1
|
Smolinská V, Boháč M, Danišovič Ľ. Current status of the applications of conditioned media derived from mesenchymal stem cells for regenerative medicine. Physiol Res 2023; 72:S233-S245. [PMID: 37888967 PMCID: PMC10669946 DOI: 10.33549/physiolres.935186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 12/01/2023] Open
Abstract
Recently published studies suggest that the paracrine substances released by mesenchymal stem cells (MSCs) are the primary motive behind the therapeutic action reported in these cells. Pre-clinical and clinical research on MSCs has produced promising outcomes. Furthermore, these cells are generally safe for therapeutic use and may be extracted from a variety of anatomical regions. Recent research has indicated, however, that transplanted cells do not live long and that the advantages of MSC treatment may be attributable to the large diversity of bioactive substances they create, which play a crucial role in the control of essential physiological processes. Secretome derivatives, such as conditioned media or exosomes, may provide significant benefits over cells in terms of manufacture, preservation, handling, longevity of the product, and potential as a ready-to-use biologic product. Despite their immunophenotypic similarities, the secretome of MSCs appears to vary greatly depending on the host's age and the niches in which the cells live. The secretome's effect on multiple biological processes such as angiogenesis, neurogenesis, tissue repair, immunomodulation, wound healing, anti-fibrotic, and anti-tumor for tissue maintenance and regeneration has been discovered. Defining the secretome of cultured cultivated MSC populations by conditioned media analysis will allow us to assess its potential as a novel treatment approach. This review will concentrate on accumulating data from pre-clinical and clinical trials pointing to the therapeutic value of the conditioned medium. At last, the necessity of characterizing the conditioned medium for determining its potential for cell-free treatment therapy will be emphasized in this study.
Collapse
|
2
|
Zhong Y, Zhang Y, Yu A, Zhang Z, Deng Z, Xiong K, Wang Q, Zhang J. Therapeutic role of exosomes and conditioned medium in keloid and hypertrophic scar and possible mechanisms. Front Physiol 2023; 14:1247734. [PMID: 37781228 PMCID: PMC10536244 DOI: 10.3389/fphys.2023.1247734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
Exosomes, ranging from 40 to 160 nm in diameter, are extracellular lipid bilayer microvesicles that regulate the body's physiological and pathological processes and are secreted by cells that contain proteins, nucleic acids, amino acids and other metabolites. Previous studies suggested that mesenchymal stem cell (MSC)-derived exosomes could either suppress or support keloid and hypertrophic scar progression. Although previous research has identified the potential value of MSC-exosomes in keloid and hypertrophic scar, a comprehensive analysis of different sources of MSC-exosome in keloid and hypertrophic scar is still lacking. This review mainly discusses different insights regarding the roles of MSC-exosomes in keloid and hypertrophic scar treatment and summarizes possible underlying mechanisms.
Collapse
Affiliation(s)
- Yixiu Zhong
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Youfan Zhang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aijiao Yu
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwen Zhang
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenjun Deng
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Kaifen Xiong
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Sun J, Wang L, Matthews RC, Walcott GP, Yu-An L, Wei Y, Zhou Y, Zangi L, Zhang J. CCND2 Modified mRNA Activates Cell Cycle of Cardiomyocytes in Hearts With Myocardial Infarction in Mice and Pigs. Circ Res 2023; 133:484-504. [PMID: 37565345 PMCID: PMC10529295 DOI: 10.1161/circresaha.123.322929] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Experiments in mammalian models of cardiac injury suggest that the cardiomyocyte-specific overexpression of CCND2 (cyclin D2, in humans) improves recovery from myocardial infarction (MI). The primary objective of this investigation was to demonstrate that our specific modified mRNA translation system (SMRTs) can induce CCND2 expression in cardiomyocytes and replicate the benefits observed in other studies of cardiomyocyte-specific CCND2 overexpression for myocardial repair. METHODS The CCND2-cardiomyocyte-specific modified mRNA translation system (cardiomyocyte SMRTs) consists of 2 modRNA constructs: one codes for CCND2 and contains a binding site for L7Ae, and the other codes for L7Ae and contains recognition elements for the cardiomyocyte-specific microRNAs miR-1 and miR-208. Thus, L7Ae suppresses CCND2 translation in noncardiomyocytes but is itself suppressed by endogenous miR-1 and -208 in cardiomyocytes, thereby facilitating cardiomyocyte-specific CCND2 expression. Experiments were conducted in both mouse and pig models of MI, and control assessments were performed in animals treated with an SMRTs coding for the cardiomyocyte-specific expression of luciferase or green fluorescent protein (GFP), in animals treated with L7Ae modRNA alone or with the delivery vehicle, and in Sham-operated animals. RESULTS CCND2 was abundantly expressed in cultured, postmitotic cardiomyocytes 2 days after transfection with the CCND2-cardiomyocyte SMRTs, and the increase was accompanied by the upregulation of markers for cell-cycle activation and proliferation (eg, Ki67 and Aurora B kinase). When the GFP-cardiomyocyte SMRTs were intramyocardially injected into infarcted mouse hearts, the GFP signal was observed in cardiomyocytes but no other cell type. In both MI models, cardiomyocyte proliferation (on day 7 and day 3 after treatment administration in mice and pigs, respectively) was significantly greater, left-ventricular ejection fractions (days 7 and 28 in mice, days 10 and 28 in pigs) were significantly higher, and infarcts (day 28 in both species) were significantly smaller in animals treated with the CCND2-cardiomyocyte SMRTs than in any other group that underwent MI induction. CONCLUSIONS Intramyocardial injections of the CCND2-cardiomyocyte SMRTs promoted cardiomyocyte proliferation, reduced infarct size, and improved cardiac performance in small and large mammalian hearts with MI.
Collapse
Affiliation(s)
- Jiacheng Sun
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
- These authors contributed equally to this work
| | - Lu Wang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
- These authors contributed equally to this work
| | - Rachel C. Matthews
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Gregory P. Walcott
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
- Department of Medicine, Division of Cardiovascular Disease, School of Medicine, University of Alabama at Birmingham
| | - Lu Yu-An
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Yuhua Wei
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Yang Zhou
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham
- Department of Medicine, Division of Cardiovascular Disease, School of Medicine, University of Alabama at Birmingham
| |
Collapse
|
4
|
Payushina OV, Tsomartova DA, Chereshneva YV, Ivanova MY, Lomanovskaya TA, Pavlova MS, Kuznetsov SL. Experimental Transplantation of Mesenchymal Stromal Cells as an Approach to Studying Their Differentiation In Vivo (Review). BIOL BULL+ 2022. [DOI: 10.1134/s1062359022060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Notoginsenoside R1 Promotes Migration, Adhesin, Spreading, and Osteogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stromal Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113403. [PMID: 35684342 PMCID: PMC9182421 DOI: 10.3390/molecules27113403] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/18/2022]
Abstract
Cellular activities, such as attachment, spreading, proliferation, migration, and differentiation are indispensable for the success of bone tissue engineering. Mesenchymal stromal cells (MSCs) are the key precursor cells to regenerate bone. Bioactive compounds from natural products had shown bone regenerative potential. Notoginsenoside R1 (NGR1) is a primary bioactive natural compound that regulates various biological activities, including cardiovascular protection, neuro-protection, and anti-cancer effects. However, the effect of NGR1 on migration, adhesion, spreading, and osteogenic differentiation of MSCs required for bone tissue engineering application has not been tested properly. In this study, we aimed to analyze the effect of NGR1 on the cellular activities of MSCs. Since human adipose-derived stromal cells (hASCs) are commonly used MSCs for bone tissue engineering, we used hASCs as a model of MSCs. The optimal concentration of 0.05 μg/mL NGR1 was biocompatible and promoted migration and osteogenic differentiation of hASCs. Pro-angiogenic factor VEGF expression was upregulated in NGR1-treated hASCs. NGR1 enhanced the adhesion and spreading of hASCs on the bio-inert glass surface. NGR1 robustly promoted hASCs adhesion and survival in 3D-printed TCP scaffold both in vitro and in vivo. NGR1 mitigated LPS-induced expression of inflammatory markers IL-1β, IL-6, and TNF-α in hASCs as well as inhibited the RANKL/OPG expression ratio. In conclusion, the biocompatible NGR1 promoted the migration, adhesion, spreading, osteogenic differentiation, and anti-inflammatory properties of hASCs.
Collapse
|
6
|
Lin H, Chen H, Zhao X, Chen Z, Zhang P, Tian Y, Wang Y, Ding T, Wang L, Shen Y. Advances in mesenchymal stem cell conditioned medium-mediated periodontal tissue regeneration. J Transl Med 2021; 19:456. [PMID: 34736500 PMCID: PMC8567704 DOI: 10.1186/s12967-021-03125-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/22/2021] [Indexed: 12/19/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease that leads to the destruction of both soft and hard periodontal tissues. Complete periodontal regeneration in clinics using the currently available treatment approaches is still a challenge. Mesenchymal stem cells (MSCs) have shown promising potential to regenerate periodontal tissue in various preclinical and clinical studies. The poor survival rate of MSCs during in vivo transplantation and host immunogenic reaction towards MSCs are the main drawbacks of direct use of MSCs in periodontal tissue regeneration. Autologous MSCs have limited sources and possess patient morbidity during harvesting. Direct use of allogenic MSCs could induce host immune reaction. Therefore, the MSC-based indirect treatment approach could be beneficial for periodontal regeneration in clinics. MSC culture conditioned medium (CM) contains secretomes that had shown immunomodulatory and tissue regenerative potential in pre-clinical and clinical studies. MSC-CM contains a cocktail of growth factors, cytokines, chemokines, enzymes, and exosomes, extracellular vesicles, etc. MSC-CM-based indirect treatment has the potential to eliminate the drawbacks of direct use of MSCs for periodontal tissue regeneration. MSC-CM holds the tremendous potential of bench-to-bed translation in periodontal regeneration applications. This review focuses on the accumulating evidence indicating the therapeutic potential of the MSC-CM in periodontal regeneration-related pre-clinical and clinical studies. Recent advances on MSC-CM-based periodontal regeneration, existing challenges, and prospects are well summarized as guidance to improve the effectiveness of MSC-CM on periodontal regeneration in clinics.
Collapse
Affiliation(s)
- Hongbing Lin
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Huishan Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Xuetao Zhao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Zhen Chen
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Peipei Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Yue Tian
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Yawei Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Tong Ding
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Lijing Wang
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China.,Vascular Biology Research Institute, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuqin Shen
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
7
|
Jiang LL, Li H, Liu L. Xenogeneic stem cell transplantation: Research progress and clinical prospects. World J Clin Cases 2021; 9:3826-3837. [PMID: 34141739 PMCID: PMC8180210 DOI: 10.12998/wjcc.v9.i16.3826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Organ transplantation is the ultimate treatment for end-stage diseases such as heart and liver failure. However, the severe shortage of donor organs has limited the organ transplantation progress. Xenogeneic stem cell transplantation provides a new strategy to solve this problem. Researchers have shown that xenogeneic stem cell transplantation has significant therapeutic effects and broad application prospects in treating liver failure, myocardial infarction, advanced type 1 diabetes mellitus, myelosuppression, and other end-stage diseases by replacing the dysfunctional cells directly or improving the endogenous regenerative milieu. In this review, the sources, problems and solutions, and potential clinical applications of xenogeneic stem cell transplantation will be discussed.
