1
|
Ege D, Lu HH, Boccaccini AR. Bioactive Glass and Silica Particles for Skeletal and Cardiac Muscle Tissue Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:448-461. [PMID: 38126329 DOI: 10.1089/ten.teb.2023.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
When skeletal and cardiac tissues are damaged, surgical approaches are not always successful and tissue regeneration approaches are investigated. Reports in the literature indicate that silica nanoparticles and bioactive glasses (BGs), including silicate bioactive glasses (e.g., 45S5 BG), phosphate glass fibers, boron-doped mesoporous BGs, borosilicate glasses, and aluminoborates, are promising for repairing skeletal muscle tissue. Silica nanoparticles and BGs have been combined with polymers to obtain aligned nanofibers and to maintain controlled delivery of nanoparticles for skeletal muscle repair. The literature indicates that cardiac muscle regeneration can be also triggered by the ionic products of BGs. This was observed to be due to the release of vascular endothelial growth factor and other growth factors from cardiomyocytes, which regulate endothelial cells to form capillary structures (angiogenesis). Specific studies, including both in vitro and in vivo approaches, are reviewed in this article. The analysis of the literature indicates that although the research field is still very limited, BGs are showing great promise for muscle tissue engineering and further research in the field should be carried out to expand our basic knowledge on the application of BGs in muscle (skeletal and cardiac) tissue regeneration. Impact statement This review highlights the potential of silica particles and bioactive glasses (BGs) for skeletal and cardiac tissue regeneration. These biomaterials create scaffolds triggering muscle cell differentiation. Ionic products from BGs stimulate growth factors, supporting angiogenesis in cardiac tissue repair. Further research is required to expand our know-how on silica particles and BGs in muscle tissue engineering.
Collapse
Affiliation(s)
- Duygu Ege
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hsuan-Heng Lu
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
2
|
Pomella S, Melaiu O, Dri M, Martelli M, Gargari M, Barillari G. Effects of Angiogenic Factors on the Epithelial-to-Mesenchymal Transition and Their Impact on the Onset and Progression of Oral Squamous Cell Carcinoma: An Overview. Cells 2024; 13:1294. [PMID: 39120324 PMCID: PMC11311310 DOI: 10.3390/cells13151294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development of new blood and lymphatic vessels that nourish the growing OPMDs or OSCCs and provide the latter with metastatic routes. Notably, VEGF, FGF-2, and ANG-2 are also linked to the epithelial-to-mesenchymal transition (EMT), a trans-differentiation process that respectively promotes or exasperates the invasiveness of normal and neoplastic oral epithelial cells. Here, we have summarized published work regarding the impact that the interplay among VEGF, FGF-2, ANG-2, vessel generation, and EMT has on oral carcinogenesis. Results from the reviewed studies indicate that VEGF, FGF-2, and ANG-2 spark either protein kinase B (AKT) or mitogen-activated protein kinases (MAPK), two signaling pathways that can promote both EMT and new vessels' formation in OPMDs and OSCCs. Since EMT and vessel generation are key to the onset and progression of OSCC, as well as to its radio- and chemo-resistance, these data encourage including AKT or MAPK inhibitors and/or antiangiogenic drugs in the treatment of this malignancy.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Maria Dri
- Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Mirko Martelli
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Marco Gargari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier, 00133 Rome, Italy; (S.P.); (O.M.); (M.M.); (M.G.)
| |
Collapse
|
3
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
4
|
Castillo MG, Peralta TM, Locatelli P, Velazquez C, Herrero Y, Crottogini AJ, Olea FD, Cuniberti LA. Promoting early neovascularization by allotransplanted adipose-derived Muse cells in an ovine model of acute myocardial infarction. PLoS One 2023; 18:e0277442. [PMID: 36662847 PMCID: PMC9858827 DOI: 10.1371/journal.pone.0277442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/26/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recent preclinical studies have demonstrated that bone marrow (BM)-derived Muse cells have a homing mechanism to reach damaged cardiac tissue while also being able to reduce myocardial infarct size and improve cardiac function; however, the potential of BM-Muse cells to foster new blood-vessel formation has not been fully assessed. Up to date, adipose tissue (AT)-derived Muse cells remain to be studied in acute myocardial infarction (AMI). The aim of the present study was to analyze in vitro and in vivo the neovascularization capacity of AT-Muse cells while exploring their biodistribution and differentiation potential in a translational ovine model of AMI. METHODS AND RESULTS AT-Muse cells were successfully isolated from ovine adipose tissue. In adult sheep, one or more diagonal branches of the left anterior descending coronary artery were permanently ligated for thirty minutes. Sheep were randomized in two groups and treated with intramyocardial injections: Vehicle (PBS, n = 4) and AT-Muse (2x107 AT-Muse cells labeled with PKH26 Red Fluorescent Dye, n = 4). Molecular characterization showed higher expression of angiogenic genes (VEGF, PGF and ANG) and increased number of tube formation in AT-Muse cells group compared to Adipose-derived mesenchymal stromal cells (ASCs) group. At 7 days post-IAM, the AT-Muse group showed significantly more arterioles and capillaries than the Vehicle group. Co-localization of PKH26+ cells with desmin, sarcomeric actin and troponin T implied the differentiation of Muse cells to a cardiac fate; moreover, PKH26+ cells also co-localized with a lectin marker, suggesting a possible differentiation to a vascular lineage. CONCLUSION Intramyocardially administered AT-Muse cells displayed a significant neovascularization activity and survival capacity in an ovine model of AMI.
Collapse
Affiliation(s)
- Martha G. Castillo
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Tomás M. Peralta
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Paola Locatelli
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Candela Velazquez
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Yamila Herrero
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto J. Crottogini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda D. Olea
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Luis A. Cuniberti
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
5
|
Hamel KM, Liimatta KQ, Belgodere JA, Bunnell BA, Gimble JM, Martin EC. Adipose-Derived Stromal/Stem Cell Response to Tumors and Wounds: Evaluation of Patient Age. Stem Cells Dev 2022; 31:579-592. [PMID: 35262397 PMCID: PMC9836707 DOI: 10.1089/scd.2021.0280] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/05/2022] [Indexed: 01/22/2023] Open
Abstract
Tumors were characterized as nonhealing wounds by Virchow in 1858 and Dvorak in 1986. Since then, researchers have analyzed tumors from a new perspective. The parallels between tumorigenesis and physiological wound healing can provide a new framework for developing antitumor therapeutics. One commonality between tumors and wounds is the involvement of the stromal environment, particularly adipose stromal/stem cells (ASCs). ASCs exhibit dual functions, in which they stimulate tumor progression and assist in tissue repair and regeneration. Numerous studies have focused on the role of ASCs in cancer and wound healing, but none to date has linked age, cancer, and wound healing. Furthermore, very few studies have focused on the role of donor-specific characteristics of ASCs, such as age and their role in facilitating ASC behavior in cancer and wound healing. This review article is designed to provide important insights into the impact of donor age on ASC tumor and wound response and their role in facilitating ASC behavior in cancer and wound healing.
Collapse
Affiliation(s)
- Katie M. Hamel
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Kara Q. Liimatta
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Jorge A. Belgodere
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bruce A. Bunnell
- University of North Texas Health Sciences Center, Fort Worth, Texas, USA
| | | | - Elizabeth C. Martin
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
6
|
DİLMAÇ S, ERTOSUN MG, AÇIKGÖZ E, TANRIÖVER G. Kök hücreler kanser hücrelerinin mikroçevresindeki sitokin yanıtlarını etkiler: Meme kanseri ve dental pulpa kök hücreleri arasındaki etkileşim. EGE TIP DERGISI 2021. [DOI: 10.19161/etd.1036959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
7
|
Liang Z, Liu H, Zhang Y, Xiong L, Zeng Z, He X, Wang F, Wu X, Lan P. Cyr61 from adipose-derived stem cells promotes colorectal cancer metastasis and vasculogenic mimicry formation via integrin α V β 5. Mol Oncol 2021; 15:3447-3467. [PMID: 33999512 PMCID: PMC8637569 DOI: 10.1002/1878-0261.12998] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/14/2021] [Accepted: 05/14/2021] [Indexed: 01/11/2023] Open
Abstract
Adipose‐derived stem cells (ADSCs) play a vital role in colorectal cancer (CRC) progression, but the mechanism remains largely unknown. Herein, we found that ADSCs isolated from CRC patients produced more cysteine‐rich 61 (Cyr61) than those from healthy donors, and the elevated serum Cyr61 levels were associated with advanced TNM stages. Moreover, serum Cyr61 displayed a better diagnostic value for CRC compared to carcinoembryonic antigen (CEA) and carbohydrate antigen (CA19‐9). Mechanistically, integrin αVβ5 was identified as the functional receptor by which Cyr61 promotes CRC cell metastasis in vitro and in vivo by activating the αVβ5/FAK/NF‐κB signaling pathway. In addition, Cyr61 promotes vasculogenic mimicry (VM) formation, thereby promoting tumor growth and metastasis through a αVβ5/FAK/HIF‐1α/STAT3/MMP2 signaling cascade. Histologically, xenografts and clinical samples of CRC both exhibited VM, which was correlated with HIF‐1α and MMP2 activation. Notably, we demonstrated the synergistic effect of combined anti‐VM therapy (integrin αVβ5 inhibitor) and anti‐VEGF therapy (bevacizumab) in patient‐derived xenograft models. Further investigation showed that CRC cell‐derived exosomal STAT3 promoted Cyr61 transcription in ADSCs. These findings indicate that Cyr61 derived from ADSCs plays a critical role in promoting CRC progression via integrin αVβ5 and provides a novel antitumor strategy by targeting Cyr61/αVβ5.
Collapse
Affiliation(s)
- Zhenxing Liang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Huashan Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Yunfeng Zhang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Xiong
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziwei Zeng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaowen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengwei Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xianrui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
8
|
Shi M, Zhao F, Sun L, Tang F, Gao W, Xie W, Cao X, Zhuang J, Chen X. Bioactive glass activates VEGF paracrine signaling of cardiomyocytes to promote cardiac angiogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112077. [PMID: 33947569 DOI: 10.1016/j.msec.2021.112077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022]
Abstract
The heart contains a wide range of cell types, which are not isolated but interact with one another via multifarious paracrine, autocrine and endocrine factors. In terms of cardiac angiogenesis, previous studies have proved that regulating the communication between cardiomyocytes and endothelial cells is efficacious to promote capillary formation. Firstly, this study investigated the effect and underlying mechanism of bioactive glass (BG) acted on vascular endothelial growth factor (VEGF) paracrine signaling in cardiomyocytes. We found that bioactive ions released from BG significantly promoted the VEGF production and secretion of cardiomyocytes. Subsequently, we proved that cardiomyocyte-derived VEGF played an important role in mediating the behavior of endothelial cells. Further research showed that the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/hypoxia-inducible factor 1α (HIF-1α) signaling pathway was upregulated by BG, which was involved in VEGF expression of cardiomyocytes. This study revealed that by means of modulating cellular crosstalk via paracrine signaling of host cells in heart is a new direction for the application of BGs in cardiac angiogenesis.
