1
|
Li H, Lin J, Wang L, He R, Li J, Chen M, Zhang W, Zhang C. Interleukin 4 improved adipose-derived stem cells engraftment via interacting with fibro/adipogenic progenitors in dystrophic mice. Cell Mol Life Sci 2023; 80:375. [PMID: 38010513 PMCID: PMC10682070 DOI: 10.1007/s00018-023-05020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023]
Abstract
Adipose-derived stem cells (ADSC) therapy shows promise as an effective treatment for dystrophinopathy. Fibro-/adipogenic progenitors (FAPs) play an essential role in the myogenesis of muscle satellite cells and contribute to muscle fibrosis and adipocyte infiltration. The interleukin 4 (IL-4) pathway acts as a switch that regulates the functions of FAPs. The interaction between FAPs and engrafted cells remains unclear. In this study, we used a co-culture system to investigate possible crosstalk between the FAPs of dystrophic mice and ADSC overexpressing IL4 (IL4-ADSC) and control ADSC. Systemic transplantation of IL4-ADSC and control ADSC in dystrophic mice was conducted for 16 weeks, after which motor function and molecular improvements were evaluated. Overexpression of IL4 in ADSC significantly promoted myogenesis in vitro, increasing the expression of Pax7, Myogenin, and MyHC. Co-culture indicated that although myoblasts derived from control ADSC promoted adipogenic and fibrogenic differentiation of FAPs, FAPs did not significantly affect myogenesis of ADSC-derived myoblasts. However, overexpression of IL4 in ADSC inhibited their myotube-dependent promotion of FAPs differentiation on the one hand and promoted FAPs to enhance myogenesis on the other. Dystrophic mice administered with IL4-ADSC-derived myoblasts displayed significantly better motor ability, more engrafted cells showing dystrophin expression, and less muscle fibrosis, intramuscular adipocytes, and macrophage infiltration than mice administered control-ADSC-derived myoblasts. In conclusion, IL4 activation enhanced the therapeutic potential of ADSC transplantation in dystrophic mice, possibly by improving the myogenesis of IL4-ADSC and altering the crosstalk between engrafted stem cells and resident FAPs.
Collapse
Affiliation(s)
- Huan Li
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Jinfu Lin
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Liang Wang
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Ruojie He
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Jing Li
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Menglong Chen
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, 510080, China
| | - Weixi Zhang
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China.
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Pang KT, Loo LSW, Chia S, Ong FYT, Yu H, Walsh I. Insight into muscle stem cell regeneration and mechanobiology. Stem Cell Res Ther 2023; 14:129. [PMID: 37173707 PMCID: PMC10176686 DOI: 10.1186/s13287-023-03363-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Stem cells possess the unique ability to differentiate into specialized cell types. These specialized cell types can be used for regenerative medicine purposes such as cell therapy. Myosatellite cells, also known as skeletal muscle stem cells (MuSCs), play important roles in the growth, repair, and regeneration of skeletal muscle tissues. However, despite its therapeutic potential, the successful differentiation, proliferation, and expansion processes of MuSCs remain a significant challenge due to a variety of factors. For example, the growth and differentiation of MuSCs can be greatly influenced by actively replicating the MuSCs microenvironment (known as the niche) using mechanical forces. However, the molecular role of mechanobiology in MuSC growth, proliferation, and differentiation for regenerative medicine is still poorly understood. In this present review, we comprehensively summarize, compare, and critically analyze how different mechanical cues shape stem cell growth, proliferation, differentiation, and their potential role in disease development (Fig. 1). The insights developed from the mechanobiology of stem cells will also contribute to how these applications can be used for regenerative purposes using MuSCs.
Collapse
Affiliation(s)
- Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
- School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, 62 Nanyang Drive, N1.2-B3, Singapore, 637459, Singapore.
| | - Larry Sai Weng Loo
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Francesca Yi Teng Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hanry Yu
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Interdisplinary Science and Engineering Program, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
3
|
Li H, Lin J, Wang L, He R, Li J, Chen M, Zhang W, Zhang C. Interleukin-4 improved adipose-derived stem cells engraftment via interacting with fibro/adipogenic progenitors in dystrophic mice.. [DOI: 10.21203/rs.3.rs-2321597/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Abstract
Adipose-derived stem cells (ADSC) therapy is a promising therapy for dystrophinopathy. Fibro/adipogenic progenitors (FAP) are important in regulating the myogenesis of muscle satellite cells and contribute to muscle fibrosis and adipocyte infiltration. The interleukin-4 (IL4) pathway is found to be a switcher regulating the functions of FAP. The interaction between FAP and engrafted cells has not yet been studied. We used a co-culture system to investigate the possible crosstalk between FAP of dystrophic mice and IL4-overexpressed ADSC (IL4-ADSC) and control ADSC. The systemic transplantation of IL4-ADSC and control ADSC was conducted in dystrophic mice for 16 weeks and motor function and molecular improvements of mice were evaluated. Overexpression of IL4 in ADSC significantly promoted terminal myogenesis in vitro with significant increased expression of Myogenin and MyHC. Through co-culture, we discovered that myoblasts derived from control ADSC promoted adipogenic and fibrogenic differentiation of FAP, but FAP did not significantly affect their myogenesis, while overexpression of IL4 in ADSC inhibited their myotube-dependent promotion of FAP differentiation but promoted FAP to support myogenesis. Dystrophic mice delivered with IL4-ADSC-derived myoblasts had a significant better motor ability, more engrafted cells with dystrophin expression, less muscle fibrosis, and intramuscular adipocytes and macrophage infiltration than mice delivered with control-ADSC-derived myoblasts. Our results revealed the importance of focusing on the crosstalk between engrafted cells and resident FAP in cell therapy and the positive therapeutic effect of IL4 administration combined with ADSC therapy in dystrophic mice.
Collapse
Affiliation(s)
- Huan Li
- Sun Yat-sen University First Affiliated Hospital
| | | | - Liang Wang
- Sun Yat-sen University First Affiliated Hospital
| | - Ruojie He
- Sun Yat-sen University First Affiliated Hospital
| | - Jing Li
- Sun Yat-sen University First Affiliated Hospital
| | | | - Weixi Zhang
- Sun Yat-sen University First Affiliated Hospital
| | - Cheng Zhang
- Sun Yat-sen University First Affiliated Hospital
| |
Collapse
|
4
|
Ragni E, Viganò M, De Luca P, Pedrini E, de Girolamo L. Adipose-Derived Stem/Stromal Cells, Stromal Vascular Fraction, and Microfragmented Adipose Tissue. ORTHOBIOLOGICS 2022:47-61. [DOI: 10.1007/978-3-030-84744-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Reksodiputro MH, Hutauruk SM, Koento T, Fardizza F, Hakim RYR, Audindra S, Yosia M. Randomised clinical trial: Effect of administering platelet-rich fibrin to autologous fat tissue in injection laryngoplasty for vocal cord paralysis. Ann Med Surg (Lond) 2021; 68:102564. [PMID: 34367634 PMCID: PMC8326719 DOI: 10.1016/j.amsu.2021.102564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/03/2022] Open
Abstract
The vocal cord in humans is essential in producing voice used in communication and interaction between us. Vocal cord paralysis causes dysphonia, which interferes with communication, causing disruptions towards social activity and daily activities. One of the managements for vocal cord paralysis is medialization and augmentation of the vocal cord through injection laryngoplasty. Autologous fat is one of the best fillers used in this procedure, but it is highly absorbable and can be reabsorbed very quickly when injected into body tissues. Platelet Rich Fibrin (PRF) is a biomaterial consisting of growth factors that are thought to improve fat tissue viability by increasing adipogenesis and angiogenesis. Improvement in fat viability will improve clinical outcomes after the laryngoplasty procedure, potentially reducing the number of repeated injections needed to achieve a satisfactory resolution to vocal cord paralysis. The study evaluates a combination of PRF and autologous microlobular fat compared with autologous microlobular fat alone on laryngoplasty. This single-blinded randomised control trial recruit a total of 18 patients, which are then randomised into the treatment and control groups. The evaluation was done via computerized acoustic analysis/Multidimensional Voice Program (MDVP) parameters and maximum phonation time. The MDVP results and maximum phonation time in both groups showed clinical improvement after the operation with no statistically significant differences.
