1
|
Ledford BT, Chen M, Van Dyke M, Barron C, Zhang X, Cartaya A, Zheng Y, Ceylan A, Goldstein A, He JQ. Keratose Hydrogel Drives Differentiation of Cardiac Vascular Smooth Muscle Progenitor Cells: Implications in Ischemic Treatment. Stem Cell Rev Rep 2023; 19:2341-2360. [PMID: 37392292 DOI: 10.1007/s12015-023-10574-6] [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] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
Peripheral artery disease (PAD) is a common vascular disorder in the extremity of limbs with limited clinical treatments. Stem cells hold great promise for the treatment of PAD, but their therapeutic efficiency is limited due to multiple factors, such as poor engraftment and non-optimal selection of cell type. To date, stem cells from a variety of tissue sources have been tested, but little information is available regarding vascular smooth muscle cells (VSMCs) for PAD therapy. The present study examines the effects of keratose (KOS) hydrogels on c-kit+/CD31- cardiac vascular smooth muscle progenitor cell (cVSMPC) differentiation and the therapeutic potential of the resultant VSMCs in a mouse hindlimb ischemic model of PAD. The results demonstrated that KOS but not collagen hydrogel was able to drive the majority of cVSMPCs into functional VSMCs in a defined Knockout serum replacement (SR) medium in the absence of differentiation inducers. This effect could be inhibited by TGF-β1 antagonists. Further, KOS hydrogel increased expression of TGF-β1-associated proteins and modulated the level of free TGF-β1 during differentiation. Finally, transplantation of KOS-driven VSMCs significantly increased blood flow and vascular densities of ischemic hindlimbs. These findings indicate that TGF-β1 signaling is involved in KOS hydrogel-preferred VSMC differentiation and that enhanced blood flow are likely resulted from angiogenesis and/or arteriogenesis induced by transplanted VSMCs.
Collapse
Affiliation(s)
- Benjamin T Ledford
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Miao Chen
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Mark Van Dyke
- Department of Biomedical Engineering, College of Engineering, University of Arizona, Tucson, AZ, 85721, USA
| | - Catherine Barron
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Xiaonan Zhang
- Beijing Yulong Shengshi Biotechnology, Haidian District, Beijing, 100085, China
| | - Aurora Cartaya
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Youjing Zheng
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Ahmet Ceylan
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Aaron Goldstein
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jia-Qiang He
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
2
|
Takács R, Kovács P, Ebeid RA, Almássy J, Fodor J, Ducza L, Barrett-Jolley R, Lewis R, Matta C. Ca2+-Activated K+ Channels in Progenitor Cells of Musculoskeletal Tissues: A Narrative Review. Int J Mol Sci 2023; 24:ijms24076796. [PMID: 37047767 PMCID: PMC10095002 DOI: 10.3390/ijms24076796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Musculoskeletal disorders represent one of the main causes of disability worldwide, and their prevalence is predicted to increase in the coming decades. Stem cell therapy may be a promising option for the treatment of some of the musculoskeletal diseases. Although significant progress has been made in musculoskeletal stem cell research, osteoarthritis, the most-common musculoskeletal disorder, still lacks curative treatment. To fine-tune stem-cell-based therapy, it is necessary to focus on the underlying biological mechanisms. Ion channels and the bioelectric signals they generate control the proliferation, differentiation, and migration of musculoskeletal progenitor cells. Calcium- and voltage-activated potassium (KCa) channels are key players in cell physiology in cells of the musculoskeletal system. This review article focused on the big conductance (BK) KCa channels. The regulatory function of BK channels requires interactions with diverse sets of proteins that have different functions in tissue-resident stem cells. In this narrative review article, we discuss the main ion channels of musculoskeletal stem cells, with a focus on calcium-dependent potassium channels, especially on the large conductance BK channel. We review their expression and function in progenitor cell proliferation, differentiation, and migration and highlight gaps in current knowledge on their involvement in musculoskeletal diseases.
Collapse
Affiliation(s)
- Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Rana Abdelsattar Ebeid
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, Semmelweis University, H-1428 Budapest, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Ducza
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 3GA, UK
| | - Rebecca Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
3
|
Esophageal wound healing by aligned smooth muscle cell-laden nanofibrous patch. Mater Today Bio 2023; 19:100564. [PMID: 36747583 PMCID: PMC9898453 DOI: 10.1016/j.mtbio.2023.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
The esophagus exhibits peristalsis via contraction of circularly and longitudinally aligned smooth muscles, and esophageal replacement is required if there is a critical-sized wound. In this study, we proposed to reconstruct esophageal tissues using cell electrospinning (CE), an advanced technique for encapsulating living cells into fibers that allows control of the direction of fiber deposition. After treatment with transforming growth factor-β, mesenchymal stem cell-derived smooth muscle cells (SMCs) were utilized for cell electrospinning or three-dimensional bioprinting to compare the effects of aligned micropatterns on cell morphology. CE resulted in SMCs with uniaxially arranged and elongated cell morphology with upregulated expression levels of SMC-specific markers, including connexin 43, smooth muscle protein 22 alpha (SM22α), desmin, and smoothelin. When SMC-laden nanofibrous patches were transplanted into a rat esophageal defect model, the SMC patch promoted regeneration of esophageal wounds with an increased number of newly formed blood vessels and enhanced the SMC-specific markers of SM22α and vimentin. Taken together, CE with its advantages, such as guidance of highly elongated, aligned cell morphology and accelerated SMC differentiation, can be an efficient strategy to reconstruct smooth muscle tissues and treat esophageal perforation.
Collapse
|
4
|
Monteiro JP, Ferreira HB, Melo T, Flanagan C, Urbani N, Neves J, Domingues P, Domingues MR. The plasma phospholipidome of the bottlenose dolphin ( Tursiops truncatus) is modulated by both sex and developmental stage. Mol Omics 2023; 19:35-47. [PMID: 36314173 DOI: 10.1039/d2mo00202g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lipidomics represent a valid complementary tool to the biochemical analysis of plasma in humans. However, in cetaceans, these tools have been unexplored. Here, we evaluated how the plasma lipid composition of Tursiops truncatus is modulated by developmental stage and sex, aiming at a potential use of lipidomics in integrated strategies to monitor cetacean health. We characterized the fatty acid profile and detected a total of 26 fatty acids in T. truncatus plasma. The most abundant fatty acids were palmitic acid (C16:0), stearic acid (C18:0) and oleic acid (C18:1n-9). Interestingly, there are consistent differences between the fatty acid profile of mature female and mature male specimens. Phospholipidome analysis identified 320 different lipid species belonging to phosphatidylcholine (PC, 105 lipid species), lysophosphatidylcholine (42), phosphatidylethanolamine (PE, 67), lysophosphatidylethanolamine (18), phosphatidylglycerol (14), lysophosphatidylglycerol (8), phosphatidylinositol (14), lysophosphatidylinositol (2), phosphatidylserine (3), sphingomyelin (45) and ceramides (2) classes. The statistical analysis of the phospholipidome showed that its composition allows discriminating mature animals between sexes and mature males from immature males. Notably, discrimination between sexes is mainly determined by the contents of PE plasmalogens and lysophospholipids (LPC and LPE), while the differences between mature and immature male animals were mainly determined by the levels of PC lipids. This is the first time that a correlation between developmental stage and sex and the lipid composition of the plasma has been established in cetaceans. Being able to discern between age and sex-related changes is an encouraging step towards using these tools to also detect differences related to disease/dysfunction processes.