Collapse
Affiliation(s)
- Miao Shi
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Fujian Zhao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Luyao Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Fengling Tang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Wendong Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Weihan Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Xiaodong Cao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Jian Zhuang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Guangdong General Hospital, Guangzhou 510080, PR China.
| | - Xiaofeng Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
9
|
Luo B, Wu Y, Liu SL, Li XY, Zhu HR, Zhang L, Zheng F, Liu XY, Guo LY, Wang L, Song HX, Lv YX, Cheng ZS, Chen SY, Wang JN, Tang JM. Vagus nerve stimulation optimized cardiomyocyte phenotype, sarcomere organization and energy metabolism in infarcted heart through FoxO3A-VEGF signaling. Cell Death Dis 2020; 11:971. [PMID: 33184264 PMCID: PMC7665220 DOI: 10.1038/s41419-020-03142-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/30/2022]
Abstract
Vagus nerve stimulation (VNS) restores autonomic balance, suppresses inflammation action and minimizes cardiomyocyte injury. However, little knowledge is known about the VNS’ role in cardiomyocyte phenotype, sarcomere organization, and energy metabolism of infarcted hearts. VNS in vivo and acetylcholine (ACh) in vitro optimized the levels of α/β-MHC and α-Actinin positive sarcomere organization in cardiomyocytes while reducing F-actin assembly of cardiomyocytes. Consistently, ACh improved glucose uptake while decreasing lipid deposition in myocytes, correlating both with the increase of Glut4 and CPT1α and the decrease of PDK4 in infarcted hearts in vivo and myocytes in vitro, attributing to improvement in both glycolysis by VEGF-A and lipid uptake by VEGF-B in response to Ach. This led to increased ATP levels accompanied by the repaired mitochondrial function and the decreased oxygen consumption. Functionally, VNS improved the left ventricular performance. In contrast, ACh-m/nAChR inhibitor or knockdown of VEGF-A/B by shRNA powerfully abrogated these effects mediated by VNS. On mechanism, ACh decreased the levels of nuclear translocation of FoxO3A in myocytes due to phosphorylation of FoxO3A by activating AKT. FoxO3A overexpression or knockdown could reverse the specific effects of ACh on the expression of VEGF-A/B, α/β-MHC, Glut4, and CPT1α, sarcomere organization, glucose uptake and ATP production. Taken together, VNS optimized cardiomyocytes sarcomere organization and energy metabolism to improve heart function of the infarcted heart during the process of delaying and/or blocking the switch from compensated hypertrophy to decompensated heart failure, which were associated with activation of both P13K/AKT-FoxO3A-VEGF-A/B signaling cascade.
Collapse
Affiliation(s)
- Bin Luo
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China
| | - Yan Wu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China
| | - Shu-Lin Liu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Xing-Yuan Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Hong-Rui Zhu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China
| | - Lei Zhang
- Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Xiao-Yao Liu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China
| | - Ling-Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Lu Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Hong-Xian Song
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Yan-Xia Lv
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China
| | - Zhong-Shan Cheng
- Applied Bioinformatics Center, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shi-You Chen
- The Department of Surgery, University of Missouri, Columbia, MO, USA
| | - Jia-Ning Wang
- Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Jun-Ming Tang
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China. .,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China. .,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China.
| |
Collapse
|
10
|
Heidari-Moghadam A, Bayati V, Orazizadeh M, Rashno M. Role of Vascular Endothelial Growth Factor and Human Umbilical Vein Endothelial Cells in Designing An In Vitro Vascular-Muscle Cellular Model Using Adipose-Derived Stem Cells. CELL JOURNAL 2020; 22:19-28. [PMID: 32779430 PMCID: PMC7481900 DOI: 10.22074/cellj.2020.7034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 08/26/2019] [Indexed: 11/07/2022]
Abstract
Objective Researchers have been interested in the creation of a favorable cellular model for use in vascular-muscle
tissue engineering. The main objective of this study is to determine the myogenic effects of vascular endothelial growth
factor (VEGF) and human umbilical vein endothelial cells (HUVECs) on adipose-derived stem cells (ADSCs) to achieve
an in vitro vascular-muscle cellular model.
Materials and Methods The present experimental research was conducted on two primary groups, namely ADSCs
monoculture and ADSCs/HUVECs co-culture that were divided into control, horse serum (HS), and HS/VEGF
differentiation subgroups. HUVECs were co-cultured by ADSC in a ratio of 1:1. The myogenic differentiation was
evaluated using the reverse transcription-polymerase chain reaction (RT-PCR) and immunofluorescence in different
experimental groups. The interaction between ADSCs and HUVECs, as well as the role of ADSCs conditional medium,
was investigated for endothelial tube formation assay.
Results Immunofluorescence staining indicated that Tropomyosin was positive in ADSCs and ADSCs and HUVECs
co-culture groups on HS and HS/VEGF culture medium. Furthermore, the MyHC2 gene expression significantly
increased in HS and HS/VEGF groups in comparison with the control group (P<0.001). More importantly, there was a
significant difference in the mRNA expression of this gene between ADSCs and ADSCs and HUVECs co-culture groups
on HS/VEGF culture medium (P<0.05). Current data revealed that the co-culture of ADSCs and HUVECs could develop
endothelial network formation in the VEGF-loaded group. Also, the ADSCs-conditioned medium improved the viability
and formation of the endothelial tube in the HS and VEGF groups, respectively.
Conclusion It was concluded that ADSCs/HUVECs co-culture and dual effects of VEGF can lead to the formation
of differentiated myoblasts in proximity to endothelial network formations. These in vitro cellular models could be
potentially used in vascular-muscle tissue engineering implanted into organ defects where muscle tissue and vascular
regeneration were required.
Collapse
Affiliation(s)
- Abbas Heidari-Moghadam
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. Electronic Address: .,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Hwang OK, Noh YW, Hong JT, Lee JW. Hypoxia Pretreatment Promotes Chondrocyte Differentiation of Human Adipose-Derived Stem Cells via Vascular Endothelial Growth Factor. Tissue Eng Regen Med 2020; 17:335-350. [PMID: 32451775 DOI: 10.1007/s13770-020-00265-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human adipose tissue-derived stem cells (ADSCs) are attractive multipotent stem cell sources with therapeutic potential in various fields requiring repair and regeneration, such as acute and chronically damaged tissues. ADSC is suitable for cell-based therapy, but its use has been hampered due to poor survival after administration. Potential therapeutic use of ADSC requires mass production of cells through in vitro expansion. Many studies have consistently observed the tendency of senescence by mesenchymal stem cell (MSC) proliferation upon expansion. Hypoxia has been reported to improve stem cell proliferation and survival. METHODS We investigated the effects of hypoxia pretreatment on ADCS proliferation, migration capacity, differentiation potential and cytokine production. We also analyzed the effects of vascular endothelial growth factor (VEGF) on osteogenic and chondrogenic differentiation of ADSCs by hypoxia pretreatment. RESULTS Hypoxia pretreatment increased the proliferation of ADSCs by increasing VEGF levels. Interestingly, hypoxia pretreatment significantly increased chondrogenic differentiation but decreased osteogenic differentiation compared to normoxia. The osteogenic differentiation of ADSC was decreased by the addition of VEGF but increased by the depletion of VEGF. We have shown that hypoxia pretreatment increases the chondrogenic differentiation of ADSCs while reducing osteogenic differentiation in a VEGF-dependent manner. CONCLUSION These results show that hypoxia pretreatment can provide useful information for studies that require selective inhibition of osteogenic differentiation, such as cartilage regeneration.
Collapse
Affiliation(s)
- Ok Kyung Hwang
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk, 28160, Republic of Korea.,College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk, 28160, Republic of Korea
| | - Young Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk, 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk, 28160, Republic of Korea.
| | - Je-Wook Lee
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
12
|
Sokolowska P, Zukowski K, Lasocka I, Szulc-Dabrowska L, Jastrzebska E. Human mesenchymal stem cell (hMSC) differentiation towards cardiac cells using a new microbioanalytical method. Analyst 2020; 145:3017-3028. [PMID: 32133460 DOI: 10.1039/c9an02366f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells (SCs) are more and more often applied in tissue engineering and cell therapies, e.g. in regenerative medicine. Standard methods of SC differentiation are time consuming and ineffective. Therefore, new bioanalytical methods (i.e. Lab-on-a-Chip systems) are develop to improve such type of studies. Although, microtechnology is a rapidly growing research area, there are so far not too many works which present SC differentiation into cardiomyocytes in the microsystems. Therefore, we present new microbioanalytical method of SC differentiation towards cardiac cells using a newly developed digitally controlled microdispenser integrated with a Heart-on-a-chip system. Seven-day culture of human mesenchymal stem cells (hMSCs) and their differentiation using biochemical factors such as 5-AZA (2 μM, 24 h) and VEGF (20 ng ml-1, 72 h) were investigated in the microsystem which was automatically operated using smartphone software. hMSC differentiation into the cardiac cells was confirmed using immunostaining of cardiac markers (α-actinin and troponin T). The usage of the microsystem allowed shortening the time of hMSC differentiation in comparison to macroscale method. We showed that the microsystem, in which the in vivo microenvironment is mimicked and dynamic conditions are provided by a microdispenser, favorably affect hMSC differentiation towards cardiac cells. Based on the presented research we can conclude that the developed digitally controlled microsystem could be successfully utilized as a new microbioanalytical method for stem cells differentiation and analysis of their function under dynamic conditions. In the future, this could be a helpful tool for scientists working on regenerative medicine.