Collapse
Affiliation(s)
- Lin-Li Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hui Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lei Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
8
|
Li M, Yang J, Cheng O, Peng Z, Luo Y, Ran D, Yang Y, Xiang P, Huang H, Tan X, Wang H. Effect of TO901317 on GF to promote the differentiation of human bone marrow mesenchymal stem cells into dopamine neurons on Parkinson's disease. Ther Adv Chronic Dis 2021; 12:2040622321998139. [PMID: 33796244 PMCID: PMC7985948 DOI: 10.1177/2040622321998139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 02/03/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Human bone marrow mesenchymal stem cells (hBMSCs) could differentiate into dopamine-producing cells and ameliorate behavioral deficits in Parkinson’s disease (PD) models. Liver X receptors (LXRs) are involved in the maintenance of the normal function of central nervous system myelin. Therefore, the previous work of our team has found the induction of cocktail-induced to dopaminergic (DA) phenotypes from adult rat BMSCs by using sonic hedgehog (SHH), fibroblast growth factor 8 (FGF8), basic fibroblast growth factor (bFGF), and TO901317 (an agonist of LXRs) with 87.42% of efficiency in a 6-day induction period. But we did not verify whether the induced cells had the corresponding neural function. Methods: Expressions of LXRα, LXRβ, and tyrosine hydroxylase (TH) were detected by immunofluorescence and western blot. Adenosine triphosphate-binding cassette transporter A1 (ABCA1) was detected by quantitative real-time PCR. The induced cells were transplanted into PD rats to study whether the induced cells are working. Results: The induced cells can release the dopamine transmitter; the maximum induction efficiency of differentiation of hBMSCs into DA neurons was 91.67% under conditions of combined use with TO901317 and growth factors (GF). When the induced-cells were transplanted into PD rats, the expression of TH in the striatum increased significantly, and the behavior of PD rats induced by apomorphine was significantly improved. Conclusion: The induced cells have the function of DA neurons and have the potential to treat PD. TO901317 promoted differentiation of hBMSCs into DA neurons, which may be related to activation of the LXR-ABCA1 signaling pathway.
Collapse
Affiliation(s)
- Miaomiao Li
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Junqing Yang
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Oumei Cheng
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Zhe Peng
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Yin Luo
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Dongzhi Ran
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Yang Yang
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Pu Xiang
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Haifeng Huang
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Xiaodan Tan
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Hong Wang
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China
| |
Collapse
|
9
|
Galow AM, Goldammer T, Hoeflich A. Xenogeneic and Stem Cell-Based Therapy for Cardiovascular Diseases: Genetic Engineering of Porcine Cells and Their Applications in Heart Regeneration. Int J Mol Sci 2020; 21:ijms21249686. [PMID: 33353186 PMCID: PMC7766969 DOI: 10.3390/ijms21249686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases represent a major health concern worldwide with few therapy options for ischemic injuries due to the limited regeneration potential of affected cardiomyocytes. Innovative cell replacement approaches could facilitate efficient regenerative therapy. However, despite extensive attempts to expand primary human cells in vitro, present technological limitations and the lack of human donors have so far prevented their broad clinical use. Cell xenotransplantation might provide an ethically acceptable unlimited source for cell replacement therapies and bridge the gap between waiting recipients and available donors. Pigs are considered the most suitable candidates as a source for xenogeneic cells and tissues due to their anatomical and physiological similarities with humans. The potential of porcine cells in the field of stem cell-based therapy and regenerative medicine is under intensive investigation. This review outlines the current progress and highlights the most promising approaches in xenogeneic cell therapy with a focus on the cardiovascular system.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Correspondence: ; Tel.: +49-38208-68-723
| | - Tom Goldammer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Molecular Biology and Fish Genetics Unit, Faculty of Agriculture and Environmental Sciences, University of Rostock, 18059 Rostock, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
| |
Collapse
|
10
|
Nanosheets-incorporated bio-composites containing natural and synthetic polymers/ceramics for bone tissue engineering. Int J Biol Macromol 2020; 164:1960-1972. [DOI: 10.1016/j.ijbiomac.2020.08.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/20/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
|
11
|
Fan C, Oduk Y, Zhao M, Lou X, Tang Y, Pretorius D, Valarmathi MT, Walcott GP, Yang J, Menasche P, Krishnamurthy P, Zhu W, Zhang J. Myocardial protection by nanomaterials formulated with CHIR99021 and FGF1. JCI Insight 2020; 5:e132796. [PMID: 32453715 PMCID: PMC7406256 DOI: 10.1172/jci.insight.132796] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
The mortality of patients suffering from acute myocardial infarction is linearly related to the infarct size. As regeneration of cardiomyocytes from cardiac progenitor cells is minimal in the mammalian adult heart, we have explored a new therapeutic approach, which leverages the capacity of nanomaterials to release chemicals over time to promote myocardial protection and infarct size reduction. Initial screening identified 2 chemicals, FGF1 and CHIR99021 (a Wnt1 agonist/GSK-3β antagonist), which synergistically enhance cardiomyocyte cell cycle in vitro. Poly-lactic-co-glycolic acid nanoparticles (NPs) formulated with CHIR99021 and FGF1 (CHIR + FGF1-NPs) provided an effective slow-release system for up to 4 weeks. Intramyocardial injection of CHIR + FGF1-NPs enabled myocardial protection via reducing infarct size by 20%-30% in mouse or pig models of postinfarction left ventricular (LV) remodeling. This LV structural improvement was accompanied by preservation of cardiac contractile function. Further investigation revealed that CHIR + FGF1-NPs resulted in a reduction of cardiomyocyte apoptosis and increase of angiogenesis. Thus, using a combination of chemicals and an NP-based prolonged-release system that works synergistically, this study demonstrates a potentially novel therapy for LV infarct size reduction in hearts with acute myocardial infarction.
Collapse
Affiliation(s)
- Chengming Fan
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cardiovascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Yasin Oduk
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Meng Zhao
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xi Lou
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yawen Tang
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Danielle Pretorius
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mani T. Valarmathi
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gregory P. Walcott
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jinfu Yang
- Department of Cardiovascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Philippe Menasche
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cardiovascular Surgery, Université de Paris, PARCC, INSERM, F-75015 Paris, France
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wuqiang Zhu
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, and School of Engineering, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
12
|
Browne S, Healy KE. Matrix-assisted cell transplantation for tissue vascularization. Adv Drug Deliv Rev 2019; 146:155-169. [PMID: 30605738 DOI: 10.1016/j.addr.2018.12.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/30/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022]
Abstract
Cell therapy offers much promise for the treatment of ischemic diseases by augmenting tissue vasculogenesis. Matrix-assisted cell transplantation (MACT) has been proposed as a solution to enhance cell survival and integration with host tissue following transplantation. By designing semi synthetic matrices (sECM) with the correct physical and biochemical signals, encapsulated cells are directed towards a more angiogenic phenotype. In this review, we describe the choice of cells suitable for pro-angiogenic therapies, the properties that should be considered when designing sECM for transplantation and their relative importance. Pre-clinical models where MACT has been successfully applied to promote angiogenesis are reviewed to show the great potential of this strategy to treat ischemic conditions.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA; Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
13
|
Leong YY, Ng WH, Umar Fuaad MZ, Ng CT, Ramasamy R, Lim V, Yong YK, Tan JJ. Mesenchymal stem cells facilitate cardiac differentiation in Sox2-expressing cardiac C-kit cells in coculture. J Cell Biochem 2018; 120:9104-9116. [PMID: 30548289 DOI: 10.1002/jcb.28186] [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] [Received: 08/24/2018] [Accepted: 11/08/2018] [Indexed: 01/11/2023]
Abstract
Stem cell therapy offers hope to reconstitute injured myocardium and salvage heart from failing. A recent approach using combinations of derived Cardiac-derived c-kit expressing cells (CCs) and mesenchymal stem cells (MSCs) in transplantation improved infarcted hearts with a greater functional outcome, but the effects of MSCs on CCs remain to be elucidated. We used a novel two-step protocol to clonogenically amplify colony forming c-kit expressing cells from 4- to 6-week-old C57BL/6N mice. This method yielded highly proliferative and clonogenic CCs with an average population doubling time of 17.2 ± 0.2, of which 80% were at the G1 phase. We identified two distinctly different CC populations based on its Sox2 expression, which was found to inversely related to their nkx2.5 and gata4 expression. To study CCs after MSC coculture, we developed micron-sized particles of iron oxide-based magnetic reisolation method to separate CCs from MSCs for subsequent analysis. Through validation using the sex and species mismatch CC-MSC coculture method, we confirmed that the purity of the reisolated cells was greater than 85%. In coculture experiment, we found that MSCs prominently enhanced Ctni and Mef2c expressions in Sox2 pos CCs after the induction of cardiac differentiation, and the level was higher than that of conditioned medium Sox2 pos CCs. However, these effects were not found in Sox2 neg CCs. Immunofluorescence labeling confirmed the presence of cardiac-like cells within Sox2 pos CCs after differentiation, identified by its cardiac troponin I and α-sarcomeric actinin expressions. In conclusion, this study shows that MSCs enhance CC differentiation toward cardiac myocytes. This enhancement is dependent on CC stemness state, which is determined by Sox2 expression.
Collapse
Affiliation(s)
- Yin Yee Leong
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Wai Hoe Ng
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Mimi Zulaikha Umar Fuaad
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Chin Theng Ng
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.,Department of Preclinical, Physiology Unit, Faculty of Medicine, AIMST University, Bedong, Kedah, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Vuanghao Lim
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Jun Jie Tan
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| |
Collapse
|
14
|
Haider KH. Bone marrow cell therapy and cardiac reparability: better cell characterization will enhance clinical success. Regen Med 2018; 13:457-475. [PMID: 29985118 DOI: 10.2217/rme-2017-0134] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nearly two decades of experimental and clinical research with bone marrow cells have paved the way for Phase III pivotal trials in larger groups of heart patients. Despite immense advancements, a multitude of factors are hampering the acceptance of bone marrow cell-based therapy for routine clinical use. These include uncertainties regarding purification and characterization of the cell preparation, delivery protocols, mechanistic understanding and study end points and their methods of assessment. Clinical data show mediocre outcomes in terms of sustained cardiac pump function. This review reasons that the modest outcomes observed in trials thus far are based on quality of the cell preparation with a focus on the chronological aging of cells when autologous cells are used for transplantation in elderly patients.