PRF and PRP are platelet-derived products commonly used tissue engineering procedure. PRF is an enhanced version of PRP believed to have superior growth factor release quality. Microlobular fat containing adipose stem cell is used as filler in injection laryngoplasty for medialization of vocal cord. This study observed combination of PRF and autologous microlobular fat filler for injection laryngoplasty. The MDVP results and maximum phonation time showed clinical improvement after the injection laryngoplasty.
Collapse
Affiliation(s)
- M H Reksodiputro
- Facial Plastic Reconstructive Division, Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| | - S M Hutauruk
- Larynx Pharynx Division, Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| | - T Koento
- Facial Plastic Reconstructive Division, Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| | - F Fardizza
- Larynx Pharynx Division, Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| | - R Y R Hakim
- Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| | - S Audindra
- Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| | - M Yosia
- Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital Jakarta, Indonesia
| |
Collapse
|
6
|
Abreu de Melo MI, da Silva Cunha P, Coutinho de Miranda M, Faraco CCF, Barbosa JL, da Fonseca Ferreira A, Kunrath Lima M, Faria JAQA, Rodrigues MÂ, de Goes AM, Gomes DA. Human adipose-derived stromal/stem cells are distinct from dermal fibroblasts as evaluated by biological characterization and RNA sequencing. Cell Biochem Funct 2021; 39:442-454. [PMID: 33389760 DOI: 10.1002/cbf.3610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/27/2020] [Accepted: 12/13/2020] [Indexed: 01/08/2023]
Abstract
Human adipose-derived stromal/stem cells (ASC) have immunomodulatory properties and the potential to differentiate into several cell lines, important for application in regenerative medicine. However, the contamination with dermal fibroblasts (FIB) can impair the beneficial effects of ASC in cell therapy. It is then essential to develop new strategies that contribute to the distinction between these two cell types. In this study, we performed functional assays, high-throughput RNA sequencing (RNA-Seq) and quantitative PCR (qPCR) to find new markers that can distinguish ASC and FIB. We showed that ASC have adipogenic and osteogenic differentiation capacity and alkaline phosphatase activity, not observed in FIB. Gene expression variation analysis identified more than 2000 differentially expressed genes (DEG) between these two cell types. We validated 16 genes present in the list of DEG, including the alkaline phosphatase gene (ALPL). In conclusion, we showed that ASC and FIB have distinct biological properties as demonstrated by alkaline phosphatase activity and differentiation capacity, besides having different gene expression profiles. SIGNIFICANCE OF THE STUDY: Although many differences between stromal stem cells derived from human adipose tissue (ASC) and human dermal fibroblasts (FIB) are described, it is still difficult to find specific markers to differentiate them. This problem can interfere with the therapeutic use of ASC. This work aimed to find new markers to differentiate these two cell populations. Our findings suggest that these cells can be distinguished by biological and molecular characteristics, such as adipogenic and osteogenic differentiation, alkaline phosphatase activity and differential gene expression profiles. The DEG were related to the regulation of the cell cycle, development process, structural organization of the cell and synthesis of the extracellular matrix. This study helps to find new cellular markers to distinguish the two populations and to better understand the properties of these cells, which can improve cell therapy.
Collapse
Affiliation(s)
- Mariane Izabella Abreu de Melo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pricila da Silva Cunha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Coutinho de Miranda
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Cristina Fraga Faraco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Joana Lobato Barbosa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andrea da Fonseca Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marianna Kunrath Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jerusa Araújo Quintão Arantes Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Brazil
| | - Michele Ângela Rodrigues
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alfredo Miranda de Goes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dawidson Assis Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
7
|
Kozlowska U, Krawczenko A, Futoma K, Jurek T, Rorat M, Patrzalek D, Klimczak A. Similarities and differences between mesenchymal stem/progenitor cells derived from various human tissues. World J Stem Cells 2019; 11:347-374. [PMID: 31293717 PMCID: PMC6600850 DOI: 10.4252/wjsc.v11.i6.347] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/03/2018] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) constitute a promising tool in regenerative medicine and can be isolated from different human tissues. However, their biological properties are still not fully characterized. Whereas MSCs from different tissue exhibit many common characteristics, their biological activity and some markers are different and depend on their tissue of origin. Understanding the factors that underlie MSC biology should constitute important points for consideration for researchers interested in clinical MSC application.
AIM To characterize the biological activity of MSCs during longterm culture isolated from: bone marrow (BM-MSCs), adipose tissue (AT-MSCs), skeletal muscles (SM-MSCs), and skin (SK-MSCs).
METHODS MSCs were isolated from the tissues, cultured for 10 passages, and assessed for: phenotype with immunofluorescence and flow cytometry, multipotency with differentiation capacity for osteo-, chondro-, and adipogenesis, stemness markers with qPCR for mRNA for Sox2 and Oct4, and genetic stability for p53 and c-Myc; 27 bioactive factors were screened using the multiplex ELISA array, and spontaneous fusion involving a co-culture of SM-MSCs with BM-MSCs or AT-MSCs stained with PKH26 (red) or PKH67 (green) was performed.
RESULTS All MSCs showed the basic MSC phenotype; however, their expression decreased during the follow-up period, as confirmed by fluorescence intensity. The examined MSCs express CD146 marker associated with proangiogenic properties; however their expression decreased in AT-MSCs and SM-MSCs, but was maintained in BM-MSCs. In contrast, in SK-MSCs CD146 expression increased in late passages. All MSCs, except BM-MSCs, expressed PW1, a marker associated with differentiation capacity and apoptosis. BM-MSCs and AT-MSCs expressed stemness markers Sox2 and Oct4 in long-term culture. All MSCs showed a stable p53 and c-Myc expression. BM-MSCs and AT-MSCs maintained their differentiation capacity during the follow-up period. In contrast, SK-MSCs and SM-MSCs had a limited ability to differentiate into adipocytes. BM-MSCs and AT-MSCs revealed similarities in phenotype maintenance, capacity for multilineage differentiation, and secretion of bioactive factors. Because AT-MSCs fused with SM-MSCs as effectively as BM-MSCs, AT-MSCs may constitute an alternative source for BM-MSCs.