Collapse
Affiliation(s)
- Patrycja Sokolowska
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Poland.
| | | | | | | | | |
Collapse
|
13
|
Visfatin Mediates Malignant Behaviors through Adipose-Derived Stem Cells Intermediary in Breast Cancer. Cancers (Basel) 2019; 12:cancers12010029. [PMID: 31861872 PMCID: PMC7016886 DOI: 10.3390/cancers12010029] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/31/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) have been implicated in tumor growth and metastasis in breast cancer. ADSCs exhibit tumor tropism, and are of increasing clinical relevance due to the autologous fat grafting for breast reconstruction. Although we have previously shown that a high level of the adipocytokine visfatin in human breast cancer tissues correlated with tumor progression mediated by cAbl and STAT3, the effects of visfatin in the tumor microenvironment are unclear. To understand how visfatin modulates breast cancer within the tumor-stromal environment, we examined determinants of breast cancer progression using a visfatin-primed ADSCs-tumor co-culture model. ADSCs were isolated from tumor-free adipose tissue adjacent to breast tumors. ADSCs were treated with or without visfatin for 48 h and then collected for co-culture with breast cancer cell line MDA-MB-231 for 72 h in a transwell system. We found that the MDA-MB-231 cells co-cultured with visfatin-treated ADSCs (vADSCs) had higher levels of cell viability, anchorage independent growth, migration, invasion, and tumorsphere formation than that co-cultured with untreated ADSCs (uADSCs). Growth differentiation factor 15 (GDF15) upregulation was found in the co-culture conditioned medium, with GDF15 neutralizing antibody blocking the promoting effect on MDA-MB-231 in co-culture. In addition, a GDF15-induced AKT pathway was found in MDA-MB-231 and treatment with PI3K/AKT inhibitor also reversed the promoting effect. In an orthotopic xenograft mouse model, MDA-MB-231 co-injected with vADSCs formed a larger tumor mass than with uADSCs. Positive correlations were noted between visfatin, GDF15, and phosphor-AKT expressions in human breast cancer specimens. In conclusion, visfatin activated GDF15-AKT pathway mediated via ADSCs to facilitate breast cancer progression.
Collapse
|
14
|
Namgoong S, Lee H, Lee JS, Jeong SH, Han SK, Dhong ES. Comparative Biological Effects of Human Amnion and Chorion Membrane Extracts on Human Adipose-Derived Stromal Cells. J Craniofac Surg 2019; 30:947-954. [PMID: 30817541 DOI: 10.1097/scs.0000000000005393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although therapies with human amnion/chorion are used to ameliorate acute and chronic wounds, it is unclear which component of the amnion/chorion tissue promotes wound healing. To characterize the comparative effects of amnion and chorion in wound healing, we used human adipose-derived stromal cells to assess cell viability, migration, and gel contraction after treatment with amnion membrane extract (AME) or chorion membrane extract (CME). We then correlated the possible effectors via AME and CME protein profiling, and compared them by enzyme-linked immunosorbent assay (ELISA), western blotting, and immunocytochemistry. Cell viability was significantly increased with 50 and 100 μg/mL AME treatment, but with CME treatment, a significant increase was only observed with 100 μg/mL. With CME treatment, cell migration was 2.22-fold greater than the control, and collagen gels showed 20% greater contraction. Compared to control, the expression levels of α-smooth muscle actin (SMA) and smooth muscle protein 22-alpha (SM22α) increased both with AME and CME treatments, whereas calponin expression decreased. Protein profiling revealed significantly higher tissue inhibitor of metalloproteinase-1 (TIMP-1), interleukin-8, exotoxin, and adiponectin levels in CME than in AME, and ELISA revealed 8-fold higher adiponectin levels in cells treated with CME than those treated with AME. Immunocytochemistry revealed that α-SMA, SM22α, and calponin were significantly higher in CME- than AME-treated cells; however, adiponectin treatment did not enhance α-SMA, SM22α, or calponin expression. In conclusion, amnion and chorion membrane extracts exerted differential effects on proliferation and contraction of human adipose-derived stromal cells. Amnion extract was superior at inducing cell proliferation and migration, whereas CME was superior at inducing cell contraction.
Collapse
Affiliation(s)
- Sik Namgoong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
15
|
Zheng M, Kim DY, Sung JH. Ion channels and transporters in adipose-derived stem cells. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-018-00413-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
16
|
Collagen I Promotes Adipocytogenesis in Adipose-Derived Stem Cells In Vitro. Cells 2019; 8:cells8040302. [PMID: 30939867 PMCID: PMC6523348 DOI: 10.3390/cells8040302] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 01/19/2023] Open
Abstract
A hallmark of ageing is the redistribution of body fat. Particularly, subcutaneous fat decreases paralleled by a decrease of skin collagen I are typical for age-related skin atrophy. In this paper, we hypothesize that collagen I may be a relevant molecule stimulating the differentiation of adipose-derived stem cells (ASCs) into adipocytes augmenting subcutaneous fat. In this context lipogenesis, adiponectin, and collagen I receptor expression were determined. Freshly isolated ASCs were characterized by stemness-associated surface markers by FACS analysis and then transdifferentiated into adipocytes by specific medium supplements. Lipogenesis was evaluated using Nile Red staining and documented by fluorescence microscopy or quantitatively measured by using a multiwell spectrofluorometer. Expression of adiponectin was measured by real-time RT-PCR and in cell-free supernatants by ELISA, and expression of collagen I receptors was observed by western blot analysis. It was found that supports coated with collagen I promote cell adhesion and lipogenesis of ASCs. Interestingly, a reverse correlation to adiponectin expression was observed. Moreover, we found upregulation of the collagen receptor, discoidin domain-containing receptor 2; receptors of the integrin family were absent or downregulated. These findings indicate that collagen I is able to modulate lipogenesis and adiponectin expression and therefore may contribute to metabolic dysfunctions associated with ageing.
Collapse
|
17
|
Vascular Endothelial Growth Factor Enhances Proliferation of Human Tenocytes and Promotes Tenogenic Gene Expression. Plast Reconstr Surg 2019; 142:1240-1247. [PMID: 30113440 DOI: 10.1097/prs.0000000000004920] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND In obtaining human tenocytes for tendon tissue engineering, a low proliferation rate and phenotype loss during passaging is a problem. It was the authors' aim to evaluate the influence of vascular endothelial growth factor (VEGF) on human tenocyte growth and gene expression. METHODS Human tenocytes were exposed to human VEGF in various concentrations (5, 10, and 20 ng/ml) for 5 days. Cell proliferation was counted and expression of tendon-related genes was analyzed. RESULTS Tenocyte count was 1.4 × 10(5)/ml, 2.7 × 10(5)/ml, 2.3 × 10(5)/ml, and 3.7 × 10(5)/ml for 0, 5, 10, and 20 ng/ml VEGF, respectively. Expression of Col1 was up-regulated 6.4 ± 4.2-fold, 60.1 ± 21.6-fold, and 15.8 ± 10.2-fold for 5, 10, and 20 ng/ml VEGF; all differences were significant with p < 0.05. Col3 was down-regulated to 0.2 ± 0.1-fold, 0.3 ± 0.1-fold, and 0.1 ± 0.03-fold for 5, 10, and 20 ng/ml VEGF; all differences were significant. Eln was up-regulated 2.3 ± 1.7-fold, 25.5 ± 10.9-fold, and 16.6 ± 9.0-fold for 5, 10, and 20 ng/ml VEGF; differences were significant for 10 and 20 ng/ml VEGF. TSC was down-regulated to 0.3 ± 0.1-fold and 0.3 ± 0.1-fold for 5 and 20 ng/ml VEGF; differences were significant for 5 and 20 ng/ml. SCX was up-regulated to 31.3 ± 8.5-fold, 49.1 ± 23.4-fold, and 20.9 ± 9.5-fold for 5, 10, and 20 ng/ml VEGF; all changes were significant. CONCLUSIONS VEGF enhances proliferation and expression of tendon-related genes in human tenocytes. It could therefore be a useful addition for tenocyte cultivation.
Collapse
|
18
|
Neshati V, Mollazadeh S, Fazly Bazzaz BS, Iranshahi M, Mojarrad M, Naderi-Meshkin H, Kerachian MA. Cardiogenic effects of characterized Geum urbanum extracts on adipose-derived human mesenchymal stem cells. Biochem Cell Biol 2018; 96:610-618. [DOI: 10.1139/bcb-2017-0313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Stem cell therapy is considered as a promising treatment for cardiovascular diseases. Adipose-derived mesenchymal stem cells (ADMSCs) have the ability to undergo cardiomyogenesis. Medicinal plants are effective and safe candidates for cell differentiation. Therefore, the aim of our study was to investigate cardiogenic effects of characterized (HPLC–UV) extracts of Geum urbanum on ADMSCs of adipose tissue. The methanolic extracts of the root and aerial parts of G. urbanum were obtained and MTT assay was used for studying their cytotoxic effects. Then, cells were treated with 50 or 100 μg/mL of the extracts from root and aerial parts of G. urbanum. MTT assay showed that the extracts of G. urbanum did not have any toxic effects on ADMSCs. Immunostaining results showed increase in the expression of α-actinin and cardiac troponin I (cTnI), and quantitative real-time reverse-transcription PCR data confirmed the upregulation of ACTN, ACTC1, and TNNI3 genes in ADMSCs after treatment. According to HPLC fingerprinting, some cardiogenic effects of G. urbanum extracts are probably due to ellagic and gallic acid derivatives. Our findings indicated that G. urbanum extracts effectively upregulated some essential cardiogenic markers, which confirmed the therapeutic role of this plant as a traditional cardiac medicine.
Collapse
Affiliation(s)
- Vajiheh Neshati
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture, Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Petry L, Kippenberger S, Meissner M, Kleemann J, Kaufmann R, Rieger UM, Wellenbrock S, Reichenbach G, Zöller N, Valesky E. Directing adipose-derived stem cells into keratinocyte-like cells: impact of medium composition and culture condition. J Eur Acad Dermatol Venereol 2018; 32:2010-2019. [PMID: 29705993 DOI: 10.1111/jdv.15010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/22/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Adipose-derived stem cells (ASC) are known to transdifferentiate into a wide range of different cell species in vitro including along the epidermal lineage. This property makes them a promising tool for regenerative medicine to restore the epidermal barrier. OBJECTIVE This study is dedicated to identify in vitro conditions enabling transdifferentiation to a keratinocyte-like phenotype. In particular, the impact of different culture conditions (media compositions, 2D, 3D cultures) and extracellular matrix (ECM) molecules was evaluated. METHODS Adipose-derived stem cells derived from subcutaneous abdominal fat were characterized by stemness-associated markers and subjected to different media. Epithelial differentiation in 2D cultures was monitored by pan-cytokeratin expression using flow cytometry and immunocytochemistry. To evaluate the impact of different ECM molecules on epidermal stratification, 3D cultures were produced, lifted to the air-liquid interface (ALI) and examined by histological analysis and quantitative real-time RT-PCR. RESULTS We identified a medium composition containing retinoic acid, hydrocortisone, ascorbic acid and BMP-4 enabling maximum pan-cytokeratin expression in 2D cultures. Moreover, adhesion to type IV collagen further promotes the pan-cytokeratin expression. When cultures were lifted to the ALI, significant stratification was observed, particularly in supports coated with type IV collagen or fibronectin. Moreover, epidermal differentiation markers (involucrin, cytokeratin 1 and 14) become induced. CONCLUSION Conditions with hampered wound healing such as non-healing ulcers demand new treatment regimes. The here introduced optimized protocols for transdifferentiation of ASC into keratinocyte-like cells may help to establish more effective treatment procedures.