Collapse
Affiliation(s)
- Khawaja H Haider
- Department of Basic Sciences, Sulaiman AlRajhi Medical School, Al Qassim, Al Bukayria, 51941, Kingdom of Saudi Arabia
| |
Collapse
|
15
|
Xia W, Zhuang L, Hou M. Role of lincRNA‑p21 in the protective effect of macrophage inhibition factor against hypoxia/serum deprivation‑induced apoptosis in mesenchymal stem cells. Int J Mol Med 2018; 42:2175-2184. [PMID: 30015822 DOI: 10.3892/ijmm.2018.3767] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/28/2018] [Indexed: 01/09/2023] Open
Abstract
Stem cell transplantation is a promising clinical strategy for curing ischemic cardiomyopathy. However, its efficacy is impaired by low cell survival following transplantation, partly caused by insufficient resistance of the transplanted stem cells to severe oxidative stress at the injury site. In the current study, it was demonstrated that the small‑molecule macrophage migration inhibitory factor (MIF) enhanced the defense of bone marrow‑derived mesenchymal stem cells (MSCs) against hypoxia/serum deprivation (SD)‑induced apoptosis in vitro. MIF significantly suppressed apoptosis and caspase family activities through inhibition of long intergenic noncoding (linc) RNA‑p21 to maintain activation of the Wnt/β‑catenin signaling pathway. The regulatory loop between MIF and the lincRNA‑p21‑Wnt/β‑catenin signaling pathway was identified to be associated with the inhibition of oxidative stress. The involvement of the lincRNA‑p21‑Wnt/β‑catenin signaling pathway in the effects of MIF in MSCs by overexpression of lincRNA‑p21and silencing β‑catenin using small interfering RNA was also demonstrated, both of which abolished the anti‑apoptotic and anti‑oxidative effects of MIF in MSCs under hypoxia/SD conditions. In conclusion, MIF protected MSCs from hypoxia/SD‑induced apoptosis by interacting with lincRNA‑p21, leading to activation of the downstream Wnt/β‑catenin signaling pathway and decreased oxidative stress. Thus, treatment with MIF may have important therapeutic implications in improving MSC survival and therapeutic efficiency.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lei Zhuang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
16
|
Zhu W, Zhao M, Mattapally S, Chen S, Zhang J. CCND2 Overexpression Enhances the Regenerative Potency of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Remuscularization of Injured Ventricle. Circ Res 2017; 122:88-96. [PMID: 29018036 DOI: 10.1161/circresaha.117.311504] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE The effectiveness of transplanted, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for treatment of ischemic myocardial injury is limited by the exceptionally low engraftment rate. OBJECTIVE To determine whether overexpression of the cell cycle activator CCND2 (cyclin D2) in hiPSC-CMs can increase the graft size and improve myocardial recovery in a mouse model of myocardial infarction by increasing the proliferation of grafted cells. METHODS AND RESULTS Human CCND2 was delivered to hiPSCs via lentiviral-mediated gene transfection. In cultured cells, markers for cell cycle activation and proliferation were ≈3- to 7-folds higher in CCND2-overexpressing hiPSC-CMs (hiPSC-CCND2OECMs) than in hiPSC-CMs with normal levels of CCND2 (hiPSC-CCND2WTCMs; P<0.01). The pluripotent genes (Oct 4, Sox2, and Nanog) decrease to minimal levels and undetectable levels at day 1 and 10 after differentiating to CMs. In the mouse myocardial infarction model, cardiac function, infarct size, and the number of engrafted cells were similar at week 1 after treatment with hiPSC-CCND2OECMs or hiPSC-CCND2WTCMs but was about tripled in hiPSC-CCND2OECM-treated than in hiPSC-CCND2WTCM-treated animals at week 4 (P<0.01). The cardiac function and infarct size were significantly better in both cell treatment groups' hearts than in control hearts, which was most prominent in hiPSC-CCND2OECM-treated animals (P<0.05, each). No tumor formation was observed in any hearts. CONCLUSIONS CCND2 overexpression activates cell cycle progression in hiPSC-CMs that results in a significant enhanced potency for myocardial repair as evidenced by remuscularization of injured myocardium. This left ventricular muscle regeneration and increased angiogenesis in border zone are accompanied by a significant improvement of left ventricular chamber function.
Collapse
Affiliation(s)
- Wuqiang Zhu
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (W.Z., M.Z., S.M., J.Z.); and Department of Physiology and Pathophysiology, Fudan University, Shanghai, China (M.Z., S.C.)
| | - Meng Zhao
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (W.Z., M.Z., S.M., J.Z.); and Department of Physiology and Pathophysiology, Fudan University, Shanghai, China (M.Z., S.C.)
| | - Saidulu Mattapally
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (W.Z., M.Z., S.M., J.Z.); and Department of Physiology and Pathophysiology, Fudan University, Shanghai, China (M.Z., S.C.)
| | - Sifeng Chen
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (W.Z., M.Z., S.M., J.Z.); and Department of Physiology and Pathophysiology, Fudan University, Shanghai, China (M.Z., S.C.)
| | - Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (W.Z., M.Z., S.M., J.Z.); and Department of Physiology and Pathophysiology, Fudan University, Shanghai, China (M.Z., S.C.).
| |
Collapse
|
17
|
Quantitative Assessment of Optimal Bone Marrow Site for the Isolation of Porcine Mesenchymal Stem Cells. Stem Cells Int 2017; 2017:1836960. [PMID: 28539939 PMCID: PMC5429955 DOI: 10.1155/2017/1836960] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/28/2017] [Indexed: 12/13/2022] Open
Abstract
Background. One of the most plentiful sources for MSCs is the bone marrow; however, it is unknown whether MSC yield differs among different bone marrow sites. In this study, we quantified cellular yield and evaluated resident MSC population from five bone marrow sites in the porcine model. In addition, we assessed the feasibility of a commercially available platelet concentrator (Magellan® MAR01™ Arteriocyte Medical Systems, Hopkinton, MA) as a bedside stem cell concentration device. Methods. Analyses of bone marrow aspirate (BMA) and concentrated bone marrow aspirate (cBMA) included bone marrow volume, platelet and nucleated cell yield, colony-forming unit fibroblast (CFU-F) number, flow cytometry, and assessment of differentiation potential. Results. Following processing, the concentration of platelets and nucleated cells significantly increased but was not significantly different between sites. The iliac crest had significantly less bone marrow volume; however, it yielded significantly more CFUs compared to the other bone marrow sites. Culture-expanded cells from all tested sites expressed high levels of MSC surface markers and demonstrated adipogenic and osteogenic differentiation potential. Conclusions. All anatomical bone marrow sites contained MSCs, but the iliac crest was the most abundant source of MSCs. Additionally, the Magellan can function effectively as a bedside stem cell concentrator.
Collapse
|
18
|
Gao L, Kupfer ME, Jung JP, Yang L, Zhang P, Da Sie Y, Tran Q, Ajeti V, Freeman BT, Fast VG, Campagnola PJ, Ogle BM, Zhang J. Myocardial Tissue Engineering With Cells Derived From Human-Induced Pluripotent Stem Cells and a Native-Like, High-Resolution, 3-Dimensionally Printed Scaffold. Circ Res 2017; 120:1318-1325. [PMID: 28069694 DOI: 10.1161/circresaha.116.310277] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 12/30/2016] [Accepted: 01/09/2017] [Indexed: 01/31/2023]
Abstract
RATIONALE Conventional 3-dimensional (3D) printing techniques cannot produce structures of the size at which individual cells interact. OBJECTIVE Here, we used multiphoton-excited 3D printing to generate a native-like extracellular matrix scaffold with submicron resolution and then seeded the scaffold with cardiomyocytes, smooth muscle cells, and endothelial cells that had been differentiated from human-induced pluripotent stem cells to generate a human-induced pluripotent stem cell-derived cardiac muscle patch (hCMP), which was subsequently evaluated in a murine model of myocardial infarction. METHODS AND RESULTS The scaffold was seeded with ≈50 000 human-induced pluripotent stem cell-derived cardiomyocytes, smooth muscle cells, and endothelial cells (in a 2:1:1 ratio) to generate the hCMP, which began generating calcium transients and beating synchronously within 1 day of seeding; the speeds of contraction and relaxation and the peak amplitudes of the calcium transients increased significantly over the next 7 days. When tested in mice with surgically induced myocardial infarction, measurements of cardiac function, infarct size, apoptosis, both vascular and arteriole density, and cell proliferation at week 4 after treatment were significantly better in animals treated with the hCMPs than in animals treated with cell-free scaffolds, and the rate of cell engraftment in hCMP-treated animals was 24.5% at week 1 and 11.2% at week 4. CONCLUSIONS Thus, the novel multiphoton-excited 3D printing technique produces extracellular matrix-based scaffolds with exceptional resolution and fidelity, and hCMPs fabricated with these scaffolds may significantly improve recovery from ischemic myocardial injury.
Collapse
Affiliation(s)
- Ling Gao
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Molly E Kupfer
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Jangwook P Jung
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Libang Yang
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Patrick Zhang
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Yong Da Sie
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Quyen Tran
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Visar Ajeti
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Brian T Freeman
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Vladimir G Fast
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Paul J Campagnola
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.)
| | - Brenda M Ogle
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.).
| | - Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.G., V.G.F., J.Z.); Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis (M.E.K., J.P.J., L.Y., P.Z., B.T.F., B.M.O.); and Department of Biomedical Engineering, University of Wisconsin, Madison (Y.D.S., Q.T., V.A., P.J.C.).
| |
Collapse
|
19
|
Bharti D, Shivakumar SB, Subbarao RB, Rho GJ. Research Advancements in Porcine Derived Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2016. [PMID: 26201864 PMCID: PMC5403966 DOI: 10.2174/1574888x10666150723145911] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the present era of stem cell biology, various animals such as Mouse, Bovine, Rabbit and Porcine have been tested for the efficiency of their mesenchymal stem cells (MSCs) before their actual use for stem cell based application in humans. Among them pigs have many similarities to humans in the form of organ size, physiology and their functioning, therefore they have been considered as a valuable model system for in vitro studies and preclinical assessments. Easy assessability, few ethical issues, successful MSC isolation from different origins like bone marrow, skin, umbilical cord blood, Wharton’s jelly, endometrium, amniotic fluid and peripheral blood make porcine a good model for stem cell therapy. Porcine derived MSCs (pMSCs) have shown greater in vitro differentiation and transdifferention potential towards mesenchymal lineages and specialized lineages such as cardiomyocytes, neurons, hepatocytes and pancreatic beta cells. Immunomodulatory and low immunogenic profiles as shown by autologous and heterologous MSCs proves them safe and appropriate models for xenotransplantation purposes. Furthermore, tissue engineered stem cell constructs can be of immense importance in relation to various osteochondral defects which are difficult to treat otherwise. Using pMSCs successful treatment of various disorders like Parkinson’s disease, cardiac ischemia, hepatic failure, has been reported by many studies. Here, in this review we highlight current research findings in the area of porcine mesenchymal stem cells dealing with their isolation methods, differentiation ability, transplantation applications and their therapeutic potential towards various diseases.