CONCLUSION Long-term culture affects the biological activity of MSCs obtained from various tissues. The source of MSCs and number of passages are important considerations in regenerative medicine.
Collapse
Affiliation(s)
- Urszula Kozlowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| | - Agnieszka Krawczenko
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| | - Katarzyna Futoma
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| | - Tomasz Jurek
- Department of Forensic Medicine, Wroclaw Medical University, Wroclaw 50-345, Poland
| | - Marta Rorat
- Department of Forensic Medicine, Wroclaw Medical University, Wroclaw 50-345, Poland
| | - Dariusz Patrzalek
- Faculty of Health Science, Department of Physiotherapy, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Aleksandra Klimczak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| |
Collapse
|
8
|
Metairon S, Zamboni CB, Suzuki MF, Bueno CR. Evaluation of ions and metals in the blood of GRMD dogs submitted to hASCs therapy by NAA and XRF techniques. Appl Radiat Isot 2018; 143:107-112. [PMID: 30408633 DOI: 10.1016/j.apradiso.2018.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 10/28/2022]
Abstract
The elements Br, Ca, Cl, Cr, Fe, K, Mg, Na, P, Rb, S, and Zn were investigated in the whole blood samples of Golden Retriever dogs submitted to cell therapy (hASCs). These analyses were performed over 2 years using Neutron Activation Analysis and X-Ray Fluorescence techniques. The results were compared with control and untreated dog's. A significant increase was observed in K blood levels. There was also variation in blood levels of Br, Cr, Fe, Rb, S, and Zn.
Collapse
Affiliation(s)
- Sabrina Metairon
- Instituto de Pesquisas Energéticas e Nucleares, IPEN - CNEN/SP, Centro do Reator de Pesquisa - CRPq, Av. Professor Lineu Prestes 2242, 05508-000 São Paulo, SP, Brazil.
| | - Cibele B Zamboni
- Instituto de Pesquisas Energéticas e Nucleares, IPEN - CNEN/SP, Centro do Reator de Pesquisa - CRPq, Av. Professor Lineu Prestes 2242, 05508-000 São Paulo, SP, Brazil.
| | - Miriam F Suzuki
- Instituto de Pesquisas Energéticas e Nucleares, IPEN - CNEN/SP, Centro do Reator de Pesquisa - CRPq, Av. Professor Lineu Prestes 2242, 05508-000 São Paulo, SP, Brazil.
| | - Carlos R Bueno
- Centro de Estudos do Genoma Humano, Instituto de Biociências, USP, Rua: do Matão, Travessa 13, 106, 05508-090 São Paulo, SP, Brazil.
| |
Collapse
|
9
|
Personalized gene and cell therapy for Duchenne Muscular Dystrophy. Neuromuscul Disord 2018; 28:803-824. [DOI: 10.1016/j.nmd.2018.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 01/09/2023]
|
10
|
Kaleka CC, Zucconi E, Vieira TDS, Secco M, Ferretti M, Cohen M. Evaluation of different commercial hyaluronic acids as a vehicle for injection of human adipose-derived mesenchymal stem cells. Rev Bras Ortop 2018; 53:557-563. [PMID: 30245994 PMCID: PMC6147760 DOI: 10.1016/j.rboe.2018.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 05/19/2017] [Indexed: 12/05/2022] Open
Abstract
Objective The main purpose of this study is to evaluate, in vitro, the cytotoxicity of different commercial brands of hyaluronic acids to be used as a vehicle for injection of human adipose-derived mesenchymal stem cells (AD-MSCs). Methods AD-MSCs were divided into seven groups: one control group where AD-MSCs were cultivated with phosphate-buffered saline (PBS) and six other groups where AD-MSCs were cultivated with different commercial brands of hyaluronic acid. AD-MSC viability analysis was performed after 4, 24, and 48 h in contact with each treatment, using the trypan staining method on a Countess automated cell counter (Thermo Fisher Scientific). Results The results clearly demonstrated a significant difference in cell viability when AD-MSCs were exposed to different hyaluronic acids when compared with the control group. Conclusion These data suggest that hyaluronic acid can be used as a vehicle for injection of human AD-MSCs, but caution is needed to choose the best product, aiming at its future therapeutic application.
Collapse
Affiliation(s)
- Camila Cohen Kaleka
- Instituto Cohen Ortopedia, Reabilitação e Medicina do Esporte, São Paulo, SP, Brazil.,Programa do Aparelho Locomotor, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Eder Zucconi
- Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo (USP), São Paulo, SP, Brazil.,Laboratório StemCorp de Tecnologia em Células-Tronco, São Paulo, SP, Brazil
| | | | - Mariane Secco
- Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo (USP), São Paulo, SP, Brazil.,Laboratório StemCorp de Tecnologia em Células-Tronco, São Paulo, SP, Brazil
| | - Mário Ferretti
- Programa do Aparelho Locomotor, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Moisés Cohen
- Instituto Cohen Ortopedia, Reabilitação e Medicina do Esporte, São Paulo, SP, Brazil
| |
Collapse
|
11
|
Gomes JP, Coatti GC, Valadares MC, Assoni AF, Pelatti MV, Secco M, Zatz M. Human Adipose-Derived CD146+ Stem Cells Increase Life Span of a Muscular Dystrophy Mouse Model More Efficiently than Mesenchymal Stromal Cells. DNA Cell Biol 2018; 37:798-804. [DOI: 10.1089/dna.2018.4158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Juliana P. Gomes
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Giuliana C. Coatti
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marcos C. Valadares
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Amanda F. Assoni
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mayra V. Pelatti
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mariane Secco
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mayana Zatz
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
12
|
Kaleka CC, Zucconi E, Vieira TDS, Secco M, Ferretti M, Cohen M. Avaliação de diferentes ácidos hialurônicos comerciais como veículo de injeção para células mesenquimais humanas derivadas do tecido adiposo. Rev Bras Ortop 2018. [DOI: 10.1016/j.rbo.2017.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
13
|
Park JU, Kwon ST. Potential of autologous adipose-derived stem cells to regenerate atrophied muscle in a rat model. Wound Repair Regen 2018; 25:944-955. [DOI: 10.1111/wrr.12598] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/13/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Ji-Ung Park
- Department of Plastic and Reconstructive Surgery; Seoul National University Boramae Hospital; Seoul Republic of Korea
| | - Sung-Tack Kwon
- Department of Plastic and Reconstructive Surgery; Seoul National University College of Medicine; Seoul Republic of Korea
| |
Collapse
|
14
|
Sung SE, Hwang M, Kim AY, Lee EM, Lee EJ, Hwang SK, Kim SY, Kim HK, Jeong KS. MyoD Overexpressed Equine Adipose-Derived Stem Cells Enhanced Myogenic Differentiation Potential. Cell Transplant 2018; 25:2017-2026. [PMID: 26892394 DOI: 10.3727/096368916x691015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells could potentially be used in the clinical treatment of muscle disorders and muscle regeneration. Adipose-derived stem cells (ADSCs) can be easily isolated from adipose tissue, as opposed to stem cells of other tissues. We believe that cell therapy using ADSCs could be applied to muscle disorders in horses and other species. We sought to improve the myogenic differentiation potential of equine ADSCs (eqADSCs) using a MyoD lentiviral vector. MyoD lentiviruses were transduced into eqADSCs and selected using puromycin. Cells were cultured in differentiation media containing 5% horse serum, and after 5 days the MyoD-transduced cells differentiated into myogenic cells (MyoD-eqADSCs). Using green fluorescent protein (GFP), MyoD-eqADSCs were purified and transplanted into the tibialis anterior muscles of mice after they were injured with the myotoxin notexin. The mice were sacrificed to examine any regeneration in the tibialis anterior muscle 4 weeks after the MyoD-eqADSCs were injected. The MyoD-eqADSCs cultured in growth media expressed murine and equine MyoD; however, they did not express late differentiation markers such as myogenin (MYOG). When cells were grown in differentiation media, the expression of MYOG was clearly observed. According to our reverse transcription polymerase chain reaction and immunocytochemistry results, MyoD-eqADSCs expressed terminal myogenic phase genes, such as those encoding dystrophin, myosin heavy chain, and troponin I. The MyoD-eqADSCs fused to each other, and the formation of myotube-like cells from myoblasts in differentiation media occurred between days 5 and 14 postplating. In mice, we observed GFP-positive myofibers, which had differentiated from the injected MyoD-eqADSCs. Our approaches improved the myogenic differentiation of eqADSCs through the forced expression of murine MyoD. Our findings suggest that limitations in the treatment of equine muscle disorders could be overcome using ADSCs.