Collapse
Affiliation(s)
- L Petry
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - S Kippenberger
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - M Meissner
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - J Kleemann
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - R Kaufmann
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - U M Rieger
- Department of Plastic & Aesthetic, Reconstructive & Hand Surgery, AGAPLESION Markus Hospital, Frankfurt/Main, Germany
| | - S Wellenbrock
- Department of Plastic & Aesthetic, Reconstructive & Hand Surgery, AGAPLESION Markus Hospital, Frankfurt/Main, Germany
| | - G Reichenbach
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - N Zöller
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - E Valesky
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
20
|
Abstract
Adipose-derived stem/stromal cells (ASCs), together with adipocytes, vascular endothelial cells, and vascular smooth muscle cells, are contained in fat tissue. ASCs, like the human bone marrow stromal/stem cells (BMSCs), can differentiate into several lineages (adipose cells, fibroblast, chondrocytes, osteoblasts, neuronal cells, endothelial cells, myocytes, and cardiomyocytes). They have also been shown to be immunoprivileged, and genetically stable in long-term cultures. Nevertheless, unlike the BMSCs, ASCs can be easily harvested in large amounts with minimal invasive procedures. The combination of these properties suggests that these cells may be a useful tool in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Simone Ciuffi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Roberto Zonefrati
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| |
Collapse
|
21
|
Shen T, Shen J, Zheng QQ, Li QS, Zhao HL, Cui L, Hong CY. Cell viability and extracellular matrix synthesis in a co-culture system of corneal stromal cells and adipose-derived mesenchymal stem cells. Int J Ophthalmol 2017; 10:670-678. [PMID: 28546919 DOI: 10.18240/ijo.2017.05.02] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/09/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the impact of adipose-derived mesenchymal stem cells (ADSCs) on cell viability and extracellular matrix (ECM) synthesis of corneal stromal cells (CSCs). METHODS ADSCs and CSCs were obtained from the corneas of New Zealand white rabbits and indirectly co-cultured in vitro. The proliferative capacity of CSCs in the different groups was assessed by CCK-8 assays. Annexin V-fluorescein isothiocyanate (FITC)/proliferation indices (PI) assays were used to detect the apoptosis of CSCs. The expression levels of matrix metalloproteinase (MMP), such as MMP1, MMP2, MMP9, and collagens were also evaluated by Western blot. RESULTS ADSCs significantly promoted proliferation and invasion of CSCs in the indirect co-culture assays. The co-cultural group displayed much higher ability of proliferation, especially under the co-culture conditions of ADSCs for 3d, compared with that CSCs cultured alone. The PI of CSCs in the co-culture system were increased approximately 3-8-fold compared with the control group. A significant change was observed in the proportions of cells at apoptosis (early and late) between the negative control group (6.34% and 2.06%) and the ADCSs-treated group (4.69% and 1.59%). The expression levels of MMPs were down regulated in the co-culture models. Compared with the control group, the decrease intensities of MMP-1, MMP-2 and MMP-9 in CSCs/ADSCs group were observed, 3.90-fold, 1.09-fold and 3.03-fold, respectively. However, the increase intensities of collagen type (I, II, III, IV, and V) in CSCs were observed in CSCs/ADSCs group, 3.47-fold, 4.30-fold, 2.35-fold, 2.55-fold and 2.43-fold, respectively, compared to that in the control group. The expressions of aldehyde dehydrogenase and fibronectin in CSCs were upregulated in the co-culture models. CONCLUSION ADSCs play a promotive role in CSCs' growth and invasion, which may be partially associated with MMPs decrease and collagens increase, resulting in a positive participation in the plasticity and ECM synthesis of CSCs. This provided a new insight into the extensive role of ADSCs in CSCs and a potential molecular target for corneal therapy.
Collapse
Affiliation(s)
- Ting Shen
- Zhejiang Provincial People's Hospital, Hangzhou 310014, Zhejiang Province, China
| | - Jiang Shen
- Zhejiang Provincial People's Hospital, Hangzhou 310014, Zhejiang Province, China
| | - Qing-Qing Zheng
- Zhejiang Provincial People's Hospital, Hangzhou 310014, Zhejiang Province, China
| | - Qiu-Shi Li
- Zhejiang Provincial People's Hospital, Hangzhou 310014, Zhejiang Province, China
| | - Hai-Lan Zhao
- Zhejiang Provincial People's Hospital, Hangzhou 310014, Zhejiang Province, China
| | - Lei Cui
- Beijing Shijitan Hospital, Beijing 100050, China
| | - Chao-Yang Hong
- Zhejiang Provincial People's Hospital, Hangzhou 310014, Zhejiang Province, China.,Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| |
Collapse
|
22
|
Wang X, Li C, Gong H. Morphological and functional changes in bone marrow mesenchymal stem cells in rats with heart failure. Exp Ther Med 2017; 13:2888-2892. [PMID: 28587355 PMCID: PMC5450621 DOI: 10.3892/etm.2017.4341] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 02/01/2017] [Indexed: 12/12/2022] Open
Abstract
The changes in bone marrow-derived mesenchymal stem cells (BMSCs), in terms of cell morphology and protein expression in rats with heart failure, were studied. Pressure overload chronic heart failure rat model was induced with partial constriction of the abdominal aorta. BMSCs from the model and the sham operation groups were isolated and cultured (cell density, 108 cells/l), and supernatant was collected after 72 h. Enzyme-linked immunosorbent assay was used to measure HGF, IGF-1, PDGF, SCF, FGF and VEGF levels in the supernatant. Results showed that in the model group, the minimum cell diameter, the average cell area and the protein expression in single BMSCs were significantly less than those in the sham operation group. In the model group, SCF and PDGF levels were significantly lower than those in the sham operation group. VEGF concentration in the model group was significantly higher than that in the sham operation group. Compared with normal rats, the morphology of BMSCs in rats with heart failure changed considerably, the protein expression of a single cell and the ability to secrete cytokines decreased in a meaningful way.
Collapse
Affiliation(s)
- Xiuli Wang
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| | - Chunmei Li
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| | - Haibin Gong
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
23
|
Chen L, Deng ZJ, Zhou JS, Ji RJ, Zhang X, Zhang CS, Li YQ, Yang XQ. Tbx18-dependent differentiation of brown adipose tissue-derived stem cells toward cardiac pacemaker cells. Mol Cell Biochem 2017; 433:61-77. [PMID: 28382491 DOI: 10.1007/s11010-017-3016-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 03/15/2017] [Indexed: 12/24/2022]
Abstract
A cell-sourced biological pacemaker is a promising therapeutic approach for sick sinus syndrome (SSS) or severe atrial ventricular block (AVB). Adipose tissue-derived stem cells (ATSCs), which are optimal candidate cells for possible use in regenerative therapy for acute or chronic myocardial injury, have the potential to differentiate into spontaneous beating cardiomyocytes. However, the pacemaker characteristics of the beating cells need to be confirmed, and little is known about the underlying differential mechanism. In this study, we found that brown adipose tissue-derived stem cells (BATSCs) in mice could differentiate into spontaneous beating cells in 15% FBS Dulbecco's modified Eagle's medium (DMEM) without additional treatment. Subsequently, we provide additional evidence, including data regarding ultrastructure, protein expression, electrophysiology, and pharmacology, to support the differentiation of BATSCs into a cardiac pacemaker phenotype during the course of early cultivation. Furthermore, we found that silencing Tbx18, a key transcription factor in the development of pacemaker cells, terminated the differentiation of BATSCs into a pacemaker phenotype, suggesting that Tbx18 is required to direct BATSCs toward a cardiac pacemaker fate. The expression of Tbx3 and shox2, the other two important transcription factors in the development of pacemaker cells, was decreased by silencing Tbx18, which suggests that Tbx18 mediates the differentiation of BATSCs into a pacemaker phenotype via these two downstream transcription factors.
Collapse
Affiliation(s)
- Lei Chen
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China
| | - Zi-Jun Deng
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China
| | - Jian-Sheng Zhou
- Biochemistry and Molecular Biology Department, Second Military Medical University, Shanghai, China
| | - Rui-Juan Ji
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China
| | - Xi Zhang
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China
| | - Chuan-Sen Zhang
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China
| | - Yu-Quan Li
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China.
- Department of Anatomy, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| | - Xiang-Qun Yang
- Research Center of Regenerative Medicine, Second Military Medical University, Shanghai, China.
- Department of Anatomy, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| |
Collapse
|
24
|
MEK/ERK signaling is involved in the role of VEGF and IGF1 in cardiomyocyte differentiation of mouse adipose tissue-derived stromal cells. Int J Cardiol 2017; 228:427-434. [DOI: 10.1016/j.ijcard.2016.11.199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/06/2016] [Indexed: 12/11/2022]
|
25
|
Salehi H, Amirpour N, Niapour A, Razavi S. An Overview of Neural Differentiation Potential of Human Adipose Derived Stem Cells. Stem Cell Rev Rep 2016; 12:26-41. [PMID: 26490462 DOI: 10.1007/s12015-015-9631-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide interest in application of adult stem cells due to easy to obtain with a minimal patient discomfort, capable of producing cell numbers in large quantities and their immunocompatible properties without restriction by ethical concerns. Among these stem cells, multipotent mesenchymal stem cells (MSCs) from human adipose tissue are considered as an ideal source for various regenerative medicine. In spite of mesodermal origin of human adipose-derived stem cells (hADSCs), these cells have differentiation potential toward mesodermal and non-mesodermal lineages. Up to now, several studies have shown that hADSCs can undergo transdifferentiation and produce cells outside of their lineage, especially into neural cells when they are transferred to a specific cell environment. The purpose of this literature review is to provide an overview of the existing state of knowledge of the differentiation potential of hADSCs, specifically their ability to give rise to neuronal cells. The following review discusses different protocols considered for differentiation of hADSCs to neural cells, the neural markers that are used in each procedure and possible mechanisms that are involved in this differentiation.