Collapse
Affiliation(s)
| | | | | | - Gyu-Jin Rho
- OBS/Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, 900 Gazwa, Jinju 660-701, Republic of Korea.
| |
Collapse
|
20
|
Cai M, Shen R, Song L, Lu M, Wang J, Zhao S, Tang Y, Meng X, Li Z, He ZX. Bone Marrow Mesenchymal Stem Cells (BM-MSCs) Improve Heart Function in Swine Myocardial Infarction Model through Paracrine Effects. Sci Rep 2016; 6:28250. [PMID: 27321050 PMCID: PMC4913323 DOI: 10.1038/srep28250] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/01/2016] [Indexed: 12/14/2022] Open
Abstract
Stem cells are promising for the treatment of myocardial infarction (MI) and large animal models should be used to better understand the full spectrum of stem cell actions and preclinical evidences. In this study, bone marrow mesenchymal stem cells (BM-MSCs) were transplanted into swine heart ischemia model. To detect glucose metabolism in global left ventricular myocardium and regional myocardium, combined with assessment of cardiac function, positron emission tomography-computer tomography (PET-CT) and magnetic resonance imaging (MRI) were performed. To study the changes of glucose transporters and glucose metabolism-related enzymes and the signal transduction pathway, RT-PCR, Western-blot, and immunohistochemistry were carried out. Myocardium metabolic evaluation by PET-CT showed that mean signal intensity (MSI) increased in these segments at week 4 compared with that at week 1 after BM-MSCs transplantation. Moreover, MRI demonstrated significant function enhancement in BM-MSCs group. The gene expressions of glucose transporters (GLUT1, GLUT4), glucose metabolism-related enzymes phosphofructokinase (PFK), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH)) and 70-kDa ribosomal protein S6 kinase (p70s6k) in BM-MSCs injected areas were up-regulated at week 4 after BM-MSCs transplantation and this was confirmed by Western-blot and immunohistochemistry. In conclusions, BM-MSCs transplantation could improve cardiac function in swine MI model by activation of mTOR signal transduction pathway.
Collapse
Affiliation(s)
- Min Cai
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China.,Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Shen
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Song
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Minjie Lu
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguang Wang
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Shihua Zhao
- Department of Nuclear Medicine, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Yue Tang
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Xianmin Meng
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin, China
| | - Zuo-Xiang He
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Zhilai Z, Biling M, Sujun Q, Chao D, Benchao S, Shuai H, Shun Y, Hui Z. Preconditioning in lowered oxygen enhances the therapeutic potential of human umbilical mesenchymal stem cells in a rat model of spinal cord injury. Brain Res 2016; 1642:426-435. [PMID: 27085204 DOI: 10.1016/j.brainres.2016.04.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 12/20/2022]
Abstract
Human umbilical cord mesenchymal stem cells (UCMSCs) have recently been shown to hold great therapeutic potential for the treatment of spinal cord injury (SCI). However, the number of engrafted cells has been shown to decrease dramatically post-transplantation. Physioxia is known to enhance the paracrine properties and immune modulation of stem cells, a notion that has been applied in many clinical settings. We therefore hypothesized that preconditioning of UCMSCs in physioxic environment would enhance the regenerative properties of these cells in the treatment of rat SCI. UCMSCs were pretreated with either atmospheric normoxia (21% O2, N-UCMSC) or physioxia (5% O2, P-UCMSC). The MSCs were characterized using flow cytometry, immunocytochemistry, and real-time polymerase chain reaction. Furthermore, 10(5) N-UCMSC or P-UCMSC were injected into the injured spinal cord immediately after SCI, and locomotor function as well as cellular, molecular and pathological changes were compared between the groups. We found that N-UCMSC and P-UCMSC displayed similar surface protein expression. P-UCMSC grew faster, while physioxia up-regulated the expression of trophic and growth factors, including hepatocyte growth factor (HGF), brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor(VEGF), in UCMSCs. Compared to N-UCMSC, treatment with P-UCMSC was associated with marked changes in the SCI environment, with a significant increase in axonal preservation and a decrease in the number of caspase-3+ cells and ED-1+ macrophages. These changes were accompanied by improved functional recovery. Thus, the present study indicated that preculturing UCMSCs under 5% lowered oxygen physioxic conditions prior to transplantation improves their therapeutic potential for the treatment of SCI in rats.
Collapse
Affiliation(s)
- Zhou Zhilai
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Mo Biling
- Department of Cardiology, Liwan Hospital, Guangzhou Medical University, China.
| | - Qiu Sujun
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Dong Chao
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Shi Benchao
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Huang Shuai
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Yao Shun
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Zhang Hui
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China.
| |
Collapse
|
22
|
Conditioned Medium From Mesenchymal Stem Cells Enhances Early Bone Regeneration After Maxillary Sinus Floor Elevation in Rabbits. IMPLANT DENT 2015; 24:657-63. [DOI: 10.1097/id.0000000000000335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
23
|
Li J, Zhu K, Yang S, Wang Y, Guo C, Yin K, Wang C, Lai H. Fibrin patch-based insulin-like growth factor-1 gene-modified stem cell transplantation repairs ischemic myocardium. Exp Biol Med (Maywood) 2015; 240:585-92. [PMID: 25767192 DOI: 10.1177/1535370214556946] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/08/2014] [Indexed: 02/03/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs), tissue-engineered cardiac patch, and therapeutic gene have all been proposed as promising therapy strategies for cardiac repair after myocardial infarction. In our study, BMSCs were modified with insulin-like growth factor-1 (IGF-1) gene, loaded into a fibrin patch, and then transplanted into a porcine model of ischemia/reperfusion (I/R) myocardium injury. The results demonstrated that IGF-1 gene overexpression could promote proliferation of endothelial cells and cardiomyocyte-like differentiation of BMSCs in vitro. Four weeks after transplantation of fibrin patch loaded with gene-modified BMSCs, IGF-1 overexpression could successfully promote angiogenesis, inhibit remodeling, increase grafted cell survival and reduce apoptosis. In conclusion, the integrated strategy, which combined fibrin patch with IGF-1 gene modified BMSCs, could promote the histological cardiac repair for a clinically relevant porcine model of I/R myocardium injury.
Collapse
Affiliation(s)
- Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| | - Shan Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yulin Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| | - Changfa Guo
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| | - Kanhua Yin
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China Shanghai Institute of Cardiovascular Disease, Shanghai 200032, P.R. China
| |
Collapse
|
24
|
Liu WH, Song FQ, Ren LN, Guo WQ, Wang T, Feng YX, Tang LJ, Li K. The multiple functional roles of mesenchymal stem cells in participating in treating liver diseases. J Cell Mol Med 2015; 19:511-520. [PMID: 25534251 PMCID: PMC4369809 DOI: 10.1111/jcmm.12482] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/07/2014] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a group of stem cells derived from the mesodermal mesenchyme. MSCs can be obtained from a variety of tissues, including bone marrow, umbilical cord tissue, umbilical cord blood, peripheral blood and adipose tissue. Under certain conditions, MSCs can differentiate into many cell types both in vitro and in vivo, including hepatocytes. To date, four main strategies have been developed to induce the transdifferentiation of MSCs into hepatocytes: addition of chemical compounds and cytokines, genetic modification, adjustment of the micro-environment and alteration of the physical parameters used for culturing MSCs. Although the phenomenon of transdifferentiation of MSCs into hepatocytes has been described, the detailed mechanism is far from clear. Generally, the mechanism is a cascade reaction whereby stimulating factors activate cellular signalling pathways, which in turn promote the production of transcription factors, leading to hepatic gene expression. Because MSCs can give rise to hepatocytes, they are promising to be used as a new treatment for liver dysfunction or as a bridge to liver transplantation. Numerous studies have confirmed the therapeutic effects of MSCs on hepatic fibrosis, cirrhosis and other liver diseases, which may be related to the differentiation of MSCs into functional hepatocytes. In addition to transdifferentiation into hepatocytes, when MSCs are used to treat liver disease, they may also inhibit hepatocellular apoptosis and secrete various bioactive molecules to promote liver regeneration. In this review, the capacity and molecular mechanism of MSC transdifferentiation, and the therapeutic effects of MSCs on liver diseases are thoroughly discussed.
Collapse
Affiliation(s)
- Wei-hui Liu
- General Surgery Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Fu-qiang Song
- Experimental Medical Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Li-na Ren
- General Surgery Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Wen-qiong Guo
- Nursing College, Chengdu Medical SchoolChengdu, Sichuan Province, China
| | - Tao Wang
- General Surgery Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Ya-xing Feng
- Experimental Medical Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Li-jun Tang
- General Surgery Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| | - Kun Li
- Experimental Medical Center, Chengdu Military General HospitalChengdu, Sichuan Province, China
| |
Collapse
|
25
|
Kawai T, Katagiri W, Osugi M, Sugimura Y, Hibi H, Ueda M. Secretomes from bone marrow-derived mesenchymal stromal cells enhance periodontal tissue regeneration. Cytotherapy 2015; 17:369-81. [PMID: 25595330 DOI: 10.1016/j.jcyt.2014.11.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/06/2014] [Accepted: 11/25/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Periodontal tissue regeneration with the use of mesenchymal stromal cells (MSCs) has been regarded as a future cell-based therapy. However, low survival rates and the potential tumorigenicity of implanted MSCs could undermine the efficacy of cell-based therapy. The use of conditioned media from MSCs (MSC-CM) may be a feasible approach to overcome these limitations. The aim of this study was to confirm the effect of MSC-CM on periodontal regeneration. METHODS MSC-CM were collected during their cultivation. The concentrations of the growth factors in MSC-CM were measured with the use of enzyme-linked immunoassay. Rat MSCs (rMSCs) and human umbilical vein endothelial cells cultured in MSC-CM were assessed on wound-healing and angiogenesis. The expressions of osteogenetic- and angiogenic-related genes of rMSCs cultured in MSC-CM were quantified by means of real-time reverse transcriptase-polymerase chain reaction analysis. In vivo, periodontal defects were prepared in the rat models and the collagen sponges with MSC-CM were implanted. RESULTS MSC-CM includes insulin-like growth factor-1, vascular endothelial growth factor, transforming growth factor-β1 and hepatocyte growth factor. In vitro, wound-healing and angiogenesis increased significantly in MSC-CM. The levels of expression of osteogenetic- and angiogenic-related genes were significantly upregulated in rMSCs cultured with MSC-CM. In vivo, in the MSC-CM group, 2 weeks after implantation, immunohistochemical analysis showed several CD31-, CD105-or FLK-1-positive cells occurring frequently. At 4 weeks after implantation, regenerated periodontal tissue was observed in MSC-CM groups. CONCLUSIONS The use of MSC-CM may be an alternative therapy for periodontal tissue regeneration because several cytokines included in MSC-CM will contribute to many processes of complicated periodontal tissue regeneration.