Collapse
Affiliation(s)
- Soo-Eun Sung
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Meeyul Hwang
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ah-Young Kim
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Eun-Mi Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Eun-Joo Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Su-Kyeong Hwang
- Department of Pediatrics, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Shin-Yoon Kim
- Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hong-Kyun Kim
- Department of Ophthalmology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
15
|
Milner DJ, Bionaz M, Monaco E, Cameron JA, Wheeler MB. Myogenic potential of mesenchymal stem cells isolated from porcine adipose tissue. Cell Tissue Res 2018; 372:507-522. [PMID: 29318389 DOI: 10.1007/s00441-017-2764-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/02/2017] [Indexed: 12/31/2022]
Abstract
Advances in stem cell biology and materials science have provided a basis for developing tissue engineering methods to repair muscle injury. Among stem cell populations with potential to aid muscle repair, adipose-derived mesenchymal stem cells (ASC) hold great promise. To evaluate the possibility of using porcine ASC for muscle regeneration studies, we co-cultured porcine ASC with murine C2C12 myoblasts. These experiments demonstrated that porcine ASC display significant myogenic potential. Co-culture of ASC expressing green fluorescent protein (GFP) with C2C12 cells resulted in GFP+ myotube formation, indicating fusion of ASC with myoblasts to form myotubes. The presence of porcine lamin A/C positive nuclei in myotubes and RTqPCR analysis of porcine myogenin and desmin expression confirmed that myotube nuclei derived from ASC contribute to muscle gene expression. Co-culturing GFP+ASC with porcine satellite cells demonstrated enhanced myogenic capability of ASC, as the percentage of labeled myotubes increased compared to mouse co-cultures. Enhancing myogenic potential of ASC through soluble factor treatment or expansion of ASC with innate myogenic capacity should allow for their therapeutic use to regenerate muscle tissue lost to disease or injury.
Collapse
Affiliation(s)
- Derek J Milner
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Massimo Bionaz
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Elisa Monaco
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jo Ann Cameron
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Matthew B Wheeler
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA.
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
16
|
Preferred M2 Polarization by ASC-Based Hydrogel Accelerated Angiogenesis and Myogenesis in Volumetric Muscle Loss Rats. Stem Cells Int 2017; 2017:2896874. [PMID: 28694827 PMCID: PMC5488492 DOI: 10.1155/2017/2896874] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 01/26/2023] Open
Abstract
Volumetric muscle loss (VML) injury resulted from massive muscle defects and diseases for which there are still no effective therapeutic treatments. This study aimed to investigate the effects of rat adipose-derived mesenchymal stem cells (rASCs) and rASCs-conditioned medium- (CM-) based type I collagen hydrogel on macrophage (MP) transition, myogenesis, and vascularization in the rat VML model. Laser Doppler results demonstrated much higher blood flow in the rASC- and CM-based hydrogel groups. qRT-PCR, hematoxylin and eosin, immunofluorescence, and Sirius Red staining manifested that both rASCs and CM-based hydrogel implantation accelerated muscle repair with upregulated angiogenesis and myogenesis, attenuated inflammation while facilitating M2 transition, and decreased the collagen deposition compared with the hydrogel group. In vitro experiments indicated that factors secreted from polarized M2 MPs could accelerate the migration and tube formation capacities of HUVECs. These results suggested that rASCs exerted immunomodulatory effects on MPs which further enhanced the proangiogenic potential on ECs to promote myogenesis and angiogenesis during muscle repair. These fundamental results support further clinical applications of ASCs for muscle loss injury.
Collapse
|
17
|
Naderi N, Combellack EJ, Griffin M, Sedaghati T, Javed M, Findlay MW, Wallace CG, Mosahebi A, Butler PEM, Seifalian AM, Whitaker IS. The regenerative role of adipose-derived stem cells (ADSC) in plastic and reconstructive surgery. Int Wound J 2017; 14:112-124. [PMID: 26833722 PMCID: PMC7949873 DOI: 10.1111/iwj.12569] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/24/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
The potential use of stem cell-based therapies for the repair and regeneration of various tissues and organs offers a paradigm shift in plastic and reconstructive surgery. The use of either embryonic stem cells (ESC) or induced pluripotent stem cells (iPSC) in clinical situations is limited because of regulations and ethical considerations even though these cells are theoretically highly beneficial. Adult mesenchymal stem cells appear to be an ideal stem cell population for practical regenerative medicine. Among these cells, adipose-derived stem cells (ADSC) have the potential to differentiate the mesenchymal, ectodermal and endodermal lineages and are easy to harvest. Additionally, adipose tissue yields a high number of ADSC per volume of tissue. Based on this background knowledge, the purpose of this review is to summarise and describe the proliferation and differentiation capacities of ADSC together with current preclinical data regarding the use of ADSC as regenerative tools in plastic and reconstructive surgery.