Collapse
|
26
|
Quan L, Wang Y, Liang J, Qiu T, Wang H, Zhang Y, Zhang Y, Hui Q, Tao K. Screening for genes, transcription factors and miRNAs associated with the myogenic and osteogenic differentiation of human adipose tissue-derived stem cells. Int J Mol Med 2016; 38:1839-1849. [PMID: 27779643 DOI: 10.3892/ijmm.2016.2788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/09/2016] [Indexed: 11/05/2022] Open
Abstract
In the present study, we aimed to reveal the molecular mechanisms responsible for the differentiation of human adipose tissue-derived stem cells (hASCs) into myocytes and osteoblasts. Microarray data GSE37329 were obtained from the Gene Expression Omnibus database, including three hASC cell lines from healthy donors, two osteogenic lineages and two myogenic lineages from the in vitro‑induction of hASCs. Differentially expressed genes (DEGs) in the two lineages were firstly screened. Subsequently, the underlying functions of the two sets of DEGs were investigated by Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, followed by protein-protein interaction (PPI) network construction. Regulatory relationships between transcription factors (TFs) and microRNAs (miRNAs or miRs) with target genes were finally explored using different algorithms. A total of 665 and 485 DEGs were identified from the hASC‑derived myogenic and osteogenic lineages, respectively. The shared upregulated genes (n=205) in the two sets of DEGs were mainly involved in metabolism-related pathways, whereas the shared downregulated genes (n=128) were significantly enriched in the transforming growth factor-β (TGF-β) signaling pathway. Four genes, vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF2), nerve growth factor (NGF) and interleukin 1B (IL1B), presented with relatively higher degrees in both PPI networks. The transcription factor RAD21 was predicted to target shared upregulated and downregulated genes as well as specific downregulated genes in the myogenic and the osteogenic lineages. In addition, miRNA-DEG interaction analysis revealed that hsa-miR-1 regulated the most shared DEGs in the two lineages. There may be a correlation between the four genes, VEGFA, FGF2, IL1B and NGF, and the differentiation of hASCs into myocytes and osteoblasts. The TF RAD21 and hsa-miR-1 may play important roles in regulating the expression of differentiation-associated genes.
Collapse
Affiliation(s)
- Liangliang Quan
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Yang Wang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Jiulong Liang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Tao Qiu
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Hongyi Wang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Ye Zhang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Yu Zhang
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Qiang Hui
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| | - Kai Tao
- Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
27
|
Park E, Takimoto K. A long-lasting cardiomyogenic gene expression by PEI-based transfection induces endogenous cardiac mRNAs in human adipose-derived stem cells. Biochem Biophys Res Commun 2016; 479:12-6. [PMID: 27553283 DOI: 10.1016/j.bbrc.2016.08.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/18/2016] [Indexed: 10/21/2022]
Abstract
Our previous work revealed that a polyethyleneimine (PEI)-based gene delivery causes robust and sustained expression of exogenous genes in human adipose-derived stem cells (hADSCs). Here we use this method to test whether a single introduction of cDNAs for the three cardiomyogenic reprogramming genes (GATA4, MEF2C, and TBX5) might be sufficient to induce transdifferentiation of hADSCs towards the cardiomyogenic lineage. A single transfection results in sustained expression of the introduced genes for more than two weeks. hADSCs exhibit undetectable or very low levels of mRNAs for endogenous GATA4, MEF2C and TBX5. However, mRNAs for these endogenous factors become apparent at ∼2 weeks after transfection and keep increasing until the end of experimental period at the fifth week. Concordant with these cardiomyogenic genes, Nkx2.5 mRNA becomes significant at ∼2 weeks and gradually increases until the end of experimental period. Several other cardiomyogenic mRNAs were also significant at 5 weeks. Thus, a single transfection of cDNAs for the cardiomyogenic reprogramming genes using a PEI-based method induces transdifferentiation of ADSCs.
Collapse
Affiliation(s)
- Eulsoon Park
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata 940-2188, Japan
| | - Koichi Takimoto
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata 940-2188, Japan.
| |
Collapse
|
28
|
Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation 2016; 92:41-51. [PMID: 27012163 DOI: 10.1016/j.diff.2016.02.005] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/15/2016] [Accepted: 02/25/2016] [Indexed: 11/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that represent a promising source for regenerative medicine. MSCs are capable of osteogenic, chondrogenic, adipogenic and myogenic differentiation. Efficacy of differentiated MSCs to regenerate cells in the injured tissues requires the ability to maintain the differentiation toward the desired cell fate. Since MSCs represent an attractive source for autologous transplantation, cellular and molecular signaling pathways and micro-environmental changes have been studied in order to understand the role of cytokines, chemokines, and transcription factors on the differentiation of MSCs. The differentiation of MSC into a mesenchymal lineage is genetically manipulated and promoted by specific transcription factors associated with a particular cell lineage. Recent studies have explored the integration of transcription factors, including Runx2, Sox9, PPARγ, MyoD, GATA4, and GATA6 in the differentiation of MSCs. Therefore, the overexpression of a single transcription factor in MSCs may promote trans-differentiation into specific cell lineage, which can be used for treatment of some diseases. In this review, we critically discussed and evaluated the role of transcription factors and related signaling pathways that affect the differentiation of MSCs toward adipocytes, chondrocytes, osteocytes, skeletal muscle cells, cardiomyocytes, and smooth muscle cells.
Collapse
Affiliation(s)
- Sami G Almalki
- Departments of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K Agrawal
- Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA.
| |
Collapse
|
29
|
Advances in Adipose-Derived Stem Cells Isolation, Characterization, and Application in Regenerative Tissue Engineering. Stem Cells Int 2016. [PMID: 26981130 DOI: 10.1155/+2016/3206807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Obesity is a complex, multifactorial disease that has been extensively researched in recent times. Obesity is characterized by excess deposition of adipose tissue in response to surplus energy. Despite the negative connotations of adipose tissue (AT), it serves as a critical endocrine organ. Adipose tissue is a source of several adipokines and cytokines which have been deemed important for both normal metabolic function and disease formation. The discoveries of metabolically active brown AT in adult humans and adipose tissue derived stem cells (ADSC) have been key findings in the past decade with potential therapeutic implications. ADSCs represent an enticing pool of multipotent adult stem cells because of their noncontroversial nature, relative abundance, ease of isolation, and expandability. A decade and a half since the discovery of ADSCs, the scientific community is still working to uncover their therapeutic potential in a wide range of diseases. In this review, we provide an overview of the recent developments in the field of ADSCs and examine their potential use in transplantation and cell-based therapies for the regeneration of diseased organs and systems. We also hope to provide perspective on how to best utilize this readily available, powerful pool of stem cells in the future.
Collapse
|
30
|
Advances in Adipose-Derived Stem Cells Isolation, Characterization, and Application in Regenerative Tissue Engineering. Stem Cells Int 2016; 2016:3206807. [PMID: 26981130 PMCID: PMC4766348 DOI: 10.1155/2016/3206807] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/14/2015] [Accepted: 01/14/2016] [Indexed: 12/22/2022] Open
Abstract
Obesity is a complex, multifactorial disease that has been extensively researched in recent times. Obesity is characterized by excess deposition of adipose tissue in response to surplus energy. Despite the negative connotations of adipose tissue (AT), it serves as a critical endocrine organ. Adipose tissue is a source of several adipokines and cytokines which have been deemed important for both normal metabolic function and disease formation. The discoveries of metabolically active brown AT in adult humans and adipose tissue derived stem cells (ADSC) have been key findings in the past decade with potential therapeutic implications. ADSCs represent an enticing pool of multipotent adult stem cells because of their noncontroversial nature, relative abundance, ease of isolation, and expandability. A decade and a half since the discovery of ADSCs, the scientific community is still working to uncover their therapeutic potential in a wide range of diseases. In this review, we provide an overview of the recent developments in the field of ADSCs and examine their potential use in transplantation and cell-based therapies for the regeneration of diseased organs and systems. We also hope to provide perspective on how to best utilize this readily available, powerful pool of stem cells in the future.
Collapse
|
31
|
Albulescu R, Tanase C, Codrici E, Popescu DI, Cretoiu SM, Popescu LM. The secretome of myocardial telocytes modulates the activity of cardiac stem cells. J Cell Mol Med 2015; 19:1783-1794. [PMID: 26176909 PMCID: PMC4549029 DOI: 10.1111/jcmm.12624] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/06/2015] [Indexed: 02/05/2023] Open
Abstract
Telocytes (TCs) are interstitial cells that are present in numerous organs, including the heart interstitial space and cardiac stem cell niche. TCs are completely different from fibroblasts. TCs release extracellular vesicles that may interact with cardiac stem cells (CSCs) via paracrine effects. Data on the secretory profile of TCs and the bidirectional shuttle vesicular signalling mechanism between TCs and CSCs are scarce. We aimed to characterize and understand the in vitro effect of the TC secretome on CSC fate. Therefore, we studied the protein secretory profile using supernatants from mouse cultured cardiac TCs. We also performed a comparative secretome analysis using supernatants from rat cultured cardiac TCs, a pure CSC line and TCs-CSCs in co-culture using (i) high-sensitivity on-chip electrophoresis, (ii) surface-enhanced laser desorption/ionization time-of-flight mass spectrometry and (iii) multiplex analysis by Luminex-xMAP. We identified several highly expressed molecules in the mouse cardiac TC secretory profile: interleukin (IL)-6, VEGF, macrophage inflammatory protein 1α (MIP-1α), MIP-2 and MCP-1, which are also present in the proteome of rat cardiac TCs. In addition, rat cardiac TCs secrete a slightly greater number of cytokines, IL-2, IL-10, IL-13 and some chemokines like, GRO-KC. We found that VEGF, IL-6 and some chemokines (all stimulated by IL-6 signalling) are secreted by cardiac TCs and overexpressed in co-cultures with CSCs. The expression levels of MIP-2 and MIP-1α increased twofold and fourfold, respectively, when TCs were co-cultured with CSCs, while the expression of IL-2 did not significantly differ between TCs and CSCs in mono culture and significantly decreased (twofold) in the co-culture system. These data suggest that the TC secretome plays a modulatory role in stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Radu Albulescu
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
- National Institute for Chemical Pharmaceutical Research & DevelopmentBucharest, Romania
| | - Cristiana Tanase
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Elena Codrici
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Daniela I Popescu
- Biochemistry-Proteomics Department, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Sanda M Cretoiu
- Division of Cell Biology and Histology, Carol Davila University of Medicine and PharmacyBucharest, Romania
- Department of Ultrastructural Pathology, Victor Babeş National Institute of PathologyBucharest, Romania
| | - Laurentiu M Popescu
- Division of Cell Biology and Histology, Carol Davila University of Medicine and PharmacyBucharest, Romania
- Department of Advanced Studies, Victor Babeş National Institute of PathologyBucharest, Romania
| |
Collapse
|
32
|
Cutts J, Nikkhah M, Brafman DA. Biomaterial Approaches for Stem Cell-Based Myocardial Tissue Engineering. Biomark Insights 2015; 10:77-90. [PMID: 26052226 PMCID: PMC4451817 DOI: 10.4137/bmi.s20313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 12/21/2022] Open
Abstract
Adult and pluripotent stem cells represent a ready supply of cellular raw materials that can be used to generate the functionally mature cells needed to replace damaged or diseased heart tissue. However, the use of stem cells for cardiac regenerative therapies is limited by the low efficiency by which stem cells are differentiated in vitro to cardiac lineages as well as the inability to effectively deliver stem cells and their derivatives to regions of damaged myocardium. In this review, we discuss the various biomaterial-based approaches that are being implemented to direct stem cell fate both in vitro and in vivo. First, we discuss the stem cell types available for cardiac repair and the engineering of naturally and synthetically derived biomaterials to direct their in vitro differentiation to the cell types that comprise heart tissue. Next, we describe biomaterial-based approaches that are being implemented to enhance the in vivo integration and differentiation of stem cells delivered to areas of cardiac damage. Finally, we present emerging trends of using stem cell-based biomaterial approaches to deliver pro-survival factors and fully vascularized tissue to the damaged and diseased cardiac tissue.