Collapse
Affiliation(s)
- Takamasa Kawai
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Wataru Katagiri
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Masashi Osugi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukiko Sugimura
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
26
|
Zhang L, Guo J, Zhang P, Xiong Q, Wu SC, Xia L, Roy SS, Tolar J, O'Connell TD, Kyba M, Liao K, Zhang J. Derivation and high engraftment of patient-specific cardiomyocyte sheet using induced pluripotent stem cells generated from adult cardiac fibroblast. Circ Heart Fail 2014; 8:156-66. [PMID: 25420485 DOI: 10.1161/circheartfailure.114.001317] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) can be differentiated into potentially unlimited lineages of cell types for use in autologous cell therapy. However, the efficiency of the differentiation procedure and subsequent function of the iPSC-derived cells may be influenced by epigenetic factors that the iPSCs retain from their tissues of origin; thus, iPSC-derived cells may be more effective for treatment of myocardial injury if the iPSCs were engineered from cardiac-lineage cells, rather than dermal fibroblasts. METHODS AND RESULTS We show that human cardiac iPSCs (hciPSCs) can be generated from cardiac fibroblasts and subsequently differentiated into exceptionally pure (>92%) sheets of cardiomyocytes (CMs). The hciPSCs passed through all the normal stages of differentiation before assuming a CM identity. When using the fibrin gel-enhanced delivery of hciPSC-CM sheets at the site of injury in infarcted mouse hearts, the engraftment rate was 31.91%±5.75% at Day 28 post transplantation. The hciPSC-CM in the sheet also appeared to develop a more mature, structurally aligned phenotype 28 days after transplantation and was associated with significant improvements in cardiac function, vascularity, and reduction in apoptosis. CONCLUSIONS These data strongly support the potential of hciPSC-CM sheet transplantation for the treatment of heart with acute myocardial infarction.
Collapse
Affiliation(s)
- Liying Zhang
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Jing Guo
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Pengyuan Zhang
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Qiang Xiong
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Steven C Wu
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Lily Xia
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Samit Sunny Roy
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Jakub Tolar
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Timothy D O'Connell
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Michael Kyba
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Kenneth Liao
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.)
| | - Jianyi Zhang
- From the Cardiovascular Division, Department of Medicine (L.Z., J.G., P.Z., Q.X., S.S.R., J.Z.), Department of Integrative Biology and Physiology, School of Medicine (S.C.W., T.D.O.C.), Stem Cell Institute and Department of Pediatrics (L.X., J.T., M.K.), and Department of Surgery (K.L.), University of Minnesota, Minneapolis, MN; and Department of Cardiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China (J.G.).
| |
Collapse
|
27
|
Stramandinoli-Zanicotti RT, Carvalho AL, Rebelatto CLK, Sassi LM, Torres MF, Senegaglia AC, Boldrinileite LM, Correa-Dominguez A, Kuligovsky C, Brofman PRS. Brazilian minipig as a large-animal model for basic research and stem cell-based tissue engineering. Characterization and in vitro differentiation of bone marrow-derived mesenchymal stem cells. J Appl Oral Sci 2014; 22:218-27. [PMID: 25025563 PMCID: PMC4072273 DOI: 10.1590/1678-775720130526] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/20/2014] [Indexed: 01/22/2023] Open
Abstract
Stem cell-based regenerative medicine is one of the most intensively researched
medical issues. Pre-clinical studies in a large-animal model, especially in swine or
miniature pigs, are highly relevant to human applications. Mesenchymal stem cells
(MSCs) have been isolated and expanded from different sources.
Collapse
Affiliation(s)
| | - André Lopes Carvalho
- Department of Oncology, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | | | - Laurindo Moacir Sassi
- Service of Oral and Maxillofacial Surgery, Erasto Gaertner Hospital, Curitiba, PR, Brazil
| | - Maria Fernanda Torres
- Laboratory of Experimental Surgery, Positivo University/Department of Anatomy, Federal University of Paraná (UFPR), Curitiba, PR, Brazil., Curitiba, PR, Brazil
| | | | | | | | - Crisciele Kuligovsky
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Curitiba, PR, Brazil
| | | |
Collapse
|
28
|
Zhu K, Lai H, Guo C, Li J, Wang Y, Wang L, Wang C. Nanovector-based prolyl hydroxylase domain 2 silencing system enhances the efficiency of stem cell transplantation for infarcted myocardium repair. Int J Nanomedicine 2014; 9:5203-15. [PMID: 25429216 PMCID: PMC4243506 DOI: 10.2147/ijn.s71586] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has attracted much attention in myocardial infarction therapy. One of the limitations is the poor survival of grafted cells in the ischemic microenvironment. Small interfering RNA-mediated prolyl hydroxylase domain protein 2 (PHD2) silencing in MSCs holds tremendous potential to enhance their survival and paracrine effect after transplantation. However, an efficient and biocompatible PHD2 silencing system for clinical application is lacking. Herein, we developed a novel PHD2 silencing system based on arginine-terminated generation 4 poly(amidoamine) (Arg-G4) nanoparticles. The system exhibited effective and biocompatible small interfering RNA delivery and PHD2 silencing in MSCs in vitro. After genetically modified MSC transplantation in myocardial infarction models, MSC survival and paracrine function of IGF-1 were enhanced significantly in vivo. As a result, we observed decreased cardiomyocyte apoptosis, scar size, and interstitial fibrosis, and increased angiogenesis in the diseased myocardium, which ultimately attenuated ventricular remodeling and improved heart function. This work demonstrated that an Arg-G4 nanovector-based PHD2 silencing system could enhance the efficiency of MSC transplantation for infarcted myocardium repair.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Changfa Guo
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Yulin Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| | - Lingyan Wang
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China ; Shanghai Institute of Cardiovascular Disease, Shanghai, People's Republic of China
| |
Collapse
|
29
|
Wendel JS, Ye L, Zhang P, Tranquillo RT, Zhang JJ. Functional consequences of a tissue-engineered myocardial patch for cardiac repair in a rat infarct model. Tissue Eng Part A 2014; 20:1325-35. [PMID: 24295499 DOI: 10.1089/ten.tea.2013.0312] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell therapies have emerged as a promising treatment for the prevention of heart failure after myocardial infarction (MI). This study evaluated the capacity of an aligned, fibrin-based, stretch-conditioned cardiac patch consisting of either the native population or a cardiomyocyte (CM)-depleted population (i.e., CM+ or CM- patches) of neonatal rat heart cells to ameliorate left ventricular (LV) remodeling in the acute-phase postinfarction in syngeneic, immunocompetent rats. Patches were exposed to 7 days of static culture and 7 days of cyclic stretching prior to implantation. Within 1 week of implantation, both patches became vascularized, and non-CMs began migrating from CM+ patches. By week 4, patches had been remodeled into collagenous tissue, and live, elongated, donor CMs were found within grafted CM+ patches. Significant improvement in cardiac contractile function was seen with the administration of the CM+ patch (ejection fraction increased from 35.1% ± 4.0% for MI only to 58.8% ± 7.3% with a CM+ patch, p<0.05) associated with a 77% reduction in infarct size (61.3% ± 7.9% for MI only, 13.9% ± 10.8% for CM+ patch, p<0.05), and the elimination of LV free-wall thinning. Decreased infarct size and reduced wall thinning also occurred with the administration of the CM- patch (infarct size 36.9% ± 10.2%, LV wall thickness: 1058.2 ± 135.4 μm for CM- patch, 661.3 ± 37.4 μm for MI only, p<0.05), but without improvements in cardiac function. Approximately 36.5% of the transplanted CMs survived at 4 weeks; however, they remained separated and electrically uncoupled from the host myocardium by a layer of CM-free tissue, which suggests that the benefits of CM+ patch transplantation resulted from paracrine mechanisms originating from CMs. Collectively, these observations suggest that the transplantation of CM-containing engineered heart tissue patches can lead to dramatic improvements in cardiac function and remodeling after acute MI.
Collapse
Affiliation(s)
- Jacqueline S Wendel
- 1 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | | | | | | | | |
Collapse
|
30
|
Allogeneic transplantation of fetal membrane-derived mesenchymal stem cell sheets increases neovascularization and improves cardiac function after myocardial infarction in rats. Transplantation 2013; 96:697-706. [PMID: 23912174 DOI: 10.1097/tp.0b013e31829f753d] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation has been pursued as a new method to repair damaged myocardium. We focused on the fetal membrane (FM) as an alternative source to bone marrow (BM)-derived MSCs. In this study, we investigated whether transplantation of allogeneic FM-MSC sheets could attenuate myocardial dysfunction in a rat chronic myocardial infarction (MI) model. METHODS Sheets of allogeneic FM-MSC or autologous BM-MSC were transplanted into the scarred myocardium 4 weeks after coronary ligation. RESULTS Four weeks after transplantation, both allogeneic FM-MSC and autologous BM-MSC sheets had significantly improved cardiac function and reduced myocardial fibrosis compared with the untreated MI group. In both MSC sheet-transplanted groups, the peri-infarct regional capillary density was increased. Some engrafted MSCs formed vascular structures and were positive for lectin I and α-smooth muscle actin. The numbers of engrafted cells and differentiated cells were very low after both types of MSC sheet transplantation. CD3 T cells did not increase in the transplantation site, but CD163 M2 macrophages increased in the groups transplanted with allogeneic FM-MSC and autologous BM-MSC. CONCLUSIONS Transplantation of allogeneic FM-MSC or autologous BM-MSC sheets attenuated myocardial dysfunction in a rat MI model to a similar degree. The engraftment rate of transplanted cells and immune cell infiltration into the transplanted area did not differ between the two types of MSC transplants. M2 macrophage induction has possible involvement in the therapeutic effects of MSC transplantation. Allogeneic FM-MSC sheet transplantation might be a new therapeutic strategy after MI.