Collapse
Affiliation(s)
- João P Monteiro
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Helena B Ferreira
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Tânia Melo
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | | | | | | | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
5
|
Li C, Wang B. Mesenchymal Stem/Stromal Cells in Progressive Fibrogenic Involvement and Anti-Fibrosis Therapeutic Properties. Front Cell Dev Biol 2022; 10:902677. [PMID: 35721482 PMCID: PMC9198494 DOI: 10.3389/fcell.2022.902677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/13/2022] [Indexed: 11/22/2022] Open
Abstract
Fibrosis refers to the connective tissue deposition and stiffness usually as a result of injury. Fibrosis tissue-resident mesenchymal cells, including fibroblasts, myofibroblast, smooth muscle cells, and mesenchymal stem/stromal cells (MSCs), are major players in fibrogenic processes under certain contexts. Acknowledging differentiation potential of MSCs to the aforementioned other types of mesenchymal cell lineages is essential for better understanding of MSCs’ substantial contributions to progressive fibrogenesis. MSCs may represent a potential therapeutic option for fibrosis resolution owing to their unique pleiotropic functions and therapeutic properties. Currently, clinical trial efforts using MSCs and MSC-based products are underway but clinical data collected by the early phase trials are insufficient to offer better support for the MSC-based anti-fibrotic therapies. Given that MSCs are involved in the coagulation through releasing tissue factor, MSCs can retain procoagulant activity to be associated with fibrogenic disease development. Therefore, MSCs’ functional benefits in translational applications need to be carefully balanced with their potential risks.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People’s Hospital of Zhengzhou University and Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan University, Zhengzhou, China
- *Correspondence: Chenghai Li, ; Bin Wang,
| | - Bin Wang
- Department of Neurosurgery, People’s Hospital of Zhengzhou University and Henan Provincial People’s Hospital, Zhengzhou, China
- *Correspondence: Chenghai Li, ; Bin Wang,
| |
Collapse
|
6
|
Yu L, Qiu C, Chen R. A narrative review of research advances in the study of molecular markers of airway smooth muscle cells. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:375. [PMID: 35434039 PMCID: PMC9011254 DOI: 10.21037/atm-22-800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Abstract
Background and Objective Airway smooth muscle cells (ASMCs) are an important component of the airway. Their thickening and proliferation are important in pathological situations, such as airway remodeling in asthma, but their origin remains unclear. Therefore, characterizing molecular markers of ASMCs were sought to identify the source of increased ASMCs in asthmatic airway remodeling. Methods Articles for this review were derived from a review of the literature related to surface markers and biological properties of ASMCs and smooth muscle cells (SMCs) using PubMed, Google Scholar, and Web of Science. Key Content and Findings This review discusses several SMC molecular markers, describes the different developmental stages of SMCs that express different molecular markers, and summarizes several classical SMC molecular markers. However, the establishment of a specific molecular marker detection system for ASMCs still faces great challenges. Conclusions Although there is no recognized molecular marker detection system for ASMCs, and the study of the properties and sources of increased ASMCs in asthma airway remodeling is still in a state of exploration, the future is promising. Among the SMC markers described in this review, Myosin heavy chain 11 (MYH11) is a molecular marker for mature SMCs and Transgelin (TAGLN) is an early marker for SMC differentiation, and different molecular markers or combinations of molecular markers can be selected for the identification of the properties and sources of increased ASMCs in asthma airway remodeling according to the differentiation period and research needs.
Collapse
Affiliation(s)
- Li Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Rongchang Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| |
Collapse
|
7
|
Zhang M, Che C, Cheng J, Li P, Yang Y. Ion channels in stem cells and their roles in stem cell biology and vascular diseases. J Mol Cell Cardiol 2022; 166:63-73. [PMID: 35143836 DOI: 10.1016/j.yjmcc.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
Abstract
Stem cell therapy may be a promising option for the treatment of vascular diseases. In recent years, significant progress has been made in stem cell research, especially in the mechanism of stem cell activation, homing and differentiation in vascular repair and reconstruction. Current research on stem cells focuses on protein expression and transcriptional networks. Ion channels are considered to be the basis for the generation of bioelectrical signals, which control the proliferation, differentiation and migration of various cell types. Although heterogeneity of multiple ion channels has been found in different types of stem cells, it is unclear whether the heterogeneous expression of ion channels is related to different cell subpopulations and/or different stages of the cell cycle. There is still a long way to go in clinical treatment by using the regulation of stem cell ion channels. In this review, we reviewed the main ion channels found on stem cells, their expression and function in stem cell proliferation, differentiation and migration, and the research status of stem cells' involvement in vascular diseases.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Chang Che
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| |
Collapse
|
8
|
Park R, Yoon JW, Lee JH, Hong SW, Kim JH. Phenotypic change of mesenchymal stem cells into smooth muscle cells regulated by dynamic cell-surface interactions on patterned arrays of ultrathin graphene oxide substrates. J Nanobiotechnology 2022; 20:17. [PMID: 34983551 PMCID: PMC8725258 DOI: 10.1186/s12951-021-01225-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/23/2021] [Indexed: 01/13/2023] Open
Abstract
The topographical interface of the extracellular environment has been appreciated as a principal biophysical regulator for modulating cell functions, such as adhesion, migration, proliferation, and differentiation. Despite the existed approaches that use two-dimensional nanomaterials to provide beneficial effects, opportunities evaluating their impact on stem cells remain open to elicit unprecedented cellular responses. Herein, we report an ultrathin cell-culture platform with potential-responsive nanoscale biointerfaces for monitoring mesenchymal stem cells (MSCs). We designed an intriguing nanostructured array through self-assembly of graphene oxide sheets and subsequent lithographical patterning method to produce chemophysically defined regions. MSCs cultured on anisotropic micro/nanoscale patterned substrate were spontaneously organized in a highly ordered configuration mainly due to the cell-repellent interactions. Moreover, the spatially aligned MSCs were spontaneously differentiated into smooth muscle cells upon the specific crosstalk between cells. This work provides a robust strategy for directing stem cells and differentiation, which can be utilized as a potential cell culture platform to understand cell-substrate or cell-cell interactions, further developing tissue repair and stem cell-based therapies.
Collapse
Affiliation(s)
- Rowoon Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, 46241, Busan, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, School of Medicine, Pusan National University, 50612, Yangsan, Republic of Korea
| | - Jin-Ho Lee
- Department of Biomedical Convergence Engineering, Pusan National University, 50612, Yangsan, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, Pusan National University, 46241, Busan, Republic of Korea.
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, 50612, Yangsan, Republic of Korea.
| |
Collapse
|
9
|
Modulation of Mesenchymal Stem Cells for Enhanced Therapeutic Utility in Ischemic Vascular Diseases. Int J Mol Sci 2021; 23:ijms23010249. [PMID: 35008675 PMCID: PMC8745455 DOI: 10.3390/ijms23010249] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells are multipotent stem cells isolated from various tissue sources, including but not limited to bone marrow, adipose, umbilical cord, and Wharton Jelly. Although cell-mediated mechanisms have been reported, the therapeutic effect of MSCs is now recognized to be primarily mediated via paracrine effects through the secretion of bioactive molecules, known as the “secretome”. The regenerative benefit of the secretome has been attributed to trophic factors and cytokines that play neuroprotective, anti-angiogenic/pro-angiogenic, anti-inflammatory, and immune-modulatory roles. The advancement of autologous MSCs therapy can be hindered when introduced back into a hostile/disease environment. Barriers include impaired endogenous MSCs function, limited post-transplantation cell viability, and altered immune-modulatory efficiency. Although secretome-based therapeutics have gained popularity, many translational hurdles, including the heterogeneity of MSCs, limited proliferation potential, and the complex nature of the secretome, have impeded the progress. This review will discuss the experimental and clinical impact of restoring the functional capabilities of MSCs prior to transplantation and the progress in secretome therapies involving extracellular vesicles. Modulation and utilization of MSCs–secretome are most likely to serve as an effective strategy for promoting their ultimate success as therapeutic modulators.