Collapse
Affiliation(s)
- Josh Cutts
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
33
|
Di Taranto G, Cicione C, Visconti G, Isgrò MA, Barba M, Di Stasio E, Stigliano E, Bernardini C, Michetti F, Salgarello M, Lattanzi W. Qualitative and quantitative differences of adipose-derived stromal cells from superficial and deep subcutaneous lipoaspirates: a matter of fat. Cytotherapy 2015; 17:1076-89. [PMID: 26002819 DOI: 10.1016/j.jcyt.2015.04.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS Subcutaneous fat represents a valuable reservoir of adipose-derived stem cells (ASCs) in the stromal vascular fraction (SVF), widely exploited in regenerative medicine applications, being easily harvested through lipoaspiration. The lack of standardized procedures for autologous fat grafting guided research efforts aimed at identifying possible differences related to the harvesting site, which may affect cell isolation yield, cell growth properties and clinical outcomes. Subcutaneous fat features a complex architecture: the superficial fascia separates superficial adipose tissue (SAT) from deep layer tissue (DAT). We aimed to unravel the differences between SAT and DAT, considering morphological structure, SVF composition, and ASC properties. METHODS SAT and DAT were collected from female donors and comparatively analyzed to evaluate cellular yield and viability, morphology, immunophenotype and molecular profile. ASCs were isolated in primary culture and used for in vitro differentiation assays. SAT and DAT from cadaver donors were also analyzed through histology and immunohistochemistry to assess morphology and cell localization within the hypoderm. RESULTS Liposuctioned SAT contained a higher stromal tissue compound, along with a higher proportion of CD105-positive cells, compared with DAT from the same harvesting site. Also, cells isolated from SAT displayed increased multipotency and stemness features. All differences were mainly evidenced in specimens harvested from the abdominal region. According to our results, SAT features overall increased stem properties. CONCLUSIONS Given that subcutaneous adipose tissue is currently exploited as the gold standard source for high-yield isolation of adult stem cells, these results may provide precious hints toward the definition of standardized protocols for microharvesting.
Collapse
Affiliation(s)
- Giuseppe Di Taranto
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Plastic and Reconstructive Surgery, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Claudia Cicione
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Visconti
- Department of Plastic and Reconstructive Surgery, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Maria A Isgrò
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Enrico Di Stasio
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Egidio Stigliano
- Institute of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Camilla Bernardini
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy; Latium Musculoskeletal Tissue Bank, Rome, Italy
| | - Marzia Salgarello
- Department of Plastic and Reconstructive Surgery, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy; Latium Musculoskeletal Tissue Bank, Rome, Italy.
| |
Collapse
|
34
|
Freese KE, Kokai L, Edwards RP, Philips BJ, Sheikh MA, Kelley J, Comerci J, Marra KG, Rubin JP, Linkov F. Adipose-derived stems cells and their role in human cancer development, growth, progression, and metastasis: a systematic review. Cancer Res 2015; 75:1161-8. [PMID: 25736688 DOI: 10.1158/0008-5472.can-14-2744] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/15/2014] [Indexed: 11/16/2022]
Abstract
Obesity is a well recognized risk factor for several types of cancers, many of which occur solely or disproportionately in women. Adipose tissue is a rich source of adipose-derived stem cells (ASC), which have received attention for their role in cancer behavior. The purpose of this systematic review is to present the existing literature on the role of ASCs in the growth, development, progression, and metastasis of cancer, with an emphasis on malignancies that primarily affect women. To accomplish this goal, the bibliographic database PubMed was systematically searched for articles published between 2001 and 2014 that address ASCs' relationship to human cancer. Thirty-seven articles on ASCs' role in human cancer were reviewed. Literature suggests that ASCs exhibit cancer-promoting properties, influence/are influenced by the tumor microenvironment, promote angiogenesis, and may be associated with pathogenic processes through a variety of mechanisms, such as playing a role in hypoxic tumor microenvironment. ASCs appear to be important contributors to tumor behavior, but research in areas specific to women's cancers, specifically endometrial cancer, is scarce. Also, because obesity continues to be a major health concern, it is important to continue research in this area to improve understanding of the impact adiposity has on cancer incidence.
Collapse
Affiliation(s)
- Kyle E Freese
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania. Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania.
| | - Lauren Kokai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania. McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert P Edwards
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - Brian J Philips
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - M Aamir Sheikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - Joseph Kelley
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - John Comerci
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania. McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania. McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Faina Linkov
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Women's Research Institute, Pittsburgh, Pennsylvania. Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Kai D, Prabhakaran MP, Jin G, Tian L, Ramakrishna S. Potential of VEGF-encapsulated electrospun nanofibers for in vitro
cardiomyogenic differentiation of human mesenchymal stem cells. J Tissue Eng Regen Med 2015; 11:1002-1010. [DOI: 10.1002/term.1999] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/13/2014] [Accepted: 12/12/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Dan Kai
- NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore
- Institute of Materials Research and Engineering (IMRE); Singapore
| | - Molamma P. Prabhakaran
- Centre for Nanofibers and Nanotechnology, Nanoscience and Nanotechnology Initiative; National University of Singapore
| | - Guorui Jin
- Department of Mechanical Engineering; National University of Singapore
- Institute of Materials Research and Engineering (IMRE); Singapore
| | - Lingling Tian
- Centre for Nanofibers and Nanotechnology, Nanoscience and Nanotechnology Initiative; National University of Singapore
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, Nanoscience and Nanotechnology Initiative; National University of Singapore
- Department of Mechanical Engineering; National University of Singapore
| |
Collapse
|
36
|
Siciliano C, Chimenti I, Ibrahim M, Napoletano C, Mangino G, Scafetta G, Zoccai GB, Rendina EA, Calogero A, Frati G, De Falco E. Cardiosphere conditioned media influence the plasticity of human mediastinal adipose tissue-derived mesenchymal stem cells. Cell Transplant 2015; 24:2307-2322. [PMID: 26531290 DOI: 10.3727/096368914x685771] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Nowadays, cardiac regenerative medicine is facing many limitations because of the complexity to find the most suitable stem cell source and to understand the regenerative mechanisms involved. Mesenchymal stem cells (MSCs) have shown great regenerative potential due to their intrinsic properties and ability to restore cardiac functionality, directly by transdifferentiation and indirectly by paracrine effects. Yet, how MSCs could respond to definite cardiac-committing microenvironments, such as that created by resident cardiac progenitor cells in the form of cardiospheres (CSs), has never been addressed. Recently, a putative MSC pool has been described in the mediastinal fat (hmADMSCs), but both its biology and function remain hitherto unexplored. Accordingly, we investigated the potential of hmADMSCs to be committed toward a cardiovascular lineage after preconditioning with CS-conditioned media (CCM). Results indicated that CCM affects cell proliferation. Gene expression levels of multiple cardiovascular and stemness markers (MHC, KDR, Nkx2.5, Thy-1, c-kit, SMA) are significantly modulated, and the percentage of hmADMSCs preconditioned with CCM and positive for Nkx2.5, MHC, and KDR is significantly higher relative to FBS and explant-derived cell conditioned media (EDCM, the unselected stage before CS formation). Growth factor-specific and survival signaling pathways (i.e., Erk1/2, Akt, p38, mTOR, p53) present in CCM are all equally regulated. Nonetheless, earlier BAD phosphorylation (Ser112) occurs associated with the CS microenvironment (and to a lesser extent to EDCM), whereas faster phosphorylation of PRAS40 in FBS, and of Akt (Ser473) in EDCM and 5-azacytidine occurs compared to CCM. For the first time, we demonstrated that the MSC pool held in the mediastinal fat is adequately plastic to partially differentiate in vitro toward a cardiac-like lineage. Besides, we have provided novel evidence of the potent inductive niche-like microenvironment that the CS structure can reproduce in vitro. hmADMSCs can represent an interesting tool in order to exploit their possible role in cardiovascular diseases and treatment.