Collapse
|
31
|
Ye L, Zhang P, Duval S, Su L, Xiong Q, Zhang J. Thymosin β4 increases the potency of transplanted mesenchymal stem cells for myocardial repair. Circulation 2013; 128:S32-41. [PMID: 24030419 DOI: 10.1161/circulationaha.112.000025] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Thymosin β4 (Tβ4) has been shown to enhance the survival of cultured cardiomyocytes. Here, we investigated whether the cytoprotective effects of Tβ4 can increase the effectiveness of transplanted swine mesenchymal stem cells (sMSCs) for cardiac repair in a rat model of myocardial infarction (MI). METHODS AND RESULTS Under hypoxic conditions, cellular damage (lactate dehydrogenase leakage), apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labelingc cells), and caspase-8 activity were significantly lower, whereas B-cell lymphoma-extra large protein expression was significantly higher, in sMSCs cultured with Tβ4 (1 μg/mL) than in sMSCs cultured without Tβ4, and Tβ4 also increased sMSC proliferation. For in vivo experiments, animals were treated with basal medium (MI: n=6), a fibrin patch (Patch: n=6), a patch containing sMSCs (sMSC: n=9), or a patch containing sMSCs and Tβ4 (sMSC/Tβ4: n=11); Tβ4 was encapsulated in gelatin microspheres to extend Tβ4 delivery. Four weeks after treatment, echocardiographic assessments of left-ventricular ejection fraction and fractional shortening were significantly better (P<0.05) in sMSC/Tβ4 animals (left-ventricular ejection fraction=51.7 ± 1.1%; fractional shortening=26.7 ± 0.7%) than in animals from MI (39 ± 3%; 19.5 ± 1.7%) and Patch (43 ± 1.4%; 21.6 ± 0.9%) groups. Histological assessment of infarct wall thickness was significantly higher (P<0.05) in sMSC/Tβ4 animals (50%, [45%, 80%]) than in animals from MI (25%, [20%, 25%]) group. Measurements in sMSC (left-ventricular ejection fraction=45 ± 2.6%; fractional shortening=22.9 ± 1.6%; TH = 43% [25%, 45%]), Patch, and MI animals were similar. Tβ4 administration also significantly increased vascular growth, the retention/survival of the transplanted sMSCs, and the recruitment of endogenous c-Kit(+) progenitor cells to the infarcted region. CONCLUSIONS Extended-release Tβ4 administration improves the retention, survival, and regenerative potency of transplanted sMSCs after myocardial injury.
Collapse
Affiliation(s)
- Lei Ye
- Division of Cardiology, Department of Medicine (L.Y., P.Z., S.D., L.S., Q.X., J.Z.), Stem Cell Institute (L.Y., J.Z.), and Department of Biomedical Engineering (J.Z.), University of Minnesota, Minneapolis, MN
| | | | | | | | | | | |
Collapse
|
32
|
Li J, Ezzelarab MB, Cooper DKC. Do mesenchymal stem cells function across species barriers? Relevance for xenotransplantation. Xenotransplantation 2013; 19:273-85. [PMID: 22978461 DOI: 10.1111/xen.12000] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Allogeneic mesenchymal stem (stromal) cells (MSC) are a promising therapy for various pathological conditions. Genetically modified pig MSC have been demonstrated to downregulate the human T-cell response to pig antigens in vitro. Before genetically modified pig MSC can be used clinically, however, evidence needs to be provided to indicate whether they will survive in a human (xenogeneic) host. LITERATURE SEARCH AND RESULTS A literature search through the end of 2011 identified 94 reports of the in vivo cross-species administration of MSC in a variety of experimental models. The majority (n = 89) involved the use of human MSC in various other species, with an occasional study using pig, rat, or guinea-pig MSC. When human MSC were used, they were largely derived from the bone marrow, adipose tissue, or umbilical cord blood. The routes of administration were varied, although almost half of the studies utilized the intravenous route. In 88 experiments (93.6%), there was evidence that the MSC engrafted and functioned across the species barrier, and in only six cases (6.4%) was there evidence of failure to function. Importantly, MSC function was confirmed in several different cross-species models. For example, human MSC functioned in no fewer than seven different recipient species. CONCLUSIONS The data provided by this literature search strengthen the hypothesis that pig MSC will function satisfactorily in a different species, for example, humans. The data also suggest that our own in vitro observations on the efficacy of pig MSC in downregulating the strength of the human T-cell response to pig antigens will likely be reproduced in vivo in pre-clinical large animal models and in clinical trials.
Collapse
Affiliation(s)
- Jiang Li
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
33
|
Cho JW, Lee CY, Ko Y. Therapeutic potential of mesenchymal stem cells overexpressing human forkhead box A2 gene in the regeneration of damaged liver tissues. J Gastroenterol Hepatol 2012; 27:1362-70. [PMID: 22432472 PMCID: PMC3492917 DOI: 10.1111/j.1440-1746.2012.07137.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM Although a liver transplantation is considered to be the only effective long-term treatment in many cases of liver diseases, it is limited by a lack of donor organs and immune rejection. As an autologous stem cell approach, this study was conducted to assess whether forkhead box A2 (Foxa2) gene overexpression in bone marrow-derived mesenchymal stem cells (MSC) could protect the liver from hepatic diseases by stimulating tissue regeneration after cell transplantation. METHODS Rat MSC (rMSC) were isolated, characterized, and induced to hepatocytes that expressed liver-specific markers. Four different treatments (control [phosphate-buffered saline], rMSC alone, rMSC/pIRES-enhanced green fluorescent protein (EGFP) vector, and rMSC/pIRES-EGFP/human Foxa2) were injected into the spleen of carbon tetrachloride-injured rats. Biochemical and histological analyses on days 30, 60, and 90 post-transplantation were performed to evaluate the therapeutic capacities of MSC overexpressing hFoxa2. RESULTS rMSC transfected with hFoxa2 were induced into hepatogenic linage and expressed several liver-specific genes, such as, Foxa2, α-fetoprotein, cytokeratin-18, hepatocyte nuclear factor-1α, and hepatocyte growth factor. A group of animals treated with MSC/hFoxa2 showed significant recovery of liver-specific enzyme expressions to normal levels at the end of the study (90 days). Furthermore, when compared to the fibrotic areas of the samples treated with MSC alone or MSC/vector, the fibrotic area of the samples treated with rMSC/hFoxa2 for 90 days significantly decreased, until they were completely gone. CONCLUSIONS Human Foxa2 efficiently promoted the incorporation of MSC into liver grafts, suggesting that hFoxa2 genes could be used for the structural or functional recovery of damaged liver cells.
Collapse
Affiliation(s)
- Jong-Woo Cho
- Department of Biotechnology, Korea UniversitySeoul, Korea
| | - Chul-Young Lee
- Department of Animal Material Engineering, College of Science and Natural Resource, Gyeongnam National University of Science and TechnologyJinju, Korea
| | - Yong Ko
- Department of Biotechnology, Korea UniversitySeoul, Korea
| |
Collapse
|
34
|
Gu M, Nguyen PK, Lee AS, Xu D, Hu S, Plews JR, Han L, Huber BC, Lee WH, Gong Y, de Almeida PE, Lyons J, Ikeno F, Pacharinsak C, Connolly AJ, Gambhir SS, Robbins RC, Longaker MT, Wu JC. Microfluidic single-cell analysis shows that porcine induced pluripotent stem cell-derived endothelial cells improve myocardial function by paracrine activation. Circ Res 2012; 111:882-93. [PMID: 22821929 DOI: 10.1161/circresaha.112.269001] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
RATIONALE Induced pluripotent stem cells (iPSCs) hold great promise for the development of patient-specific therapies for cardiovascular disease. However, clinical translation will require preclinical optimization and validation of large-animal iPSC models. OBJECTIVE To successfully derive endothelial cells from porcine iPSCs and demonstrate their potential utility for the treatment of myocardial ischemia. METHODS AND RESULTS Porcine adipose stromal cells were reprogrammed to generate porcine iPSCs (piPSCs). Immunohistochemistry, quantitative PCR, microarray hybridization, and angiogenic assays confirmed that piPSC-derived endothelial cells (piPSC-ECs) shared similar morphological and functional properties as endothelial cells isolated from the autologous pig aorta. To demonstrate their therapeutic potential, piPSC-ECs were transplanted into mice with myocardial infarction. Compared with control, animals transplanted with piPSC-ECs showed significant functional improvement measured by echocardiography (fractional shortening at week 4: 27.2±1.3% versus 22.3±1.1%; P<0.001) and MRI (ejection fraction at week 4: 45.8±1.3% versus 42.3±0.9%; P<0.05). Quantitative protein assays and microfluidic single-cell PCR profiling showed that piPSC-ECs released proangiogenic and antiapoptotic factors in the ischemic microenvironment, which promoted neovascularization and cardiomyocyte survival, respectively. Release of paracrine factors varied significantly among subpopulations of transplanted cells, suggesting that transplantation of specific cell populations may result in greater functional recovery. CONCLUSIONS In summary, this is the first study to successfully differentiate piPSCs-ECs from piPSCs and demonstrate that transplantation of piPSC-ECs improved cardiac function after myocardial infarction via paracrine activation. Further development of these large animal iPSC models will yield significant insights into their therapeutic potential and accelerate the clinical translation of autologous iPSC-based therapy.