Collapse
|
10
|
Walters B, Turner PA, Rolauffs B, Hart ML, Stegemann JP. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021; 10:cells10113123. [PMID: 34831345 PMCID: PMC8624888 DOI: 10.3390/cells10113123] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are an abundant and easily accessible multipotent stem cell source with potential application in smooth muscle regeneration strategies. In 3D collagen hydrogels, we investigated whether sustained release of growth factors (GF) PDGF-AB and TGF-β1 from GF-loaded microspheres could induce a smooth muscle cell (SMC) phenotype in ASCs, and if the addition of uniaxial cyclic stretch could enhance the differentiation level. This study demonstrated that the combination of cyclic stretch and GF release over time from loaded microspheres potentiated the differentiation of ASCs, as quantified by protein expression of early to late SMC differentiation markers (SMA, TGLN and smooth muscle MHC). The delivery of GFs via microspheres produced large ASCs with a spindle-shaped, elongated SMC-like morphology. Cyclic strain produced the largest, longest, and most spindle-shaped cells regardless of the presence or absence of growth factors or the growth factor delivery method. Protein expression and cell morphology data confirmed that the sustained release of GFs from GF-loaded microspheres can be used to promote the differentiation of ASCs into SMCs and that the addition of uniaxial cyclic stretch significantly enhances the differentiation level, as quantified by intermediate and late SMC markers and a SMC-like elongated cell morphology.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Paul A. Turner
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
| | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| |
Collapse
|
11
|
Khazaei S, Keshavarz G, Bozorgi A, Nazari H, Khazaei M. Adipose tissue-derived stem cells: a comparative review on isolation, culture, and differentiation methods. Cell Tissue Bank 2021; 23:1-16. [PMID: 33616792 DOI: 10.1007/s10561-021-09905-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023]
Abstract
Adipose tissue-derived stem cells (ADSCs) are an available source of mesenchymal stem cells with the appropriate capacity to in vitro survive, propagate, and differentiate into cells from three lineages of ectoderm, mesoderm, and endoderm. The biological features of ADSCs depend on the donor physiology and health status, isolation procedure, culture conditions, and differentiation protocols used. Adipose tissue samples are provided by surgery and lipoaspiration-based methods and subjected to various mechanical and chemical digestion techniques to finally generate a heterogeneous mixture named stromal vascular fraction (SVF). ADSCs are purified through varied cell populations that exist within SVF and cultured under standard conditions to give rise to a highly rich resource of stem cells directly applied in the clinic or differentiated into a wide range of cells. The development and optimization of conventional isolation, expansion, and differentiation methods seem noteworthy to preserve the desirable biological functions of ADSCs in pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Saber Khazaei
- Department of Endodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazal Keshavarz
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Azam Bozorgi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Nazari
- Department of Orofacial Surgery, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
12
|
Song A, Wang J, Tong Y, Fang J, Zhang Y, Zhang H, Ruan H, Wang K, Liu Y. BKCa channels regulate the immunomodulatory properties of WJ-MSCs by affecting the exosome protein profiles during the inflammatory response. Stem Cell Res Ther 2020; 11:440. [PMID: 33059770 PMCID: PMC7560248 DOI: 10.1186/s13287-020-01952-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) from the human umbilical cord have been studied extensively due to their immunomodulatory functions. Large-conductance Ca2+-activated K+ (BKCa channels) channels are involved in many inflammatory responses, but their involvement in the anti-inflammatory activity of WJ-MSCs is unknown. The underlying molecular mechanism, through which BKCa channels mediate the immunomodulation of WJ-MSC, which may include changes in exosomes proteomics, has not yet been clarified. Methods Alizarin staining, Oil Red O staining, and flow cytometry were used to identify WJ-MSCs, which were isolated from human umbilical cord Wharton’s jelly. BKCa channels were detected in WJ-MSCs using western blotting, real-time polymerase chain reaction (real-time PCR), and electrophysiology, and cytokine expression was examined using real-time PCR and enzyme-linked immunosorbent assays (ELISAs). Exosomes were characterized using transmission electron microscopy and nanoparticle tracking analysis. Proteomics analysis was performed to explore exosomal proteomic profiles. Results The cells derived from human umbilical cord Wharton’s jelly were identified as MSCs. BKCa channels were detected in the isolated WJ-MSCs, and the expression of these channels increased after lipopolysaccharide (LPS) stimulation. BKCa channels blockade in LPS-treated WJ-MSCs induced apoptosis and decreased interleukin-6 (IL-6) expression. Furthermore, THP-1 cells (human monocytic cell line) stimulated with LPS/interferon gamma (IFN-γ) produced more anti-inflammatory cytokines after treatment with exosomes derived from BKCa channel-knockdown WJ-MSCs (si-exo). We also observed altered expression of mitochondrial ATP synthase alpha subunit (ATP5A1), filamin B, and other proteins in si-exo, which might increase the anti-inflammatory activity of macrophages. Conclusions Our study described the functional expression of BKCa channels in WJ-MSCs, and BKCa channels regulated the immunomodulatory properties of WJ-MSCs by affecting the exosomal protein profiles during the inflammatory response.
Collapse
Affiliation(s)
- Ahui Song
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Jingjing Wang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Yan Tong
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Junyan Fang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Yi Zhang
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Huiping Zhang
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Hongqiang Ruan
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Kai Wang
- The Clinical and Translational Research Center Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yingli Liu
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China.