Collapse
Affiliation(s)
- Naghmeh Naderi
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| | - Emman J Combellack
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| | - Michelle Griffin
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
| | - Tina Sedaghati
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
| | - Muhammad Javed
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| | - Michael W Findlay
- Plastic & Reconstructive SurgeryStanford University Medical CentreStanfordCAUSA
| | | | - Afshin Mosahebi
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
- Department of Plastic SurgeryRoyal Free NHS Foundation TrustLondonUK
| | - Peter EM Butler
- Department of Plastic SurgeryRoyal Free NHS Foundation TrustLondonUK
| | - Alexander M Seifalian
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| |
Collapse
|
18
|
Devi MG, Sharma A, Mohanty S, Jain N, Verma K, Padma MV, Pal P, Chabbra HS, Khadilkar S, Prabhakar S, Singh G. Report: Stem cell applications in neurological practice, an expert group consensus appraisal. Ann Indian Acad Neurol 2016; 19:367-73. [PMID: 27570390 PMCID: PMC4980961 DOI: 10.4103/0972-2327.186825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Introduction: Neurologists in their clinical practice are faced with inquiries about the suitability of stem cell approaches by patients with a variety of acute and chronic (namely neurodegenerative) disorders. The challenge is to provide these patients with accurate information about the scope of stem cell use as well as at the same time, empowering patients with the capacity to make an autonomous decision regarding the use of stem cells. Methods: The Indian Academy of Neurology commissioned an Expert Group Meeting to formulate an advisory to practicing neurologists to counsel patients seeking information and advice about stem cell approaches. Results and Conclusions: In the course of such counselling, it should be emphasized that the information provided by many lay websites might be unsubstantiated. Besides, standard recommendations for the stem cell research, in particular, the application of several layers of oversight should be strictly adhered in order to ensure safety and ethical use of stem cells in neurological disorders.
Collapse
Affiliation(s)
- M Gourie Devi
- Department of Neurology, Institute of Human Behavior and Allied Sciences, New Delhi, India
| | - Alka Sharma
- Department of Biotechnology, Government of India, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Neeraj Jain
- National Brain Research Institute, Gurgaon, Haryana, India
| | - Kusum Verma
- Department of Pathology, Sir Ganga Ram Hospital, New Delhi, India
| | - M Vasantha Padma
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Pramod Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - H S Chabbra
- Indian Spinal Injuries Center, New Delhi, India
| | - Satish Khadilkar
- Department of Neurology, Grant Medical College and Sir JJ Group of Hospitals, Mumbai, Maharashtra, India
| | - Sudesh Prabhakar
- Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gagandeep Singh
- Department of Neurology, Dayanand Medical College, Ludhiana, Punjab, India
| |
Collapse
|
19
|
Muscle Satellite Cells: Exploring the Basic Biology to Rule Them. Stem Cells Int 2016; 2016:1078686. [PMID: 27042182 PMCID: PMC4794588 DOI: 10.1155/2016/1078686] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/24/2016] [Indexed: 12/12/2022] Open
Abstract
Adult skeletal muscle is a postmitotic tissue with an enormous capacity to regenerate upon injury. This is accomplished by resident stem cells, named satellite cells, which were identified more than 50 years ago. Since their discovery, many researchers have been concentrating efforts to answer questions about their origin and role in muscle development, the way they contribute to muscle regeneration, and their potential to cell-based therapies. Satellite cells are maintained in a quiescent state and upon requirement are activated, proliferating, and fusing with other cells to form or repair myofibers. In addition, they are able to self-renew and replenish the stem pool. Every phase of satellite cell activity is highly regulated and orchestrated by many molecules and signaling pathways; the elucidation of players and mechanisms involved in satellite cell biology is of extreme importance, being the first step to expose the crucial points that could be modulated to extract the optimal response from these cells in therapeutic strategies. Here, we review the basic aspects about satellite cells biology and briefly discuss recent findings about therapeutic attempts, trying to raise questions about how basic biology could provide a solid scaffold to more successful use of these cells in clinics.
Collapse
|
20
|
Cao JQ, Liang YY, Li YQ, Zhang HL, Zhu YL, Geng J, Yang LQ, Feng SW, Yang J, Kong J, Zhang C. Adipose-derived stem cells enhance myogenic differentiation in the mdx mouse model of muscular dystrophy via paracrine signaling. Neural Regen Res 2016; 11:1638-1643. [PMID: 27904496 PMCID: PMC5116844 DOI: 10.4103/1673-5374.193244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adipose-derived stem cells have been shown to promote peripheral nerve regeneration through the paracrine secretion of neurotrophic factors. However, it is unclear whether these cells can promote myogenic differentiation in muscular dystrophy. Adipose-derived stem cells (6 × 106) were injected into the gastrocnemius muscle of mdx mice at various sites. Dystrophin expression was found in the muscle fibers. Phosphorylation levels of Akt, mammalian target of rapamycin (mTOR), eIF-4E binding protein 1 and S6 kinase 1 were increased, and the Akt/mTOR pathway was activated. Simultaneously, myogenin levels were increased, whereas cleaved caspase 3 and vimentin levels were decreased. Necrosis and fibrosis were reduced in the muscle fibers. These findings suggest that adipose-derived stem cells promote the regeneration and survival of muscle cells by inhibiting apoptosis and fibrosis, thereby alleviating muscle damage in muscular dystrophy.
Collapse
Affiliation(s)
- Ji-Qing Cao
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Key Laboratory for the Diagnosis and Treatment of Major Neurological Disease, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ying-Yin Liang
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Key Laboratory for the Diagnosis and Treatment of Major Neurological Disease, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ya-Qin Li
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Key Laboratory for the Diagnosis and Treatment of Major Neurological Disease, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hui-Li Zhang
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Key Laboratory for the Diagnosis and Treatment of Major Neurological Disease, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yu-Ling Zhu
- First Affiliated Hospital, Kunming Medical College, Kunming, Yunnan Province, China
| | - Jia Geng
- Department of Neurology, Yantai Yuhuangding Hospital, Yantai, Shandong Province, China
| | - Li-Qing Yang
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shan-Wei Feng
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Juan Yang
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Key Laboratory for the Diagnosis and Treatment of Major Neurological Disease, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jie Kong
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Key Laboratory for the Diagnosis and Treatment of Major Neurological Disease, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Cheng Zhang
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
21
|
Świerczek B, Ciemerych MA, Archacka K. From pluripotency to myogenesis: a multistep process in the dish. J Muscle Res Cell Motil 2015; 36:363-75. [PMID: 26715014 PMCID: PMC4762919 DOI: 10.1007/s10974-015-9436-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells (PSCs), such as embryonic stem cells or induced pluripotent stem cells are a promising source of cells for regenerative medicine as they can differentiate into all cell types building a mammalian body. However, protocols leading to efficient and safe in vitro generation of desired cell types must be perfected before PSCs can be used in cell therapies or tissue engineering. In vivo, i.e. in developing mouse embryo or teratoma, PSCs can differentiate into skeletal muscle, but in vitro their spontaneous differentiation into myogenic cells is inefficient. Numerous attempts have been undertaken to enhance this process. Many of them involved mimicking the interactions occurring during embryonic myogenesis. The key regulators of embryonic myogenesis, such as Wnts proteins, fibroblast growth factor 2, and retinoic acid, have been tested to improve the frequency of in vitro myogenic differentiation of PSCs. This review summarizes the current state of the art, comparing spontaneous and directed myogenic differentiation of PSCs as well as the protocols developed this far to facilitate this process.