Collapse
Affiliation(s)
- Mingxia Gu
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
den Haan MC, Grauss RW, Smits AM, Winter EM, van Tuyn J, Pijnappels DA, Steendijk P, Gittenberger-De Groot AC, van der Laarse A, Fibbe WE, de Vries AAF, Schalij MJ, Doevendans PA, Goumans MJ, Atsma DE. Cardiomyogenic differentiation-independent improvement of cardiac function by human cardiomyocyte progenitor cell injection in ischaemic mouse hearts. J Cell Mol Med 2012; 16:1508-21. [PMID: 22003890 PMCID: PMC3823219 DOI: 10.1111/j.1582-4934.2011.01468.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 09/08/2011] [Indexed: 12/31/2022] Open
Abstract
We previously showed that human cardiomyocyte progenitor cells (hCMPCs) injected after myocardial infarction (MI) had differentiated into cardiomyocytes in vivo 3 months after MI. Here, we investigated the short-term (2 weeks) effects of hCMPCs on the infarcted mouse myocardium. MI was induced in immunocompromised (NOD/scid) mice, immediately followed by intramyocardial injection of hCMPCs labelled with enhanced green fluorescent protein (hCMPC group) or vehicle only (control group). Sham-operated mice served as reference. Cardiac performance was measured 2 and 14 days after MI by magnetic resonance imaging at 9.4 T. Left ventricular (LV) pressure-volume measurements were performed at day 15 followed by extensive immunohistological analysis. Animals injected with hCMPCs demonstrated a higher LV ejection fraction, lower LV end-systolic volume and smaller relaxation time constant than control animals 14 days after MI. hCMPCs engrafted in the infarcted myocardium, did not differentiate into cardiomyocytes, but increased vascular density and proliferation rate in the infarcted and border zone area of the hCMPC group. Injected hCMPCs engraft into murine infarcted myocardium where they improve LV systolic function and attenuate the ventricular remodelling process 2 weeks after MI. Since no cardiac differentiation of hCMPCs was evident after 2 weeks, the observed beneficial effects were most likely mediated by paracrine factors, targeting amongst others vascular homeostasis. These results demonstrate that hCMPCs can be applied to repair infarcted myocardium without the need to undergo differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Melina C den Haan
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Robert W Grauss
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Anke M Smits
- Department of Molecular Cell Biology, Leiden University Medical CenterLeiden, The Netherlands
| | - Elizabeth M Winter
- Department of Anatomy and Embryology, Leiden University Medical CenterLeiden, The Netherlands
| | - John van Tuyn
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
- Department of Molecular Cell Biology, Leiden University Medical CenterLeiden, The Netherlands
| | - Daniël A Pijnappels
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Paul Steendijk
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
| | | | | | - Willem E Fibbe
- Department of Immunohematology and Blood Transfusion, Leiden University Medical CenterLeiden, The Netherlands
| | - Antoine A F de Vries
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
- Department of Molecular Cell Biology, Leiden University Medical CenterLeiden, The Netherlands
| | - Martin J Schalij
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, University Medical Center UtrechtUtrecht, The Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical CenterLeiden, The Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical CenterLeiden, The Netherlands
| |
Collapse
|
36
|
Xiong Q, Ye L, Zhang P, Lepley M, Swingen C, Zhang L, Kaufman DS, Zhang J. Bioenergetic and functional consequences of cellular therapy: activation of endogenous cardiovascular progenitor cells. Circ Res 2012; 111:455-68. [PMID: 22723295 DOI: 10.1161/circresaha.112.269894] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE The mechanism by which endogenous progenitor cells contribute to functional and beneficial effects in stem cell therapy remains unknown. OBJECTIVE Utilizing a novel (31)P magnetic resonance spectroscopy-2-dimensional chemical shift imaging method, this study examined the heterogeneity and bioenergetic consequences of postinfarction left ventricular (LV) remodeling and the mechanisms of endogenous progenitor cell contribution to the cellular therapy. METHODS AND RESULTS Human embryonic stem cell-derived vascular cells (hESC-VCs) that stably express green fluorescent protein and firefly luciferase (GFP(+)/Luc(+)) were used for the transplantation. hESC-VCs may release various cytokines to promote angiogenesis, prosurvival, and antiapoptotic effects. Both in vitro and in vivo experiments demonstrated that hESC-VCs effectively inhibit myocyte apoptosis. In the mouse model, a fibrin patch-based cell delivery resulted in a significantly better cell engraftment rate that was accompanied by a better ejection fraction. In the swine model of ischemia-reperfusion, the patch-enhanced delivery of hESC-VCs resulted in alleviation of abnormalities including border zone myocardial perfusion, contractile dysfunction, and LV wall stress. These results were also accompanied by a pronounced recruitment of endogenous c-kit(+) cells to the injury site. These improvements were directly associated with a remarkable improvement in myocardial energetics, as measured by a novel in vivo (31)P magnetic resonance spectroscopy-2-dimensional chemical shift imaging technology. CONCLUSIONS The findings of this study demonstrate that a severely abnormal heterogeneity of myocardial bioenergetics in hearts with postinfarction LV remodeling can be alleviated by the hESC-VCs therapy. These findings suggest an important therapeutic target of peri-scar border zone and a promising therapeutic potential for using hESC-VCs together with the fibrin patch-based delivery system.
Collapse
Affiliation(s)
- Qiang Xiong
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Mesenchymal stem cells for cardiac regeneration: translation to bedside reality. Stem Cells Int 2012; 2012:646038. [PMID: 22754578 PMCID: PMC3382381 DOI: 10.1155/2012/646038] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/03/2012] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. According to the World Health Organization (WHO), an estimate of 17.3 million people died from CVDs in 2008 and by 2030, the number of deaths is estimated to reach almost 23.6 million. Despite the development of a variety of treatment options, heart failure management has failed to inhibit myocardial scar formation and replace the lost cardiomyocyte mass with new functional contractile cells. This shortage is complicated by the limited ability of the heart for self-regeneration. Accordingly, novel management approaches have been introduced into the field of cardiovascular research, leading to the evolution of gene- and cell-based therapies. Stem cell-based therapy (aka, cardiomyoplasty) is a rapidly growing alternative for regenerating the damaged myocardium and attenuating ischemic heart disease. However, the optimal cell type to achieve this goal has not been established yet, even after a decade of cardiovascular stem cell research. Mesenchymal stem cells (MSCs) in particular have been extensively investigated as a potential therapeutic approach for cardiac regeneration, due to their distinctive characteristics. In this paper, we focus on the therapeutic applications of MSCs and their transition from the experimental benchside to the clinical bedside.
Collapse
|
38
|
Zhang EY, Xiong Q, Ye L, Suntharalingam P, Wang X, Astle CM, Zhang J, Harrison DE. Fetal myocardium in the kidney capsule: an in vivo model of repopulation of myocytes by bone marrow cells. PLoS One 2012; 7:e31099. [PMID: 22383995 PMCID: PMC3285614 DOI: 10.1371/journal.pone.0031099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 01/02/2012] [Indexed: 11/25/2022] Open
Abstract
Debate surrounds the question of whether the heart is a post-mitotic organ in part due to the lack of an in vivo model in which myocytes are able to actively regenerate. The current study describes the first such mouse model — a fetal myocardial environment grafted into the adult kidney capsule. Here it is used to test whether cells descended from bone marrow can regenerate cardiac myocytes. One week after receiving the fetal heart grafts, recipients were lethally irradiated and transplanted with marrow from green fluorescent protein (GFP)-expressing C57Bl/6J (B6) donors using normal B6 recipients and fetal donors. Levels of myocyte regeneration from GFP marrow within both fetal myocardium and adult hearts of recipients were evaluated histologically. Fetal myocardium transplants had rich neovascularization and beat regularly after 2 weeks, continuing at checkpoints of 1, 2, 4, 6, 8 and12 months after transplantation. At each time point, GFP-expressing rod-shaped myocytes were found in the fetal myocardium, but only a few were found in the adult hearts. The average count of repopulated myocardium with green rod-shaped myocytes was 996.8 cells per gram of fetal myocardial tissue, and 28.7 cells per adult heart tissue, representing a thirty-five fold increase in fetal myocardium compared to the adult heart at 12 months (when numbers of green rod-shaped myocytes were normalized to per gram of myocardial tissue). Thus, bone marrow cells can differentiate to myocytes in the fetal myocardial environment. The novel in vivo model of fetal myocardium in the kidney capsule appears to be valuable for testing repopulating abilities of potential cardiac progenitors.
Collapse
Affiliation(s)
- Eric Y. Zhang
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Qiang Xiong
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Lei Ye
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Piradeep Suntharalingam
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Xiaohong Wang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - C. Michael Astle
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Jianyi Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- * E-mail: (JZ); david.harrison@.jax.org (DEH)
| | - David E. Harrison
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- * E-mail: (JZ); david.harrison@.jax.org (DEH)
| |
Collapse
|
39
|
Arnous S, Mozid A, Martin J, Mathur A. Bone marrow mononuclear cells and acute myocardial infarction. Stem Cell Res Ther 2012; 3:2. [PMID: 22264393 PMCID: PMC3340546 DOI: 10.1186/scrt93] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cell transplantation is emerging as a potential therapy to treat heart diseases. Promising results from early animal studies led to an explosion of small, non-controlled clinical trials that created even further excitement by showing that stem cell transplantation improved left ventricular systolic function and enhanced remodelling. However, the specific mechanisms by which these cells improve heart function remain largely unknown. A large variety of cell types have been considered to possess the regenerative ability needed to repair the damaged heart. One of the most studied cell types is the bone marrow-derived mononuclear cells and these form the focus of this review. This review article aims to provide an overview of their use in the setting of acute myocardial infarction, the challenges it faces and the future of stem cell therapy in heart disease.
Collapse
Affiliation(s)
- Samer Arnous
- Department of Cardiology, London Chest Hospital, Bonner Road, London E2 9JX, UK
| | | | | | | |
Collapse
|
40
|
Kitada M. Mesenchymal cell populations: development of the induction systems for Schwann cells and neuronal cells and finding the unique stem cell population. Anat Sci Int 2012; 87:24-44. [PMID: 22237924 DOI: 10.1007/s12565-011-0128-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 12/20/2011] [Indexed: 01/01/2023]
Abstract
Mesenchymal cell populations, referred to as mesenchymal stem cells or multipotent stromal cells (MSCs), which include bone marrow stromal cells (BMSCs), umbilical cord stromal cells and adipose stromal cells (ASCs), participate in tissue repair when transplanted into damaged or degenerating tissues. The trophic support and immunomodulation provided by MSCs can protect against tissue damage, and the differentiation potential of these cells may help to replace lost cells. MSCs are easily accessible and can be expanded on a large scale. In addition, BMSCs and ASCs can be harvested from the patient himself. Thus, MSCs are considered promising candidates for cell therapy. In this review, I will discuss recently discovered high-efficiency induction systems for deriving Schwann cells and neurons from MSCs. Other features of MSCs that are important for tissue repair include the self-renewing property of stem cells and their potential for differentiation. Thus, I will also discuss the stemness of MSCs and describe the discovery of a certain stem cell type among adult MSCs that can self-renew and differentiate into cells of all three germ layers. Furthermore, I will explore the prospects of using this cell population for cell therapy.
Collapse
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
41
|
Wang X, From AH, Zhang J. Myocardial Regeneration. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:195-215. [DOI: 10.1016/b978-0-12-398459-3.00009-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Xiong Q, Hill KL, Li Q, Suntharalingam P, Mansoor A, Wang X, Jameel MN, Zhang P, Swingen C, Kaufman DS, Zhang J. A fibrin patch-based enhanced delivery of human embryonic stem cell-derived vascular cell transplantation in a porcine model of postinfarction left ventricular remodeling. Stem Cells 2011; 29:367-75. [PMID: 21732493 DOI: 10.1002/stem.580] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
It is unknown how to use human embryonic stem cell (hESC) to effectively treat hearts with postinfarction left ventricular (LV) remodeling. Using a porcine model of postinfarction LV remodeling, this study examined the functional improvement of enhanced delivery of combined transplantation of hESC-derived endothelial cells (ECs) and hESC-derived smooth muscle cells (SMCs) with a fibrin three-dimensional (3D) porous scaffold biomatrix. To facilitate tracking the transplanted cells, the hESCs were genetically modified to stably express green fluorescent protein and luciferase (GFP/Luc). Myocardial infarction (MI) was created by ligating the first diagonal coronary artery for 60 minutes followed by reperfusion. Two million each of GFP/Luc hESC-derived ECs and SMCs were seeded in the 3D porous biomatrix patch and applied to the region of ischemia/reperfusion for cell group (MI+P+C, n = 6), whereas biomatrix without cell (MI+P, n = 5), or saline only (MI, n = 5) were applied to control group hearts with same coronary artery ligation. Functional outcome (1 and 4 weeks follow-up) of stem cell transplantation was assessed by cardiac magnetic resonance imaging. The transplantation of hESC-derived vascular cells resulted in significant LV functional improvement. Significant engraftment of hESC-derived cells was confirmed by both in vivo and ex vivo bioluminescent imaging. The mechanism underlying the functional beneficial effects of cardiac progenitor transplantation is attributed to the increased neovascularization. These findings demonstrate a promising therapeutic potential of using these hESC-derived vascular cell types and the mode of patch delivery.