| |
Collapse
|
13
|
Thurner M, Deutsch M, Janke K, Messner F, Kreutzer C, Beyl S, Couillard-Després S, Hering S, Troppmair J, Marksteiner R. Generation of myogenic progenitor cell-derived smooth muscle cells for sphincter regeneration. Stem Cell Res Ther 2020; 11:233. [PMID: 32532320 PMCID: PMC7291744 DOI: 10.1186/s13287-020-01749-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/15/2020] [Accepted: 05/28/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Degeneration of smooth muscles in sphincters can cause debilitating diseases such as fecal incontinence. Skeletal muscle-derived cells have been effectively used in clinics for the regeneration of the skeletal muscle sphincters, such as the external anal or urinary sphincter. However, little is known about the in vitro smooth muscle differentiation potential and in vivo regenerative potential of skeletal muscle-derived cells. METHODS Myogenic progenitor cells (MPC) were isolated from the skeletal muscle and analyzed by flow cytometry and in vitro differentiation assays. The differentiation of MPC to smooth muscle cells (MPC-SMC) was evaluated by immunofluorescence, flow cytometry, patch-clamp, collagen contraction, and microarray gene expression analysis. In vivo engraftment of MPC-SMC was monitored by transplanting reporter protein-expressing cells into the pyloric sphincter of immunodeficient mice. RESULTS MPC derived from human skeletal muscle expressed mesenchymal surface markers and exhibit skeletal myogenic differentiation potential in vitro. In contrast, they lack hematopoietic surface marker, as well as adipogenic, osteogenic, and chondrogenic differentiation potential in vitro. Cultivation of MPC in smooth muscle differentiation medium significantly increases the fraction of alpha smooth muscle actin (aSMA) and smoothelin-positive cells, while leaving the number of desmin-positive cells unchanged. Smooth muscle-differentiated MPC (MPC-SMC) exhibit increased expression of smooth muscle-related genes, significantly enhanced numbers of CD146- and CD49a-positive cells, and in vitro contractility and express functional Cav and Kv channels. MPC to MPC-SMC differentiation was also accompanied by a reduction in their skeletal muscle differentiation potential. Upon removal of the smooth muscle differentiation medium, a major fraction of MPC-SMC remained positive for aSMA, suggesting the definitive acquisition of their phenotype. Transplantation of murine MPC-SMC into the mouse pyloric sphincter revealed engraftment of MPC-SMC based on aSMA protein expression within the host smooth muscle tissue. CONCLUSIONS Our work confirms the ability of MPC to give rise to smooth muscle cells (MPC-SMC) with a well-defined and stable phenotype. Moreover, the engraftment of in vitro-differentiated murine MPC-SMC into the pyloric sphincter in vivo underscores the potential of this cell population as a novel cell therapeutic treatment for smooth muscle regeneration of sphincters.
Collapse
Affiliation(s)
- Marco Thurner
- Innovacell Biotechnologie AG, Mitterweg 24, 6020, Innsbruck, Austria.
- Daniel Swarovski Research Laboratory (DSL), Visceral Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria.
| | - Martin Deutsch
- Innovacell Biotechnologie AG, Mitterweg 24, 6020, Innsbruck, Austria
| | - Katrin Janke
- Innovacell Biotechnologie AG, Mitterweg 24, 6020, Innsbruck, Austria
| | - Franka Messner
- Daniel Swarovski Research Laboratory (DSL), Visceral Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Kreutzer
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Stanislav Beyl
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Sébastien Couillard-Després
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Steffen Hering
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory (DSL), Visceral Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
14
|
Ni H, Zhao Y, Ji Y, Shen J, Xiang M, Xie Y. Adipose-derived stem cells contribute to cardiovascular remodeling. Aging (Albany NY) 2019; 11:11756-11769. [PMID: 31800397 PMCID: PMC6932876 DOI: 10.18632/aging.102491] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/17/2019] [Indexed: 02/06/2023]
Abstract
Obesity is an independent risk factor for cardiovascular disease. Adipose tissue was initially thought to be involved in metabolism through paracrine. Recent researches discovered mesenchymal stem cells inside adipose tissue which could differentiate into vascular lineages in vitro and in vivo, participating vascular remodeling. However, there were few researches focusing on distinct characteristics and functions of adipose-derived stem cells (ADSCs) from different regions. This is the first comprehensive review demonstrating the variances of ADSCs from the perspective of their origins.
Collapse
Affiliation(s)
- Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiming Zhao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Lau S, Klingenberg M, Mrugalla A, Helms F, Sedding D, Haverich A, Wilhelmi M, Böer U. Biochemical Myogenic Differentiation of Adipogenic Stem Cells Is Donor Dependent and Requires Sound Characterization. Tissue Eng Part A 2019; 25:936-948. [PMID: 30648499 DOI: 10.1089/ten.tea.2018.0172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPACT STATEMENT We here showed that even under optimized conditions for biochemical differentiation of adipose-derived stem cells (with respect to a pronounced marker protein expression for a reasonable period of time) it was not possible to obtain functional smooth muscle cells from all donors. Moreover, an underestimated role may play the effect of the scaffold material on smooth muscle cell functionality. Both aspects are crucial for the successful tissue engineering of the vascular medial layer combining autologous cells with a suitable scaffold material and thus should be thoroughly addressed in each individualized therapeutic approach.
Collapse
Affiliation(s)
- Skadi Lau
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Melanie Klingenberg
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Anna Mrugalla
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany
| | - Florian Helms
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany
| | - Daniel Sedding
- 3Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Mathias Wilhelmi
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ulrike Böer
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Di Stefano AB, Massihnia D, Grisafi F, Castiglia M, Toia F, Montesano L, Russo A, Moschella F, Cordova A. Adipose tissue, angiogenesis and angio-MIR under physiological and pathological conditions. Eur J Cell Biol 2019; 98:53-64. [DOI: 10.1016/j.ejcb.2018.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
|
17
|
Zhang X, Simmons CA, Santerre JP. Alterations of MEK1/2-ERK1/2, IFNγ and Smad2/3 associated Signalling pathways during cryopreservation of ASCs affect their differentiation towards VSMC-like cells. Stem Cell Res 2018; 32:115-125. [DOI: 10.1016/j.scr.2018.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/06/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
|
18
|
Tang X, Li B, Ding J, Zhang L, Zhu L. A gene expression profile analysis of the differentiation of muscle-derived stem cells into smooth muscle cells from sheep. Am J Transl Res 2018; 10:1195-1204. [PMID: 29736212 PMCID: PMC5934578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To analyze gene expression profiles analysis during the differentiation of sheep muscle-derived stem cells (MDSCs) into smooth muscle cells (SMCs) in sheep. METHODS A modified preplate technique was employed to isolation of the MDSCs from sheep. The MDSCs were subjected to flow cytometry analysis targeting CD44, CD31, CD45, CD14, and CD49f and were treated with TGF-β1 at a concentration of 10 ng/ml for ten days. The expression levels of smooth muscle α-actin (α-SMA) and calponin after treatment with TGF-β1 were determined by western blotting and immunofluorescence staining. A microarray analysis was performed to screen for differentially expressed genes (DEGs) during MDSC differentiation using total RNA extracted from MDSCs and SMCs generated from MDSCs. Molecule Annotation System (MAS) 3.0, which employs KEGG (Kyoto Encyclopedia of Genes and Genomes) and GO (Gene Ontology) Consortium annotations, was used to identify global biological trends in the gene expression data. RESULTS The expression levels of the SMC-specific contractile proteins α-SMA and calponin were dramatically increased after treatment with TGF-β1. Immunofluorescece staining showed that the TGF-β1-treated MDSCs were positive for α-SMA. We identified 486 genes that were differentially expressed between the MDSCs and TGF-β1-treated MDSCs. 260 of which were up-regulated and 226 of which were down-regulated. Twenty-one genes exhibited a greater than ten-fold change, 13 of which were up-regulated and 8 of which were down-regulated. TGF-β1 treatment up-regulated both the SMAD and MAPK signaling pathways during the differentiation of these sheep cells. The PPAR and Wnt signaling pathways were also found to be involved in the differentiation process. CONCLUSIONS TGF-β1 can successfully induce the differentiation of sheep MDSCs into SMCs. For the first time, we analyzed the gene expression profiles associated with this differentiation process, and the results showed that both the SMAD and MAPK signaling pathways are involved. This study indicated that multiple signaling networks coordinate the development and differentiation of MDSCs into SMCs.