Collapse
Affiliation(s)
- Barbara Świerczek
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland.
| |
Collapse
|
22
|
Feisst V, Meidinger S, Locke MB. From bench to bedside: use of human adipose-derived stem cells. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:149-62. [PMID: 26586955 PMCID: PMC4636091 DOI: 10.2147/sccaa.s64373] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since the discovery of adipose-derived stem cells (ASC) in human adipose tissue nearly 15 years ago, significant advances have been made in progressing this promising cell therapy tool from the laboratory bench to bedside usage. Standardization of nomenclature around the different cell types used is finally being adopted, which facilitates comparison of results between research groups. In vitro studies have assessed the ability of ASC to undergo mesenchymal differentiation as well as differentiation along alternate lineages (transdifferentiation). Recently, focus has shifted to the immune modulatory and paracrine effects of transplanted ASC, with growing interest in the ASC secretome as a source of clinical effect. Bedside use of ASC is advancing alongside basic research. An increasing number of safety-focused Phase I and Phase IIb trials have been published without identifying any significant risks or adverse events in the short term. Phase III trials to assess efficacy are currently underway. In many countries, regulatory frameworks are being developed to monitor their use and assure their safety. As many trials rely on ASC injected at a distant site from the area of clinical need, strategies to improve the homing and efficacy of transplanted cells are also being explored. This review highlights each of these aspects of the bench-to-bedside use of ASC and summarizes their clinical utility across a variety of medical specialties.
Collapse
Affiliation(s)
- Vaughan Feisst
- Dunbar Laboratory, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Sarah Meidinger
- Dunbar Laboratory, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Michelle B Locke
- Department of Surgery, Faculty of Medicine and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
23
|
Zhang Y, Zhu Y, Li Y, Cao J, Zhang H, Chen M, Wang L, Zhang C. Long-term engraftment of myogenic progenitors from adipose-derived stem cells and muscle regeneration in dystrophic mice. Hum Mol Genet 2015; 24:6029-40. [DOI: 10.1093/hmg/ddv316] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/31/2015] [Indexed: 12/12/2022] Open
|
24
|
Abstract
Regenerative capacity of skeletal muscles resides in satellite cells, a self-renewing population of muscle cells. Several studies are investigating epigenetic mechanisms that control myogenic proliferation and differentiation to find new approaches that could boost regeneration of endogenous myogenic progenitor populations. In recent years, a lot of effort has been applied to purify, expand and manipulate adult stem cells from muscle tissue. However, this population of endogenous myogenic progenitors in adults is limited and their access is difficult and invasive. Therefore, other sources of stem cells with potential to regenerate muscles need to be examined. An excellent candidate could be a population of adult stromal cells within fat characterized by mesenchymal properties, which have been termed adipose-derived stem cells (ASCs). These progenitor adult stem cells have been successfully differentiated in vitro to osteogenic, chondrogenic, neurogenic and myogenic lineages. Autologous ASCs are multipotent and can be harvested with low morbidity; thus, they hold promise for a range of therapeutic applications. This review will summarize the use of ASCs in muscle regenerative approaches.
Collapse
Affiliation(s)
- Sonia-V Forcales
- Genetics and Epigenetics of Cancer, Institute of Predictive and Personalized Medicine of Cancer Barcelona, Spain
| |
Collapse
|
25
|
Human Mesenchymal Stromal Cells Transplantation May Enhance or Inhibit 4T1 Murine Breast Adenocarcinoma through Different Approaches. Stem Cells Int 2015; 2015:796215. [PMID: 26000020 PMCID: PMC4427122 DOI: 10.1155/2015/796215] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 12/26/2022] Open
Abstract
The use of Mesenchymal Stromal Cells (MSCs) aiming to treat cancer has shown very contradictory results. In an attempt to clarify the contradictory results reported in the literature and the possible role of human fallopian tube Mesenchymal Stromal Cells (htMSCs) against breast cancer, the aim of this study was to evaluate the clinical effect of htMSCs in murine mammary adenocarcinoma using two different approaches: (1) coinjections of htMSCs and 4T1 murine tumor cell lineage and (2) injections of htMSCs in mice at the initial stage of mammary adenocarcinoma development. Coinjected animals had a more severe course of the disease and a reduced survival, while tumor-bearing animals treated with 2 intraperitoneal injections of 106 htMSCs showed significantly reduced tumor growth and increased lifespan as compared with control animals. Coculture of htMSCs and 4T1 tumor cells revealed an increase in IL-8 and MCP-1 and decreased VEGF production. For the first time, we show that MSCs isolated from a single source and donor when injected in the same animal model and tumor can lead to opposite results depending on the experimental protocol. Also, our results demonstrated that htMSCs can have an inhibitory effect on the development of murine mammary adenocarcinoma.
Collapse
|
26
|
Hosseini M, Moghadas M, Edalatmanesh MA, Hashemzadeh MR. Xenotransplantation of human adipose derived mesenchymal stem cells in a rodent model of Huntington's disease: motor and non-motor outcomes. Neurol Res 2015; 37:309-319. [PMID: 25376132 DOI: 10.1179/1743132814y.0000000456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Human mesenchymal stem cells (hMSCs) have been presented as alternative sources of cells to be transplanted into the brain in neurodegenerative disorders. In this regard, the efficacy of hMSCs transplants in reducing motor and non-motor deficits in a quinolinic acid (QA) rat model of Huntington's disease (HD) was tested in the present study. After unilateral lesions in striatum by QA, the isolated and purified hMSCs from liposuction of healthy male donors were transplanted into the damaged striatum of the rats. The cells were stably transfected with a vector containing TurboGFP and JRed to make it possible to trace them after transplantation. Animals were tested by motor and non-motor function tests at different times after the cell transplantation. The hMSCs survived 7 weeks in the brains. An improvement was observed in behavioral tests such as apomophine-induced rotation, hanging wire, and rotarod for the hMSC-treated rats. Anxiety like behaviors were decreased in hMSCs-treated animals when they were examined using open field, elevated plus maze, light and dark box, and novelty suppressed feeding tests. Compared to QA, the hMSCs treatment decreased motor activities. These results confirmed the potential efficacy of hMSCs in treatment of behavioral defects in HD. Generally, the data demonstrated that xenologous transplantation of hMSCs could be considered as an ideal candidate for treatment of neurodegenerative diseases, especially HD.
Collapse
|
27
|
Mesenchymal stromal cells for sphincter regeneration. Adv Drug Deliv Rev 2015; 82-83:123-36. [PMID: 25451135 DOI: 10.1016/j.addr.2014.10.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/29/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023]
Abstract
Stress urinary incontinence (SUI), defined as the involuntary loss of considerable amounts of urine during increased abdominal pressure (exertion, effort, sneezing, coughing, etc.), is a severe problem to the individuals affected and a significant medical, social and economic challenge. SUI is associated with pelvic floor debility, absence of detrusor contraction, or a loss of control over the sphincter muscle apparatus. The pathology includes an increasing loss of muscle cells, replacement of muscular tissue with fibrous tissue, and general aging associated processes of the sphincter complex. When current therapies fail to cure or improve SUI, application of regeneration-competent cells may be an alternative therapeutic option. Here we discuss different aspects of the biology of mesenchymal stromal cells, which are relevant to their clinical applications and for regenerating the sphincter complex. However, there are reports in favor of and against cell-based therapies. We therefore summarize the potential and the risks of cell-based therapies for the treatment of SUI.