Collapse
Affiliation(s)
- Qiang Xiong
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota 5545, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hsiao FSH, Lian WS, Lin SP, Lin CJ, Lin YS, Cheng ECH, Liu CW, Cheng CC, Cheng PH, Ding ST, Lee KH, Kuo TF, Cheng CF, Cheng WTK, Wu SC. Toward an ideal animal model to trace donor cell fates after stem cell therapy: Production of stably labeled multipotent mesenchymal stem cells from bone marrow of transgenic pigs harboring enhanced green fluorescence protein gene1. J Anim Sci 2011; 89:3460-72. [DOI: 10.2527/jas.2011-3889] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
44
|
Cross DP, Wang C. Stromal-derived factor-1 alpha-loaded PLGA microspheres for stem cell recruitment. Pharm Res 2011; 28:2477-89. [PMID: 21614634 PMCID: PMC4353616 DOI: 10.1007/s11095-011-0474-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 05/03/2011] [Indexed: 01/27/2023]
Abstract
PURPOSE Stromal-derived factor-1 alpha (SDF-1α) is a chemoattractant that has been investigated for treating various diseases, with the goal of recruiting endogenous stem cells to the site of injury. Biodegradable PLGA microspheres were investigated as a means to deliver SDF-1α in a sustained-release manner. METHODS We encapsulated SDF-1α into biodegradable poly(lactide-co-glycolide) (PLGA) microspheres using a double-emulsion solvent extraction/evaporation technique. We varied several formulation parameters, characterized the in vitro release profile of SDF-1α and the size and morphology of microspheres, and determined the bioactivity of the released SDF-1α of stimulating migration of mesenchymal stem cells (MSCs). RESULTS We found that microspheres fabricated using end-capped PLGA, BSA as an excipient, and low solvent volumes yielded a high encapsulation efficiency (>64%) and released SDF-1α over a >50-day timeframe. The released SDF-1α was bioactive and caused significant migration of MSCs throughout the duration of release from the microspheres. CONCLUSIONS We have identified several variables that led to successful encapsulation of SDF-1α into PLGA microspheres. We envision that SDF-lα-loaded microspheres may serve as injectable sources of sustained-release chemokine for promoting the recruitment of endogenous stem cells to the site of injury.
Collapse
Affiliation(s)
- Daisy P. Cross
- Department of Biomedical Engineering, University of Minnesota 7-105 Hasselmo Hall, 312 Church Street S. E. Minneapolis, MN 55455, USA
| | - Chun Wang
- Department of Biomedical Engineering, University of Minnesota 7-105 Hasselmo Hall, 312 Church Street S. E. Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Suzuki G, Iyer V, Lee TC, Canty JM. Autologous mesenchymal stem cells mobilize cKit+ and CD133+ bone marrow progenitor cells and improve regional function in hibernating myocardium. Circ Res 2011; 109:1044-54. [PMID: 21885831 DOI: 10.1161/circresaha.111.245969] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Mesenchymal stem cells (MSCs) improve function after infarction, but their mechanism of action remains unclear, and the importance of reduced scar volume, cardiomyocyte proliferation, and perfusion is uncertain. OBJECTIVE The present study was conducted to test the hypothesis that MSCs mobilize bone marrow progenitor cells and improve function by stimulating myocyte proliferation in collateral-dependent hibe rnating myocardium. METHODS AND RESULTS Swine with chronic hibernating myocardium received autologous intracoronary MSCs (icMSCs; ≈44 ×10(6) cells, n = 10) 4 months after instrumentation and were studied up to 6 weeks later. Physiological and immunohistochemical findings were compared with untreated hibernating animals (n = 7), sham-normal animals (n = 5), and icMSC-treated sham-normal animals (n = 6). In hibernating myocardium, icMSCs increased function (percent wall thickening of the left anterior descending coronary artery 24 ± 4% to 43 ± 5%, P < 0.05), although left anterior descending coronary artery flow reserve (adenosine/rest) remained critically impaired (1.2 ± 0.1 versus 1.2 ± 0.1). Circulating cKit+ and CD133+ bone marrow progenitor cells increased transiently after icMSC administration, with a corresponding increase in myocardial cKit+/CD133+ and cKit+/CD133- bone marrow progenitor cells (total cKit+ from 223 ± 49 to 4415 ± 866/10(6) cardiomyocytes, P < 0.05). In hibernating hearts, icMSCs increased Ki67+ cardiomyocytes (from 410 ± 83 to 2460 ± 610/10(6) nuclei, P < 0.05) and phospho-histone H3-positive cardiomyocytes (from 9 ± 5 to 116 ± 12/10(6) nuclei, P < 0.05). Myocyte nuclear number (from 75 336 ± 5037 to 114 424 ± 9564 nuclei/mm3, P < 0.01) and left ventricular mass (from 2.5 ± 0.1 to 2.8 ± 0.1 g/kg, P < 0.05) increased, yet myocytes were smaller (14.5 ± 0.4 versus 16.5 ± 0.4 μm, P < 0.05), which supports endogenous cardiomyocyte proliferation. In sham-normal animals, icMSCs increased myocardial bone marrow progenitor cells with no effect on myocyte proliferation or regional function. CONCLUSIONS Our results indicate that icMSCs improve function in hibernating myocardium independent of coronary flow or reduced scar volume. This arises from stimulation of myocyte proliferation with increases in cKit+/CD133+ bone marrow progenitor cells and cKit+/CD133- resident stem cells, which increase myocyte number and reduce cellular hypertrophy.
Collapse
Affiliation(s)
- Gen Suzuki
- VA WNY Health Care System, Buffalo, NY, USA
| | | | | | | |
Collapse
|
46
|
Choh SY, Cross D, Wang C. Facile Synthesis and Characterization of Disulfide-Cross-Linked Hyaluronic Acid Hydrogels for Protein Delivery and Cell Encapsulation. Biomacromolecules 2011; 12:1126-36. [DOI: 10.1021/bm101451k] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Sun-Young Choh
- Department of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo Hall, 312 Church Street South East, Minneapolis, Minnesota 55455, United States
| | - Daisy Cross
- Department of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo Hall, 312 Church Street South East, Minneapolis, Minnesota 55455, United States
| | - Chun Wang
- Department of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo Hall, 312 Church Street South East, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
47
|
Allogeneic administration of fetal membrane-derived mesenchymal stem cells attenuates acute myocarditis in rats. J Mol Cell Cardiol 2010; 49:753-61. [DOI: 10.1016/j.yjmcc.2010.07.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 05/31/2010] [Accepted: 07/30/2010] [Indexed: 01/14/2023]
|
48
|
Zhou J, Yu G, Cao C, Pang J, Chen X. Bone morphogenetic protein-7 promotes chondrogenesis in human amniotic epithelial cells. INTERNATIONAL ORTHOPAEDICS 2010; 35:941-8. [PMID: 20803292 DOI: 10.1007/s00264-010-1116-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 08/03/2010] [Accepted: 08/10/2010] [Indexed: 12/16/2022]
Abstract
Bone morphogenetic proteins (BMPs) play important roles at multiple stages of chondrogenesis. This study was undertaken to investigate the potential role of bone morphogenetic protein-7 (BMP-7) in the differentiation of chondrocytes using tissue engineering techniques. The impact of BMP-7 on human amniotic epithelial cells (hAECs) was tested. The hAECs were treated either with recombinant human BMP-7 cDNA or with transforming growth factor beta 1 (TGF-β1) as a positive control for three weeks in vitro. Cartilaginous differentiation and proliferation were assayed by quantitative RT-PCR, histology, and in situ hybridization. Our results were such that hAECs treated with either BMP-7 or TGF-β1 expressed cartilage markers (aggrecan, Sox9, CEP-68, and type II and X collagens) within three weeks. Compared with a control vector, BMP-7 induced a decrease in type I collagen expression, while the transcription of the cartilage-specific type II collagen remained stable. In induction experiments, BMP-7 transgenic hAECs exhibited the largest amount of matrix synthesis. In conclusion, these data indicate that BMP-7 plays an important role in inducing the production of cartilage by hAECs in vitro. Cartilage differentiation and matrix maturation can be promoted by BMPs in a cartilage engineering paradigm. These properties make BMPs promising tools in the engineering of cartilaginous joint bio-prostheses and as candidate biological agents or genes for cartilage stabilisation.
Collapse
Affiliation(s)
- Junjie Zhou
- Department of Orthopaedic Surgery, Tongji Hospital of Tongji University, Shanghai 200065, China
| | | | | | | | | |
Collapse
|
49
|
Shi XL, Gu JY, Han B, Xu HY, Fang L, Ding YT. Magnetically labeled mesenchymal stem cells after autologous transplantation into acutely injured liver. World J Gastroenterol 2010; 16:3674-9. [PMID: 20677339 PMCID: PMC2915427 DOI: 10.3748/wjg.v16.i29.3674] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate tracking of magnetically labeled mesenchymal stem cells (MSCs) after intraportal transplantation.
METHODS: Mononuclear cells were isolated from bone marrow aspirates of pigs by density gradient centrifugation, cultured and expanded, after which, they were incubated with super paramagnetic iron oxide (SPIO). Prussian blue staining was performed to highlight intracellular iron. To establish swine models of acute liver injury, 0.5 g/kg D-galactosamine was administrated to 10 pigs, six of which were injected via their portal veins with SPIO-labeled MSCs, while the remaining four were injected with unlabeled cells. Magnetic resonance imaging (MRI) was performed with a clinical 1.5T MR scanner immediately before transplantation and 6 h, 3 d, 7 d and 14 d after transplantation. Prussian blue staining was again performed with the tissue slices at the endpoint.
RESULTS: Prussian blue staining of SPIO-labeled MSCs had a labeling efficiency of almost 100%. Signal intensity loss in the liver by SPIO labeling on the FFE (T2*WI) sequence persisted until 14 d after transplantation. Histological analysis by Prussian blue staining confirmed homing of labeled MSCs in the liver after 14 d; primarily distributed in hepatic sinusoids and liver parenchyma.
CONCLUSION: MSCs were successfully labeled with SPIO in vitro. MRI can monitor magnetically labeled MSCs transplanted into the liver.
Collapse
|
50
|
Trivedi P, Tray N, Nguyen T, Nigam N, Gallicano GI. Mesenchymal Stem Cell Therapy for Treatment of Cardiovascular Disease: Helping People Sooner or Later. Stem Cells Dev 2010; 19:1109-20. [DOI: 10.1089/scd.2009.0465] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Premal Trivedi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Nancy Tray
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Thuy Nguyen
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Neha Nigam
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - G. Ian Gallicano
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|