Collapse
Affiliation(s)
- Xiang Tang
- Department of Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical UniversityGuangzhou, China
| | - Bin Li
- Department of Gynecology and Obstetrics, Shaanxi Provincial People’s HospitalXi’an, China
| | - Jing Ding
- Department of Gynecology, Harbin Medical University Cancer HospitalHarbin, China
| | - Lei Zhang
- Department of Gynecology and Obstetrics, Peking University First HospitalPeking, China
| | - Lan Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijing, China
| |
Collapse
|
19
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
20
|
Deriving vascular smooth muscle cells from mesenchymal stromal cells: Evolving differentiation strategies and current understanding of their mechanisms. Biomaterials 2017; 145:9-22. [PMID: 28843066 DOI: 10.1016/j.biomaterials.2017.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play essential roles in regulating blood vessel form and function. Regeneration of functional vascular smooth muscle tissue to repair vascular diseases is an area of intense research in tissue engineering and regenerative medicine. For functional vascular smooth muscle tissue regeneration to become a practical therapy over the next decade, the field will need to have access to VSMC sources that are effective, robust and safe. While pluripotent stem cells hold good future promise to this end, more immediate translation is expected to come from approaches that generate functional VSMCs from adult sources of multipotent adipose-derived and bone marrow-derived mesenchymal stromal cells (ASCs and BMSCs). The research to this end is extensive and is dominated by studies relating to classical biochemical signalling molecules used to induce differentiation of ASCs and BMSCs. However, prolonged use of the biochemical induction factors is costly and can cause potential endotoxin contamination in the culture. Over recent years several non-traditional differentiation approaches have been devised to mimic defined aspects of the native micro-environment in which VSMCs reside to contribute to the differentiation of VSMC-like cells from ASCs and BMSCs. In this review, the promises and limitations of several non-traditional culture approaches (e.g., co-culture, biomechanical, and biomaterial stimuli) targeting VSMC differentiation are discussed. The extensive crosstalk between the underlying signalling cascades are delineated and put into a translational context. It is expected that this review will not only provide significant insight into VSMC differentiation strategies for vascular smooth muscle tissue engineering applications, but will also highlight the fundamental importance of engineering the cellular microenvironment on multiple scales (with consideration of different combinatorial pathways) in order to direct cell differentiation fate and obtain cells of a desired and stable phenotype. These strategies may ultimately be applied to different sources of stem cells in the future for a range of biomaterial and tissue engineering disciplines.
Collapse
|
21
|
Yipeng J, Yongde X, Yuanyi W, Jilei S, Jiaxiang G, Jiangping G, Yong Y. Microtissues Enhance Smooth Muscle Differentiation and Cell Viability of hADSCs for Three Dimensional Bioprinting. Front Physiol 2017; 8:534. [PMID: 28790931 PMCID: PMC5524823 DOI: 10.3389/fphys.2017.00534] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/10/2017] [Indexed: 12/22/2022] Open
Abstract
Smooth muscle differentiated human adipose derived stem cells (hADSCs) provide a crucial stem cell source for urinary tissue engineering, but the induction of hADSCs for smooth muscle differentiation still has several issues to overcome, including a relatively long induction time and equipment dependence, which limits access to abundant stem cells within a short period of time for further application. Three-dimensional (3D) bioprinting holds great promise in regenerative medicine due to its controllable construction of a designed 3D structure. When evenly mixed with bioink, stem cells can be spatially distributed within a bioprinted 3D structure, thus avoiding drawbacks such as, stem cell detachment in a conventional cell-scaffold strategy. Notwithstanding the advantages mentioned above, cell viability is often compromised during 3D bioprinting, which is often due to pressure during the bioprinting process. The objective of our study was to improve the efficiency of hADSC smooth muscle differentiation and cell viability of a 3D bioprinted structure. Here, we employed the hanging-drop method to generate hADSC microtissues in a smooth muscle inductive medium containing human transforming growth factor β1 and bioprinted the induced microtissues onto a 3D structure. After 3 days of smooth muscle induction, the expression of α-smooth muscle actin and smoothelin was higher in microtissues than in their counterpart monolayer cultured hADSCs, as confirmed by immunofluorescence and western blotting analysis. The semi-quantitative assay showed that the expression of α-smooth muscle actin (α-SMA) was 0.218 ± 0.077 in MTs and 0.082 ± 0.007 in Controls; smoothelin expression was 0.319 ± 0.02 in MTs and 0.178 ± 0.06 in Controls. Induced MTs maintained their phenotype after the bioprinting process. Live/dead and cell count kit 8 assays showed that cell viability and cell proliferation in the 3D structure printed with microtissues were higher at all time points compared to the conventional single-cell bioprinting strategy (mean cell viability was 88.16 ± 3.98 vs. 61.76 ± 15% for microtissues and single-cells, respectively). These results provide a novel way to enhance the smooth muscle differentiation of hADSCs and a simple method to maintain better cell viability in 3D bioprinting.
Collapse
Affiliation(s)
- Jin Yipeng
- Department of Urology, Chinese PLA General HospitalBeijing, China
| | - Xu Yongde
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Wu Yuanyi
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Sun Jilei
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Guo Jiaxiang
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| | - Gao Jiangping
- Department of Urology, Chinese PLA General HospitalBeijing, China
| | - Yang Yong
- Department of Urology, First Affiliated Hospital of Chinese PLA General HospitalBeijing, China
| |
Collapse
|
22
|
Kim E, Park WS, Hong SH. Expression of Ion Channels in Perivascular Stem Cells derived from Human Umbilical Cords. Dev Reprod 2017; 21:11-18. [PMID: 28484740 PMCID: PMC5409206 DOI: 10.12717/dr.2017.21.1.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 12/29/2016] [Accepted: 01/05/2017] [Indexed: 11/17/2022]
Abstract
Potassium channels, the largest group of pore proteins, selectively regulate the flow of potassium (K+) ions across cell membranes. The activity and expression of K+ channels are critical for the maintenance of normal functions in vessels and neurons, and for the regulation of cell differentiation and maturation. However, their role and expression in stem cells have been poorly understood. In this study, we isolated perivascular stem cells (PVCs) from human umbilical cords and investigated the expression patterns of big-conductance Ca2+-activated K+ (BKCa) and voltage-dependent K+ (Kv) channels using the reverse transcription polymerase chain reaction. We also examined the effect of high glucose (HG, 25 mM) on expression levels of BKCa and Kv channels in PVCs. KCa1.1, KCaβ3, Kv1.3, Kv3.2, and Kv6.1 were detected in undifferentiated PVCs. In addition, HG treatment increased the amounts of BKCaβ3a, BKCaβ4, Kv1.3, Kv1.6, and Kv6.1 transcripts. These results suggested that ion channels may have important functions in the growth and differentiation of PVCs, which could be influenced by HG exposure.