Collapse
|
28
|
Indumathi S, Mishra R, Harikrishnan R, Dhanasekaran M. Subcutaneous Adipose Tissue-Derived Stem Cells: Advancement and Applications in Regenerative Medicine. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
29
|
Kim JA, Shon YH, Lim JO, Yoo JJ, Shin HI, Park EK. MYOD mediates skeletal myogenic differentiation of human amniotic fluid stem cells and regeneration of muscle injury. Stem Cell Res Ther 2014; 4:147. [PMID: 24331373 PMCID: PMC4054934 DOI: 10.1186/scrt358] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/18/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022] Open
Abstract
Introduction Human amniotic fluid stem (hAFS) cells have been shown to differentiate into multiple lineages, including myoblasts. However, molecular mechanisms underlying the myogenic differentiation of hAFS cells and their regenerative potential for muscle injury remain to be elucidated. Methods In order to induce myogenic differentiation of hAFS cells, lentiviruses for MYOD were constructed and transduced into hAFS cells. Formation of myotube-like cells was analyzed by immunocytochemistry, and expression of molecular markers for myoblasts was analyzed by reverse transcription polymerase chain reaction and Western blotting. For in vivo muscle regeneration, MYOD transduced hAFS cells were injected into left tibialis anterior (TA) muscles injured with cardiotoxin, and muscle regeneration was analyzed using hematoxylin and eosin, immunocytochemistry and formation of neuro-muscular junction. Results MYOD expression in hAFS cells successfully induced differentiation into multinucleated myotube-like cells. Consistently, significant expression of myogenic marker genes, such as MYOG, DES, DMD and MYH, was induced by MYOD. Analysis of pre-myogenic factors showed that expression of PAX3, MEOX1 and EYA2 was significantly increased by MYOD. MYOD was phosphorylated and localized in the nucleus. These results suggest that in hAFS cells, MYOD is phosphorylated and localized in the nucleus, thus inducing expression of myogenic factors, resulting in myogenic differentiation of hAFS cells. To test regenerative potential of MYOD-transduced hAFS cells, we transplanted them into injured muscles of immunodeficient BALB/cSlc-nu mice. The results showed a substantial increase in the volume of TA muscle injected with MYOD-hAFS cells. In addition, TA muscle tissue injected with MYOD-hAFS cells has more numbers of neuro-muscular junctions compared to controls, indicating functional restoration of muscle injury by MYOD-hAFS cells. Conclusions Collectively, our data suggest that transduction of hAFS cells with MYOD lentiviruses induces skeletal myogenic differentiation in vitro and morphological and functional regeneration of injured muscle in vivo.
Collapse
|
30
|
Uniaxial cyclic strain enhances adipose-derived stem cell fusion with skeletal myocytes. Biochem Biophys Res Commun 2014; 450:1083-8. [DOI: 10.1016/j.bbrc.2014.06.124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 06/24/2014] [Indexed: 12/18/2022]
|
31
|
Ting CH, Ho PJ, Yen BL. Age-related decreases of serum-response factor levels in human mesenchymal stem cells are involved in skeletal muscle differentiation and engraftment capacity. Stem Cells Dev 2014; 23:1206-16. [PMID: 24576136 DOI: 10.1089/scd.2013.0231] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Skeletal muscle (SkM) comprise ∼40% of human body weight. Injury or damage to this important tissue can result in physical disability, and in severe cases is difficult for its endogenous stem cell-the satellite cell-to reverse effectively. Mesenchymal stem cells (MSC) are postnatal progenitor/stem cells that possess multilineage mesodermal differentiation capacity, including toward SkM. Adult bone marrow (BM) is the best-studied source of MSCs; however, aging also decreases BMMSC numbers and can adversely affect differentiation capacity. Therefore, we asked whether human sources of developmentally early stage mesenchymal stem cells (hDE-MSCs) isolated from embryonic stem cells, fetal bone, and term placenta could be cellular sources for SkM repair. Under standard muscle-inducing conditions, hDE-MPCs differentiate toward a SkM lineage rather than cardiomyocytic or smooth muscle lineages, as evidenced by increased expression of SkM-associated markers and in vitro myotube formation. In vivo transplantation revealed that SkM-differentiated hDE-MSCs can efficiently incorporate into host SkM tissue in a mouse model of SkM injury. In contrast, adult BMMSCs do not express SkM-associated genes after in vitro SkM differentiation nor engraft in vivo. Further investigation of possible factors responsible for this difference in SkM differentiation potential revealed that, compared with adult BMMSCs, hDE-MSCs expressed higher levels of serum response factor (SRF), a transcription factor critical for SkM lineage commitment. Moreover, knockdown of SRF in hDE-MSCs resulted in decreased expression of SkM-related genes after in vitro differentiation and decreased in vivo engraftment. Our results implicate SRF as a key factor in age-related SkM differentiation capacity of MSCs, and demonstrate that hDE-MSCs are possible candidates for SkM repair.
Collapse
Affiliation(s)
- Chiao-Hsuan Ting
- 1 Graduate Institute of Life Sciences, National Defense Medical Center , Taipei, Taiwan
| | | | | |
Collapse
|
32
|
Adipose tissue-derived stem cell secreted IGF-1 protects myoblasts from the negative effect of myostatin. BIOMED RESEARCH INTERNATIONAL 2014; 2014:129048. [PMID: 24575400 PMCID: PMC3920898 DOI: 10.1155/2014/129048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 12/03/2013] [Indexed: 11/23/2022]
Abstract
Myostatin, a TGF-β family member, is associated with inhibition of muscle growth and differentiation and might interact with the IGF-1 signaling pathway. Since IGF-1 is secreted at a bioactive level by adipose tissue-derived mesenchymal stem cells (ASCs), these cells (ASCs) provide a therapeutic option for Duchenne Muscular Dystrophy (DMD). But the protective effect of stem cell secreted IGF-1 on myoblast under high level of myostatin remains unclear. In the present study murine myoblasts were exposed to myostatin under presence of ASCs conditioned medium and investigated for proliferation and apoptosis. The protective effect of IGF-1 was further examined by using IGF-1 neutralizing and receptor antibodies as well as gene silencing RNAi technology. MyoD expression was detected to identify impact of IGF-1 on myoblasts differentiation when exposed to myostatin. IGF-1 was accountable for 43.6% of the antiapoptotic impact and 48.8% for the proliferative effect of ASCs conditioned medium. Furthermore, IGF-1 restored mRNA and protein MyoD expression of myoblasts under risk. Beside fusion and transdifferentiation the beneficial effect of ASCs is mediated by paracrine secreted cytokines, particularly IGF-1. The present study underlines the potential of ASCs as a therapeutic option for Duchenne muscular dystrophy and other dystrophic muscle diseases.