Collapse
Affiliation(s)
- Camilla Siciliano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Adipose-Derived Stem Cells for Therapeutic Applications. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
38
|
Human Adipose-Derived Stem Cells (ASC): Their Efficacy in Clinical Applications. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
39
|
Adipose-derived stem cells inhibit epidermal melanocytes through an interleukin-6-mediated mechanism. Plast Reconstr Surg 2014; 134:470-480. [PMID: 25158706 DOI: 10.1097/prs.0000000000000431] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Several investigators have postulated that human adipose-derived stem cells can be used for skin rejuvenation, but there have been few reports about their direct effects on human epidermal melanocytes. The authors studied the effects on melanocytes, and the causative agent of those effects was further investigated in this study. METHODS Human epidermal melanocytes were divided into three groups and cultured in adipose-derived stem cell-conditioned medium, human dermal fibroblast-conditioned medium, or control medium. Concentrations of melanogenic cytokines in these media were measured using enzyme-linked immunosorbent assay kits. After 3 and 7 days of incubation, cell proliferation, melanin content, tyrosinase activity, and melanogenic gene expression were measured. Interleukin-6-neutralizing antibodies were mixed with adipose-derived stem cell-conditioned medium in which human epidermal melanocytes were cultured, and melanocyte growth and melanogenesis were measured again. RESULTS Interleukin-6 concentrations in adipose-derived stem cell- and human epidermal melanocyte-conditioned media were 1373 and 495 pg/ml, respectively. Both types of medium suppressed melanocyte proliferation and melanin synthesis (p < 0.05), but adipose-derived stem cell-conditioned medium was more effective than human dermal fibroblast-conditioned medium in inhibition of human epidermal melanocyte proliferation, melanin synthesis, and tyrosinase activity (p < 0.05). Interleukin-6-neutralizing antibody sufficiently reversed the antimelanogenic effects of adipose-derived stem cell-conditioned medium such that human epidermal melanocyte proliferation, melanin content, tyrosinase activity, and tyrosinase mRNA levels were restored (p < 0.05). CONCLUSIONS Adipose-derived stem cell-conditioned medium inhibited melanocyte proliferation and melanin synthesis by down-regulating melanogenic enzymes. Interleukin-6 plays a pivotal role in inhibition of melanocytes.
Collapse
|
40
|
Zheng L, Han P, Liu J, Li R, Yin W, Wang T, Zhang W, Kang YJ. Role of copper in regression of cardiac hypertrophy. Pharmacol Ther 2014; 148:66-84. [PMID: 25476109 DOI: 10.1016/j.pharmthera.2014.11.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 11/17/2014] [Indexed: 02/07/2023]
Abstract
Pressure overload causes an accumulation of homocysteine in the heart, which is accompanied by copper depletion through the formation of copper-homocysteine complexes and the excretion of the complexes. Copper supplementation recovers cytochrome c oxidase (CCO) activity and promotes myocardial angiogenesis, along with the regression of cardiac hypertrophy and the recovery of cardiac contractile function. Increased copper availability is responsible for the recovery of CCO activity. Copper promoted expression of angiogenesis factors including vascular endothelial growth factor (VEGF) in endothelial cells is responsible for angiogenesis. VEGF receptor-2 (VEGFR-2) is critical for hypertrophic growth of cardiomyocytes and VEGFR-1 is essential for the regression of cardiomyocyte hypertrophy. Copper, through promoting VEGF production and suppressing VEGFR-2, switches the VEGF signaling pathway from VEGFR-2-dependent to VEGFR-1-dependent, leading to the regression of cardiomyocyte hypertrophy. Copper is also required for hypoxia-inducible factor-1 (HIF-1) transcriptional activity, acting on the interaction between HIF-1 and the hypoxia responsible element and the formation of HIF-1 transcriptional complex by inhibiting the factor inhibiting HIF-1. Therefore, therapeutic targets for copper supplementation-induced regression of cardiac hypertrophy include: (1) the recovery of copper availability for CCO and other critical cellular events; (2) the activation of HIF-1 transcriptional complex leading to the promotion of angiogenesis in the endothelial cells by VEGF and other factors; (3) the activation of VEGFR-1-dependent regression signaling pathway in the cardiomyocytes; and (4) the inhibition of VEGFR-2 through post-translational regulation in the hypertrophic cardiomyocytes. Future studies should focus on target-specific delivery of copper for the development of clinical application.
Collapse
Affiliation(s)
- Lily Zheng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Pengfei Han
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jiaming Liu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Rui Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wen Yin
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tao Wang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wenjing Zhang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
41
|
Karam JP, Bonafè F, Sindji L, Muscari C, Montero-Menei CN. Adipose-derived stem cell adhesion on laminin-coated microcarriers improves commitment toward the cardiomyogenic lineage. J Biomed Mater Res A 2014; 103:1828-39. [PMID: 25098676 DOI: 10.1002/jbm.a.35304] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/14/2014] [Accepted: 07/31/2014] [Indexed: 12/27/2022]
Abstract
For tissue-engineering studies of the infarcted heart it is essential to identify a source of cells that may provide cardiomyocyte progenitors, which is easy to amplify, accessible in adults, and allowing autologous grafts. Preclinical studies have shown that human adipose-derived stem cells (ADSCs) can differentiate into cardiomyocyte-like cells and improve heart function in myocardial infarction. We have developed pharmacologically active microcarriers (PAMs) which are biodegradable and biocompatible polymeric microspheres conveying cells on their biomimetic surface, therefore providing an adequate three-dimensional (3D) microenvironment. Moreover, they can release a growth factor in a prolonged manner. In order to implement ADSCs and PAMs for cardiac tissue engineering we first defined the biomimetic surface by studying the influence of matrix molecules laminin (LM) and fibronectin (FN), in combination with growth factors present in the cardiogenic niche, to further enhance the in vitro cardiac differentiation of ADSCs. We demonstrated that LM increased the expression of cardiac markers (Nkx2.5, GATA4, MEF2C) by ADSCs after 2 weeks in vitro. Interestingly, our results suggest that the 3D support provided by PAMs with a LM biomimetic surface (LM-PAMs) further enhanced the expression of cardiac markers and induced the expression of a more mature contractile protein, cardiac troponin I, compared with the 2D differentiating conditions after only 1 week in culture. The enrichment of the growth-factor cocktail with TGF-β1 potentiated the cardiomyogenic differentiation. These results suggest that PAMs offering a LM biomimetic surface may be efficiently used for applications combining adult stem cells in tissue-engineering strategies of the ischemic heart.
Collapse
Affiliation(s)
- Jean-Pierre Karam
- LUNAM Université, UMR S-1066 F-49933, Angers, France; NSERM U1066, MINT "Micro et nanomédecines biomimétiques,", F-49933, Angers, France; INRC-National Institute for Cardiovascular Research, 40126, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | | | | | | | | |
Collapse
|
42
|
Chen L, Qin F, Ge M, Shu Q, Xu J. Application of adipose-derived stem cells in heart disease. J Cardiovasc Transl Res 2014; 7:651-63. [PMID: 25205213 DOI: 10.1007/s12265-014-9585-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 08/05/2014] [Indexed: 12/14/2022]
Abstract
Therapy with mesenchymal stem cells is one of the promising tools to improve outcomes after myocardial infarction. Adipose-derived stem cells (ASCs) are an ideal source of mesenchymal stem cells due to their abundance and ease of preparation. Studies in animal models of myocardial infarction have demonstrated the ability of injected ASCs to engraft and differentiate into cardiomyocytes and vasculature cells. ASCs secrete a wide array of angiogenic and anti-apoptotic paracrine factors such as vascular endothelial growth factor, hepatocyte growth factor, and insulin-like growth factor 1. ASCs are capable of enhancing heart function, reducing myocardial infarction, promoting vascularization, and reversing remodeling in the ischemically injured hearts. Furthermore, several ongoing clinical trials using ASCs are producing promising results for heart diseases. This article reviews the isolation, differentiation, immunoregulatory properties, mechanisms of action, animal models, and ongoing clinical trials of ASCs for cardiac disease.
Collapse
Affiliation(s)
- Lina Chen
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | | | | | | | | |
Collapse
|
43
|
Girão-Silva T, Bassaneze V, Campos LCG, Barauna VG, Dallan LAO, Krieger JE, Miyakawa AA. Short-term mechanical stretch fails to differentiate human adipose-derived stem cells into cardiovascular cell phenotypes. Biomed Eng Online 2014; 13:54. [PMID: 24885410 PMCID: PMC4012171 DOI: 10.1186/1475-925x-13-54] [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: 01/09/2014] [Accepted: 04/22/2014] [Indexed: 12/31/2022] Open
Abstract
Background We and others have previously demonstrated that adipose-derived stem cells (ASCs) transplantation improve cardiac dysfunction post-myocardium infarction (MI) under hemodynamic stress in rats. The beneficial effects appear to be associated with pleiotropic factors due to a complex interplay between the transplanted ASCs and the microenvironment in the absence of cell transdifferentiation. In the present work, we tested the hypothesis that mechanical stretch per se could change human ASCs (hASCs) into cardiovascular cell phenotypes that might influence post-MI outcomes. Methods Human ASCs were obtained from patients undergoing liposuction procedures. These cells were stretched 12%, 1Hz up to 96 hours by using Flexercell 4000 system. Protein and gene expression were evaluated to identify cardiovascular cell markers. Culture medium was analyzed to determine cell releasing factors, and contraction potential was also evaluated. Results Mechanical stretch, which is associated with extracellular signal-regulated kinase (ERK) phosphorylation, failed to induce the expression of cardiovascular cell markers in human ASCs, and mesenchymal cell surface markers (CD29; CD90) remained unchanged. hASCs and smooth muscle cells (SMCs) displayed comparable contraction ability. In addition, these cells demonstrated a profound ability to secrete an array of cytokines. These two properties of human ASCs were not influenced by mechanical stretch. Conclusions Altogether, our findings demonstrate that hASCs secrete an array of cytokines and display contraction ability even in the absence of induction of cardiovascular cell markers or the loss of mesenchymal surface markers when exposed to mechanical stretch. These properties may contribute to beneficial post-MI cardiovascular outcomes and deserve to be further explored under the controlled influence of other microenvironment components associated with myocardial infarction, such as tissue hypoxia.
Collapse
Affiliation(s)
| | | | | | | | | | - Jose Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) - University of São Paulo School of Medicine, Avenue Dr, Eneas de Carvalho Aguiar, 44, São Paulo, SP 05403-000, Brazil.
| | | |
Collapse
|
44
|
Dong Y, Hassan WU, Kennedy R, Greiser U, Pandit A, Garcia Y, Wang W. Performance of an in situ formed bioactive hydrogel dressing from a PEG-based hyperbranched multifunctional copolymer. Acta Biomater 2014; 10:2076-85. [PMID: 24389319 DOI: 10.1016/j.actbio.2013.12.045] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 12/11/2013] [Accepted: 12/17/2013] [Indexed: 01/08/2023]
Abstract
Hydrogel dressings have been widely used for wound management due to their ability to maintain a hydrated wound environment, restore the skin's physical barrier and facilitate regular dressing replacement. However, the therapeutic functions of standard hydrogel dressings are restricted. In this study, an injectable hybrid hydrogel dressing system was prepared from a polyethylene glycol (PEG)-based thermoresponsive hyperbranched multiacrylate functional copolymer and thiol-modified hyaluronic acid in combination with adipose-derived stem cells (ADSCs). The cell viability, proliferation and metabolic activity of the encapsulated ADSCs were studied in vitro, and a rat dorsal full-thickness wound model was used to evaluate this bioactive hydrogel dressing in vivo. It was found that long-term cell viability could be achieved for both in vitro (21days) and in vivo (14days) studies. With ADSCs, this hydrogel system prevented wound contraction and enhanced angiogenesis, showing the potential of this system as a bioactive hydrogel dressing for wound healing.