Collapse
Affiliation(s)
- Eunbi Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
23
|
Elçin AE, Parmaksiz M, Dogan A, Seker S, Durkut S, Dalva K, Elçin YM. Differential gene expression profiling of human adipose stem cells differentiating into smooth muscle-like cells by TGFβ1/BMP4. Exp Cell Res 2017; 352:207-217. [DOI: 10.1016/j.yexcr.2017.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 12/18/2022]
|
24
|
Feng D, Nan H, Wang W, Yan L, Du P, Zuo L, Zhang K, Zhao M, Cui G. Expression and alteration of BK Ca channels in the sphincter of Oddi's from rabbits with hypercholesterolemia. Channels (Austin) 2017; 11:236-244. [PMID: 28102743 DOI: 10.1080/19336950.2017.1279369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study aimed to investigate the expression and function of BKCa channels in the Sphincter of Oddi (SO) in a rabbit model of hypercholesterolemia (HC). New Zealand white rabbits were randomly divided into 2 groups: the control group was fed standard chow (n = 18) whereas the high-cholesterol group was fed cholesterol-enriched chow containing 1.5% cholesterol (n = 18). The serum cholesterol level was significantly greater in the HC groups than in the control group, but there was no significant difference in body weight between the control and HC groups. Although the total protein expression of BKCa α- and β1-subunit was not significantly different between the control and HC groups, the Tyr-phosphorylation of BKCa α-subunit was significantly decreased in the HC group than in the control group. In addition, hypercholesterolemia significantly increased Acetylcholine (ACh)-induced contraction of the SO rings. Pretreatment with 30 μM NS1619, a BKCa channel agonist, significantly reduced ACh-induced contraction of the SO rings in HC rabbits. Moreover, pretreatment with 100 μM Na3OV4, a protein tyrosine phosphatase inhibitor, significantly reduced ACh-induced contraction of the SO rings in HC rabbits, whereas it significantly increased upon pretreating with 10 μM Genistein, a tyrosine kinase inhibitor. Whole-cell patch clamp recordings showed that BKCa current density was significantly lower in SOSMCs from HC group than that from control group. Our findings suggest that hypercholesterolemia-induced downregulation of BKCa channel, and Tyr-phosphorylation of BKCa α-subunit may contribute to the hyperresponsiveness of the SO ring in HC rabbits.
Collapse
Affiliation(s)
- Dan Feng
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Haiyan Nan
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Wen Wang
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Linfeng Yan
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Pang Du
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Lin Zuo
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Kun Zhang
- b Department of Pharmacology, School of Pharmacy , Fourth Military Medical University , Xi'an , China
| | - Minggao Zhao
- b Department of Pharmacology, School of Pharmacy , Fourth Military Medical University , Xi'an , China
| | - Guangbin Cui
- a Department of Radiology , Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| |
Collapse
|
25
|
Han KH, Kim AK, Kim DI. Therapeutic Potential of Human Mesenchymal Stem Cells for Treating Ischemic Limb Diseases. Int J Stem Cells 2016; 9:163-168. [PMID: 27871151 PMCID: PMC5155711 DOI: 10.15283/ijsc16053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2016] [Indexed: 12/19/2022] Open
Abstract
Ischemic limb diseases are induced by different obstructions of peripheral arteries. These obstructions result in insufficient nutrient and oxygen supplies to the extremities, thereby leading to severe tissue damage that is in turn related to severe morbidities and mortalities. Mesenchymal stem cells (MSCs) have been isolated from various sources. These cells are multipotent with respect to differentiation and are also characterized by migration, immune suppression, and secretion of paracrine factors. Mesenchymal stem cells have been proposed to have therapeutic potential for the treatment of ischemic limb diseases. In preclinical experiments, injection of single MSCs has been shown to increase angiogenesis and blood flow in ischemic hindlimb animal models; several molecular mechanisms of angiogenesis have also been elucidated. Furthermore, modified strategies have been developed for enhancing angiogenesis and the efficacy of MSCs. These strategies have demonstrated significant effects in pre-clinical studies. In clinical trials, MSCs have shown significant effects in the treatment of ischemic limb diseases. In this review, we focus on the therapeutic properties of human MSCs and the modified methods for enhancing angiogenesis in pre-clinical experiments. We also discuss the clinical applications of MSCs for treating limb ischemia.
Collapse
Affiliation(s)
- Kyu-Hyun Han
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ae-Kyeong Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Björninen M, Gilmore K, Pelto J, Seppänen-Kaijansinkko R, Kellomäki M, Miettinen S, Wallace G, Grijpma D, Haimi S. Electrically Stimulated Adipose Stem Cells on Polypyrrole-Coated Scaffolds for Smooth Muscle Tissue Engineering. Ann Biomed Eng 2016; 45:1015-1026. [DOI: 10.1007/s10439-016-1755-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/31/2016] [Indexed: 01/08/2023]
|
27
|
Heidari Kani M, Chan EC, Young RC, Butler T, Smith R, Paul JW. 3D Cell Culturing and Possibilities for Myometrial Tissue Engineering. Ann Biomed Eng 2016; 45:1746-1757. [PMID: 27770218 DOI: 10.1007/s10439-016-1749-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022]
Abstract
Research insights into uterine function and the mechanisms of labour have been hindered by the lack of suitable animal and cellular models. The use of traditional culturing methods limits the exploration of complex uterine functions, such as cell interactions, connectivity and contractile behaviour, as it fails to mimic the three-dimensional (3D) nature of uterine cell interactions in vivo. Animal models are an option, however, use of these models is constrained by ethical considerations as well as translational limitations to humans. Evidence indicates that these limitations can be overcome by using 3D culture systems, or 3D Bioprinters, to model the in vivo cytological architecture of the tissue in an in vitro environment. 3D cultured or 3D printed cells can be used to form an artificial tissue. This artificial tissue can not only be used as an appropriate model in which to study cellular function and organisation, but could also be used for regenerative medicine purposes including organ or tissue transplantation, organ donation and obstetric care. The current review describes recent developments in cell culture that can facilitate the development of myometrial 3D structures and tissue engineering applications.
Collapse
Affiliation(s)
- Minoo Heidari Kani
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia. .,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia. .,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia.
| | - Eng-Cheng Chan
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Roger C Young
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Trent Butler
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia.,John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Jonathan W Paul
- Mothers and Babies Research Centre, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia.,Priority Research Centre of Reproductive Science, University of Newcastle, Callaghan, NSW, 2308, Australia
| |
Collapse
|
28
|
Li D, Liu Q, Qi L, Dai X, Liu H, Wang Y. Low levels of TGF-β1 enhance human umbilical cord-derived mesenchymal stem cell fibronectin production and extend survival time in a rat model of lipopolysaccharide-induced acute lung injury. Mol Med Rep 2016; 14:1681-92. [PMID: 27357811 DOI: 10.3892/mmr.2016.5416] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 06/03/2016] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive cellular source for cell‑based therapy, tissue engineering and regenerative medicine. However, the use of MSCs is limited by their low incorporation rate in the graft environment. The majority of cells are lost from the graft within 1 month, due to reduced microenvironment or local inflammation at the graft site. The extracellular matrix (ECM) may assist the survival and expansion of MSCs. The present study aimed to identify an effective approach to increase ECM expression levels by MSCs in order to enhance the therapeutic effect and survival rate of MSCs at the injury site. The concentration‑dependent effect of transforming growth factor (TGF)‑β1 on human umbilical cord (hUC)‑MSC proliferation and expression of ECM genes was investigated. MSCs were successfully isolated, cultured and expanded from hUC. A low concentration of TGF‑β1 (0.1 ng/ml) exhibited the optimal effect on hUC‑MSC proliferation and markedly stimulated the expression of ECM genes, particularly fibronectin (FN). Furthermore, treatment with TGF‑β1 caused no alteration in the immunophenotype and differentiation capacity of MSCs. In vivo experiments in rats demonstrated that intravenous injection of control UC-MSCs or TGF-β1-pre-treated UC-MSCs reduced the severity of lipopolysaccharide-induced lung injury, assessed using histology, measurements of the wet‑dry lung weight ratio, and neutrophil count and protein concentration in bronchoalveolar lavage fluid. However, the short‑term (48 h) therapeutic effects of untreated and TGF‑β1‑pre‑treated UC‑MSCs were similar. The survival of MSCs in damaged lungs, determined by Sry gene expression levels, were significantly increased in MSCs pre‑treated with TGF‑β1. In conclusion, pre‑treatment of MSCs with a low concentration of TGF‑β1 enhanced the expression of ECM components, particularly FN, thus, improving the survival and potential therapeutic benefits of MSCs. Pre‑treatment of MSCs with TGF‑β1 may prolong the effective therapy time and represent an efficient therapeutic approach for tissue repair.
Collapse
Affiliation(s)
- Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qingshen Liu
- First Department of Surgery, Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Lei Qi
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaoyu Dai
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Huan Liu
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yunshan Wang
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250100, P.R. China
| |
Collapse
|
29
|
Parvizi M, Bolhuis-Versteeg LA, Poot AA, Harmsen MC. Efficient generation of smooth muscle cells from adipose-derived stromal cells by 3D mechanical stimulation can substitute the use of growth factors in vascular tissue engineering. Biotechnol J 2016; 11:932-44. [DOI: 10.1002/biot.201500519] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 12/24/2015] [Accepted: 03/09/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Mojtaba Parvizi
- University of Groningen; University Medical Center Groningen, Department of Pathology and Medical Biology; Groningen the Netherlands
| | | | - André A. Poot
- Biomaterials Science and Technology; University of Twente; Enschede the Netherlands
| | - Martin C. Harmsen
- University of Groningen; University Medical Center Groningen, Department of Pathology and Medical Biology; Groningen the Netherlands
| |
Collapse
|
30
|
Tao H, Han Z, Han ZC, Li Z. Proangiogenic Features of Mesenchymal Stem Cells and Their Therapeutic Applications. Stem Cells Int 2016; 2016:1314709. [PMID: 26880933 PMCID: PMC4736816 DOI: 10.1155/2016/1314709] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/04/2015] [Accepted: 11/29/2015] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown their therapeutic potency for treatment of cardiovascular diseases owing to their low immunogenicity, ease of isolation and expansion, and multipotency. As multipotent progenitors, MSCs have revealed their ability to differentiate into various cell types and could promote endogenous angiogenesis via microenvironmental modulation. Studies on cardiovascular diseases have demonstrated that transplanted MSCs could engraft at the injured sites and differentiate into cardiomyocytes and endothelial cells as well. Accordingly, several clinical trials using MSCs have been performed and revealed that MSCs may improve relevant clinical parameters in patients with vascular diseases. To fully comprehend the characteristics of MSCs, understanding their intrinsic property and associated modulations in tuning their behaviors as well as functions is indispensable for future clinical translation of MSC therapy. This review will focus on recent progresses on endothelial differentiation and potential clinical application of MSCs, with emphasis on therapeutic angiogenesis for treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Hongyan Tao
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| | - Zhibo Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zhong Chao Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| |
Collapse
|
31
|
Brun J, Lutz KA, Neumayer KMH, Klein G, Seeger T, Uynuk-Ool T, Wörgötter K, Schmid S, Kraushaar U, Guenther E, Rolauffs B, Aicher WK, Hart ML. Smooth Muscle-Like Cells Generated from Human Mesenchymal Stromal Cells Display Marker Gene Expression and Electrophysiological Competence Comparable to Bladder Smooth Muscle Cells. PLoS One 2015; 10:e0145153. [PMID: 26673782 PMCID: PMC4684225 DOI: 10.1371/journal.pone.0145153] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/30/2015] [Indexed: 12/19/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) differentiated toward a smooth muscle cell (SMC) phenotype may provide an alternative for investigators interested in regenerating urinary tract organs such as the bladder where autologous smooth muscle cells cannot be used or are unavailable. In this study we measured the effects of good manufacturing practice (GMP)-compliant expansion followed by myogenic differentiation of human MSCs on the expression of a range of contractile (from early to late) myogenic markers in relation to the electrophysiological parameters to assess the functional role of the differentiated MSCs and found that differentiation of MSCs associated with electrophysiological competence comparable to bladder SMCs. Within 1-2 weeks of myogenic differentiation, differentiating MSCs significantly expressed alpha smooth muscle actin (αSMA; ACTA2), transgelin (TAGLN), calponin (CNN1), and smooth muscle myosin heavy chain (SM-MHC; MYH11) according to qRT-PCR and/or immunofluorescence and Western blot. Voltage-gated Na+ current levels also increased within the same time period following myogenic differentiation. In contrast to undifferentiated MSCs, differentiated MSCs and bladder SMCs exhibited elevated cytosolic Ca2+ transients in response to K+-induced depolarization and contracted in response to K+ indicating functional maturation of differentiated MSCs. Depolarization was suppressed by Cd2+, an inhibitor of voltage-gated Ca2+-channels. The expression of Na+-channels was pharmacologically identified as the Nav1.4 subtype, while the K+ and Ca2+ ion channels were identified by gene expression of KCNMA1, CACNA1C and CACNA1H which encode for the large conductance Ca2+-activated K+ channel BKCa channels, Cav1.2 L-type Ca2+ channels and Cav3.2 T-type Ca2+ channels, respectively. This protocol may be used to differentiate adult MSCs into smooth muscle-like cells with an intermediate-to-late SMC contractile phenotype exhibiting voltage-gated ion channel activity comparable to bladder SMCs which may be important for urological regenerative medicine applications.
Collapse
Affiliation(s)
- Juliane Brun
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Katrin A. Lutz
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Katharina M. H. Neumayer
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Gerd Klein
- Center for Medical Research, University Medical Clinic, Department II, University of Tübingen, Tübingen, Germany
| | - Tanja Seeger
- Center for Medical Research, University Medical Clinic, Department II, University of Tübingen, Tübingen, Germany
| | - Tatiana Uynuk-Ool
- Siegfried Weller Institute for Trauma Research, Laboratory for Molecular Biomechanics, University of Tübingen, Tübingen, Germany
| | - Katharina Wörgötter
- Siegfried Weller Institute for Trauma Research, Laboratory for Molecular Biomechanics, University of Tübingen, Tübingen, Germany
| | - Sandra Schmid
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Department of Electrophysiology, Reutlingen, Germany
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Department of Electrophysiology, Reutlingen, Germany
| | - Elke Guenther
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Department of Electrophysiology, Reutlingen, Germany
| | - Bernd Rolauffs
- Siegfried Weller Institute for Trauma Research, Laboratory for Molecular Biomechanics, University of Tübingen, Tübingen, Germany
| | - Wilhelm K. Aicher
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Melanie L. Hart
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
- Siegfried Weller Institute for Trauma Research, Laboratory for Molecular Biomechanics, University of Tübingen, Tübingen, Germany
| |
Collapse
|
32
|
Dogan C, Halici Z, Topcu A, Cadirci E, Karakus E, Bayir Y, Selli J. Effects of amlodipine on ischaemia/reperfusion injury in the rat testis. Andrologia 2015; 48:441-52. [PMID: 26259852 DOI: 10.1111/and.12464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2015] [Indexed: 12/27/2022] Open
Abstract
The aim of this study was to examine the effects of amlodipine (AML) in rat testicular torsion/detorsion damage. In this study, rats were divided into eight groups: (i) sham; (ii) testicular ischaemia, 2 h of ischaemia; (iii) testicular ischaemia/reperfusion (I/R), 2 h of ischaemia followed by 2 h of reperfusion; (iv) ischaemia + AML (5 mg kg(-1)) administered 30 min before ischaemia; (v) ischaemia + AML (10 mg kg(-1)) administered 30 min before ischaemia; (vi) and (vii) I/R + AML (5 mg kg(-1)) and I/R + AML (10 mg kg(-1)) administered 1.5 h after the induction of ischaemia, respectively, and at the end of a 2-h ischaemia period and a 2-h reperfusion period applied; and (viii) sham + AML (10 mg kg(-1)). Significant decreases in levels of superoxide dismutase and glutathione were observed in ischaemia and reperfusion groups when compared with healthy controls. These antioxidant levels increased in AML groups while malondialdehyde levels significantly decreased. While increases in tumour necrosis factor-alpha and transforming growth factor-beta levels were found in the torsion and detorsion groups, significant decreases in the levels of these inflammatory cytokines were observed in the treatment groups. These results demonstrate that AML significantly produced protective effects on testis tissue damage that occurs in the torsion/detorsion model via biochemical, histopathological and molecular pathways.