Collapse
Affiliation(s)
- Yixiao Dong
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Waqar U Hassan
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Robert Kennedy
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Udo Greiser
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland
| | - Yolanda Garcia
- Anatomy Department, National University of Ireland, Galway, Ireland.
| | - Wenxin Wang
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
45
|
Li N, Pasha Z, Ashraf M. Reversal of ischemic cardiomyopathy with Sca-1+ stem cells modified with multiple growth factors. PLoS One 2014; 9:e93645. [PMID: 24705272 PMCID: PMC3976296 DOI: 10.1371/journal.pone.0093645] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 03/09/2014] [Indexed: 01/09/2023] Open
Abstract
Background We hypothesized that bone marrow derived Sca-1+ stem cells (BM Sca-1+) transduced with multiple therapeutic cytokines with diverse effects will induce faster angiomyogenic differentiation in the infarcted myocardium. Methods and Results BM Sca-1+ were purified from transgenic male mice expressing GFP. Plasmids encoding for select quartet of growth factors, i.e., human IGF-1, VEGF, SDF-1α and HGF were prepared and used for genetic modification of Sca-1+ cells (GFSca-1+). Scramble transfected cells (ScSca-1+) were used as a control. RT-PCR and western blotting showed significantly higher expression of the growth factors in GFSca-1+. Besides the quartet of the therapeutic growth factors, PCR based growth factor array showed upregulation of multiple angiogenic and prosurvival factors such as Ang-1, Ang-2, MMP9, Cx43, BMP2, BMP5, FGF2, and NGF in GFSca-1+ (p<0.01 vsScSca-1+). LDH and TUNEL assays showed enhanced survival of GFSca-1+ under lethal anoxia (p<0.01 vs ScSca-1+). MTS assay showed significant increased cell proliferation in GFSca-1+ (p<0.05 vsScSca-1+). For in vivo study, female mice were grouped to receive the intramyocardial injection of 15 μl DMEM without cells (group-1) or containing 2.5×105ScSca-1+ (group-2) or GFSca-1+ (group-3) immediately after coronary artery ligation. As indicated by Sry gene, a higher survival of GFSca-1+ in group-3 on day4 (2.3 fold higher vs group-2) was observed with massive mobilization of stem and progenitor cells (cKit+, Mdr1+, Cxcr4+ cells). Heart tissue sections immunostained for actinin and Cx43 at 4 weeks post engraftment showed extensive myofiber formation and expression of gap junctions. Immunostaining for vWF showed increased blood vessel density in both peri-infarct and infarct regions in group-3. Infarct size was attenuated and the global heart function was improved in group-3 as compared to group-2. Conclusions Administration of BM Sca-1+ transduced with multiple genes is a novel approach to treat infarcted heart for its regeneration.
Collapse
Affiliation(s)
- Ning Li
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Zeeshan Pasha
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Muhammad Ashraf
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
46
|
Bekhite MM, Finkensieper A, Rebhan J, Huse S, Schultze-Mosgau S, Figulla HR, Sauer H, Wartenberg M. Hypoxia, Leptin, and Vascular Endothelial Growth Factor Stimulate Vascular Endothelial Cell Differentiation of Human Adipose Tissue-Derived Stem Cells. Stem Cells Dev 2014; 23:333-51. [DOI: 10.1089/scd.2013.0268] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Mohamed M. Bekhite
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Andreas Finkensieper
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Jennifer Rebhan
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Stephanie Huse
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Stefan Schultze-Mosgau
- Department of Cranio-Maxillofacial Surgery and Plastic Surgery, Jena University Hospital, Jena, Germany
| | - Hans-Reiner Figulla
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany
| | - Maria Wartenberg
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| |
Collapse
|
47
|
Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FMP, Lacerda SMSN, Lalwani P, Santos AK, Gomes KN, Ulrich H, Kihara AH, Resende RR. Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical applications. Cytometry A 2013; 85:43-77. [DOI: 10.1002/cyto.a.22402] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Bruna R. Sousa
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Ricardo C. Parreira
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Emerson A Fonseca
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Maria J. Amaya
- Department of Internal Medicine, Section of Digestive Diseases; Yale University School of Medicine; New Haven Connecticut
| | - Fernanda M. P. Tonelli
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Samyra M. S. N. Lacerda
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Pritesh Lalwani
- Faculdade de Ciências Farmacêuticas; Universidade Federal do Amazonas; Manaus AM Brazil
| | - Anderson K. Santos
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Katia N. Gomes
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Henning Ulrich
- Departamento de Bioquímica; Instituto de Química, Universidade de São Paulo; São Paulo SP Brazil
| | - Alexandre H. Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição; Universidade Federal do ABC; Santo André SP Brazil
| | - Rodrigo R. Resende
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| |
Collapse
|
48
|
Tian L, Prabhakaran MP, Ding X, Kai D, Ramakrishna S. Emulsion electrospun nanofibers as substrates for cardiomyogenic differentiation of mesenchymal stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:2577-2587. [PMID: 23851928 DOI: 10.1007/s10856-013-5003-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 07/05/2013] [Indexed: 06/02/2023]
Abstract
The potential of cardiomyogenic differentiation of human mesenchymal stem cells (hMSCs) on emulsion electrospun scaffold containing poly(L-lactic acid)-co-poly-(ε-caprolactone), gelatin and vascular endothelial growth factor (PLCL/GV) was investigated in this study. The characterizations of the scaffold were carried out using scanning electron microscope (SEM), transmission electron microscope, water contact angle and porometer. The proliferation of hMSCs showed that 73.4% higher cell proliferation on PLCL/GV scaffolds than that on PLCL scaffold after 20 days of cell culture. Results of 5-chloromethylfluorescein diacetate staining and SEM morphology analysis indicated that hMSCs differentiated on PLCL/GV scaffolds showed irregular morphology of cardiomyocyte phenotype compared to the typical long and thin hMSC phenotype. Immunostaining results showed the expression of alpha actinin and myosin heavy chain. Our studies identified emulsion electrospinning as a method for fabrication of core-shell fibers suitable for the differentiation of stem cells to cardiac cells, with potential application in cardiac regeneration.
Collapse
Affiliation(s)
- Lingling Tian
- Key Laboratory of Textile Science & Technology, Ministry of Education of China, Donghua University, Shanghai, 201620, China
| | | | | | | | | |
Collapse
|
49
|
Pikuła M, Marek-Trzonkowska N, Wardowska A, Renkielska A, Trzonkowski P. Adipose tissue-derived stem cells in clinical applications. Expert Opin Biol Ther 2013; 13:1357-70. [PMID: 23919743 DOI: 10.1517/14712598.2013.823153] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION In the past decade human adipose tissue has been identified as a source of multipotent stem cells. Adipose tissue derived stem cells (ASCs) are characterised by immunosuppressive properties and low immunogenicity. Therefore, they can be used in regenerative medicine, as well as applied to induce graft tolerance or prevent autoimmunity. ASCs can be easily harvested with low morbidity, which is their main advantage over mesenchymal stem cells (MSCs) derived from other sources. AREAS COVERED The review focuses on reported clinical applications of ASCs and discusses technical approaches of their isolation and processing. The differences in phenotype and differentiation preferences between ASCs and other MSCs that may affect the choice of a particular cell type for the future therapy are also described. EXPERT OPINION ASCs seem to be the perfect tool for regenerative medicine and immunosuppressive cellular therapies. Nevertheless, there are some tasks that should be addressed by the future studies: i) ASCs require better characterisation; a set of markers determining ASCs should be clearly defined; ii) there is need for more studies on safety of reconstructive therapies with ASCs in cancer patients (e.g., after mastectomy); iii) release criteria should be determined for freshly isolated and ex vivo expanded ASCs designed for clinical applications.
Collapse
Affiliation(s)
- Michał Pikuła
- Medical University of Gdańsk, Department of Clinical Immunology and Transplantology , ul. Dębinki 7, 80-210 Gdańsk , Poland
| | | | | | | | | |
Collapse
|
50
|
Kim JH, Shin SH, Li TZ, Suh H. Influence of in vitro biomimicked stem cell 'niche' for regulation of proliferation and differentiation of human bone marrow-derived mesenchymal stem cells to myocardial phenotypes: serum starvation without aid of chemical agents and prevention of spontaneous stem cell transformation enhanced by the matrix environment. J Tissue Eng Regen Med 2013; 10:E1-13. [PMID: 23897724 DOI: 10.1002/term.1754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 02/14/2013] [Accepted: 03/20/2013] [Indexed: 12/28/2022]
Abstract
Niche appears important for preventing the spontaneous differentiation or senescence that cells undergo during in vitro expansion. In the present study, it was revealed that human bone marrow-derived mesenchymal stem cells (hBM-MSCs) undergo senescence-related differentiation into the myocardial lineage in vitro without any induction treatment. This phenomenon occurred over the whole population of MCSs, much different from conventional differentiation with limited frequency of occurrence, and was accompanied by a change of morphology into large, flat cells with impeded proliferation, which are the representative indications of MSC senescence. By culturing MSCs under several culture conditions, it was determined that induction treatment with 5-azacytidine was not associated with the phenomenon, but the serum-starvation condition, under which proliferation is severely hampered, caused senescence progression and upregulation of cardiac markers. Nevertheless, MSCs gradually developed a myocardial phenotype under normal culture conditions over a prolonged culture period and heterogeneous populations were formed. In perspectives of clinical applications, this must be prevented for fair and consistent outcomes. Hence, the biomimetic 'niche' was constituted for hBM-MSCs by cultivating on a conventionally available extracellular matrix (ECM). Consequently, cells on ECM regained a spindle-shape morphology, increased in proliferation rate by two-fold and showed decreased expression of cardiac markers at both the mRNA and protein levels. In conclusion, the outcome indicates that progression of MSC senescence may occur via myocardial differentiation during in vitro polystyrene culture, and this can be overcome by employing appropriate ECM culture techniques.
Collapse
Affiliation(s)
- Jae Hyung Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul, 120-752, Korea
| | - Sang-Hyun Shin
- Department of Medical Engineering, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul, 120-752, Korea
| | - Tian Zhu Li
- Department of Medical Engineering, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul, 120-752, Korea
| | - Hwal Suh
- Department of Medical Engineering, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul, 120-752, Korea
| |
Collapse
|