Collapse
Affiliation(s)
- C Dogan
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Z Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - A Topcu
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - E Cadirci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - E Karakus
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Y Bayir
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - J Selli
- Department of Histology and Embryology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
33
|
Swaminathan G, Gadepalli VS, Stoilov I, Mecham RP, Rao RR, Ramamurthi A. Pro-elastogenic effects of bone marrow mesenchymal stem cell-derived smooth muscle cells on cultured aneurysmal smooth muscle cells. J Tissue Eng Regen Med 2014; 11:679-693. [DOI: 10.1002/term.1964] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 08/18/2014] [Accepted: 09/25/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Ganesh Swaminathan
- Department of Biomedical Engineering; Cleveland Clinic; Cleveland OH USA
- Department of Biology; University of Akron; Akron OH USA
| | - Venkat S. Gadepalli
- Department of Chemical and Life Science Engineering; Virginia Commonwealth University; Richmond VA USA
| | - Ivan Stoilov
- Department of Cell Biology and Physiology; Washington University; St. Louis MO USA
| | - Robert P. Mecham
- Department of Cell Biology and Physiology; Washington University; St. Louis MO USA
| | - Raj R. Rao
- Department of Chemical and Life Science Engineering; Virginia Commonwealth University; Richmond VA USA
| | - Anand Ramamurthi
- Department of Biomedical Engineering; Cleveland Clinic; Cleveland OH USA
- Department of Biology; University of Akron; Akron OH USA
| |
Collapse
|
34
|
Luo L, Lin T, Zheng S, Xie Z, Chen M, Lian G, Xu C, Wang H, Xie L. Adipose-derived stem cells attenuate pulmonary arterial hypertension and ameliorate pulmonary arterial remodeling in monocrotaline-induced pulmonary hypertensive rats. Clin Exp Hypertens 2014; 37:241-8. [PMID: 25271670 DOI: 10.3109/10641963.2014.954710] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We investigated the effect of adipose-derived stem cells (ADSCs) transplantation effects on structural remodeling and pulmonary artery pressure in monocrotaline (MCT)-induced pulmonary hypertensive rats. In the first experiment, 32 male Sprague-Dawley (SD) rats were randomly divided into four groups (n = 8/group): 3 ADSCs treated groups and normal control (Ctrl). ADSCs were administered through the left jugular vein at 10(5), 10(6) and 10(7) cells, respectively, and a cell density of 10(6)cells/ml was shown to be optimal. The GFP-tagged ADSCs were identified in the lungs and differentiated into endothelial-like cells. In the second experiment, 96 male SD rats were randomly divided into three groups (n = 32/group): Ctrl, MCT-induced pulmonary arterial hypertension (PAH), and PAH treated with ADSCs (ADSCs). Two weeks post-MCT administration, the ADSCs group received 1 × 10(6) ADSCs via the external jugular vein. Compared to PAH rats, mean pulmonary arterial pressure was decreased in rats at 1, 2, and 3 weeks after ADSCs-treatment (18.63 ± 2.15 mmHg versus 24.53 ± 2.90 mmHg; 23.07 ± 2.84 mmHg versus 33.18 ± 2.30 mmHg; 22.98 ± 2.34 mmHg versus 36.38 ± 3.28 mmHg, p < 0.05). Meanwhile, the right heart hypertrophy index (36.2 1 ± 4.27% versus 41.01 ± 1.29%; 39.47 ± 4.02% versus 48.75 ± 2 .13%; 41.02 ± 0.9% versus 50.52 ± 1.49%, p < 0.05, respectively), ratio of wall/lumen thickness, as well as the wall/lumen area were significantly reduced in PAH rats at these time points following ADSCs-treatment, as compared with untreated PAH rats. In summary, ADSCs may colonize the pulmonary arteries, attenuate pulmonary arterial hypertension and ameliorate pulmonary arterial remodeling.
Collapse
Affiliation(s)
- Li Luo
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University , Fuzhou , P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Morgan JT, Wood JA, Walker NJ, Raghunathan VK, Borjesson DL, Murphy CJ, Russell P. Human trabecular meshwork cells exhibit several characteristics of, but are distinct from, adipose-derived mesenchymal stem cells. J Ocul Pharmacol Ther 2014; 30:254-66. [PMID: 24456002 DOI: 10.1089/jop.2013.0175] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PURPOSE To support the growing promise of regenerative medicine in glaucoma, we characterized the similarities and differences between human trabecular meshwork (HTM) cells and human mesenchymal stem cells (hMSCs). METHODS HTM cells and hMSCs were phenotypically characterized by flow cytometry. Using quantitative polymerase chain reaction, the expression of myoc, angptl7, sox2, pou5f1, and notch1 was determined in both cell types with and without dexamethasone (Dex). Immunosuppressive behavior of HTM cells and hMSCs was determined using T cells activated with phytohemagglutinin. T-cell proliferation was determined using BrdU incorporation and flow cytometry. Multipotency of HTM cells and hMSCs was determined using adipogenic and osteogenic differentiation media as well as aqueous humor (AH). Alpha-smooth muscle actin (αSMA) expression was determined in HTM cells, hMSCs, and HTM tissue. RESULTS Phenotypically, HTM and hMSCs expressed CD73, CD90, CD105, and CD146 but not CD31, CD34, and CD45 and similar sox2, pou5f1, and notch1 expression. Both cell types suppressed T-cell proliferation. However, HTM cells, but not hMSCs, upregulated myoc and angptl7 in response to Dex. Additionally, HTM cells did not differentiate into adipocytes or osteocytes. Culture of hMSCs in 20%, but not 100%, AH potently induced alkaline phosphatase activity. HTM cells in culture possessed uniformly strong expression of αSMA, which contrasted with the limited expression in hMSCs and spatially discrete expression in HTM tissue. CONCLUSIONS HTM cells possess a number of important similarities with hMSCs but lack multipotency, one of the defining characteristics of stem cells. Further work is needed to explore the molecular mechanisms and functional implications underlying the phenotypic similarities.
Collapse
Affiliation(s)
- Joshua T Morgan
- 1 Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California , Davis, California
| | | | | | | | | | | | | |
Collapse
|