Collapse
|
33
|
Rao N, Grover GN, Vincent LG, Evans SC, Choi YS, Spencer KH, Hui EE, Engler AJ, Christman KL. A co-culture device with a tunable stiffness to understand combinatorial cell-cell and cell-matrix interactions. Integr Biol (Camb) 2013; 5:1344-54. [PMID: 24061208 PMCID: PMC3848881 DOI: 10.1039/c3ib40078f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell behavior on 2-D in vitro cultures is continually being improved to better mimic in vivo physiological conditions by combining niche cues including multiple cell types and substrate stiffness, which are well known to impact cell phenotype. However, no system exists in which a user can systematically examine cell behavior on a substrate with a specific stiffness (elastic modulus) in culture with a different cell type, while maintaining distinct cell populations. We demonstrate the modification of a silicon reconfigurable co-culture system with a covalently linked hydrogel of user-defined stiffness. This device allows the user to control whether two separate cell populations are in contact with each other or only experience paracrine interactions on substrates of controllable stiffness. To illustrate the utility of this device, we examined the role of substrate stiffness combined with myoblast co-culture on adipose derived stem cell (ASC) differentiation and found that the presence of myoblasts and a 10 kPa substrate stiffness increased ASC myogenesis versus co-culture on stiff substrates. As this example highlights, this technology better controls the in vitro microenvironment, allowing the user to develop a more thorough understanding of the combined effects of cell-cell and cell-matrix interactions.
Collapse
Affiliation(s)
- Nikhil Rao
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Reza AMMT, Shiwani S, Singh NK, Lohakare JD, Lee SJ, Jeong DK, Han JY, Rengaraj D, Lee BW. Keratinocyte growth factor and thiazolidinediones and linolenic acid differentiate characterized mammary fat pad adipose stem cells isolated from prepubertal Korean black goat to epithelial and adipogenic lineage. In Vitro Cell Dev Biol Anim 2013; 50:194-206. [PMID: 24101555 DOI: 10.1007/s11626-013-9690-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 09/10/2013] [Indexed: 12/25/2022]
Abstract
The study was conducted to know and investigate the mechanism involved during mesenchymal to epithelial transition to unravel questions related to mammary gland development in prepubertal Korean black goat. We, therefore, biopsied mammary fat pad and isolated adipose cells and characterized with stemness factors (CD34, CD13, CD44, CD106, and vimentin) immunologically and through their genetic expression. Furthermore, characterized cells were differentiated to adipogenic (thiazolidinediones and α-linolenic acid) and epithelial (keratinocyte growth factor) lineages. Thiazolidinediones/or in combination with α-linolenic acid demonstrated significant upregulation of adipo-Q, PPAR-γ, CEBP-α, LPL, and resistin. Adipose stem cells in induction mixture (5 μg/ml insulin, 1 μg/ml hydrocortisone, and 10 ng/ml epidermal growth factor) and subsequent treatment with 10 ng/ml keratinocyte growth factor revealed their trans-differentiating ability to epithelial lineage. From 2 d onwards, the cells under keratinocyte growth factor influenced cells to assume rectangular (2-4 d) to cuboidal (8-10 d) shapes. Ayoub-Shklar stain developed brownish-red pigment in the transformed cells. Though, expressions of K8 and K18 were noted to be highly significant (p < 0.01) but expressions of epithelial membrane antigens and epithelial specific antigens were also significant (p < 0.05) compared to 0 d. Conclusively, epithelial transformations of mammary adipose stem cells would add up knowledge to develop therapeutic regimen to deal with mammary tissue injury and diseases.
Collapse
Affiliation(s)
- A M M T Reza
- Department of Animal Biotechnology, College of Animal Life Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bayati V, Altomare L, Tanzi MC, Farè S. Adipose-derived stem cells could sense the nano-scale cues as myogenic-differentiating factors. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:2439-2447. [PMID: 23793565 DOI: 10.1007/s10856-013-4983-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 06/10/2013] [Indexed: 06/02/2023]
Abstract
Microenvironmental cues, such as surface topography and substrate stiffness, may affect stem cells adhesion, morphology, alignment, proliferation and differentiation. Adipose derived stem cells (ASCs) have attracted considerable interest in regenerative medicine due to their easy isolation, extensive in vitro expandability and ability to differentiate along a number of different tissue-specific lineages. The aim of this work was to investigate ASCs adhesion, alignment and differentiation into myogenic lineage on nanofibrous polymeric scaffolds with anisotropic topography. Nanostructured scaffolds with randomized or parallel fibers were fabricated by electrospinning using polycaprolactone (PCL) and the polycarbonate-urethane ChronoFlex AL 80A (CFAL). Cells expressed myosin (fast skeletal) and tropomyosin in all surface topographies 7 days after seeding but myotube formation was only observed on CFAL scaffolds and only few myotubes were formed on PCL scaffolds. The different cell behavior could be ascribed to two main parameters: fibers dimensions and fibers orientation of the substrates that could result in a better myotube formation on CFAL scaffolds.
Collapse
Affiliation(s)
- V Bayati
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | | |
Collapse
|
36
|
Adipose tissue-derived stem cells show considerable promise for regenerative medicine applications. Cell Mol Biol Lett 2013; 18:479-93. [PMID: 23949841 PMCID: PMC6275722 DOI: 10.2478/s11658-013-0101-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 08/09/2013] [Indexed: 01/12/2023] Open
Abstract
The stromal-vascular cell fraction (SVF) of adipose tissue can be an abundant source of both multipotent and pluripotent stem cells, known as adipose-derived stem cells or adipose tissue-derived stromal cells (ADSCs). The SVF also contains vascular cells, targeted progenitor cells, and preadipocytes. Stromal cells isolated from adipose tissue express common surface antigens, show the ability to adhere to plastic, and produce forms that resemble fibroblasts. They are characterized by a high proliferation potential and the ability to differentiate into cells of meso-, ecto- and endodermal origin. Although stem cells obtained from an adult organism have smaller capabilities for differentiation in comparison to embryonic and induced pluripotent stem cells (iPSs), the cost of obtaining them is significantly lower. The 40 years of research that mainly focused on the potential of bone marrow stem cells (BMSCs) revealed a number of negative factors: the painful sampling procedure, frequent complications, and small cell yield. The number of stem cells in adipose tissue is relatively large, and obtaining them is less invasive. Sampling through simple procedures such as liposuction performed under local anesthesia is less painful, ensuring patient comfort. The isolated cells are easily grown in culture, and they retain their properties over many passages. That is why adipose tissue has recently been treated as an attractive alternative source of stem cells. Essential aspects of ADSC biology and their use in regenerative medicine will be analyzed in this article.
Collapse
|
37
|
Systemic delivery of human mesenchymal stromal cells combined with IGF-1 enhances muscle functional recovery in LAMA2 dy/2j dystrophic mice. Stem Cell Rev Rep 2013; 9:93-109. [PMID: 22664740 DOI: 10.1007/s12015-012-9380-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The combination of cell therapy with growth factors could be a useful approach to treat progressive muscular dystrophies. Here, we demonstrate, for the first time, that IGF-1 considerably enhances the myogenesis of human umbilical cord (UC) mesenchymal stromal cells (MSCs) in vitro and that IGF-1 enhances interaction and restoration of dystrophin expression in co-cultures of MSCs and muscle cells from Duchenne patients. In vivo studies showed that human MSCs were able to reach the skeletal muscle of LAMA2(dy/2j) dystrophic mice, through systemic delivery, without immunosuppression. Moreover, we showed, for the first time, that IGF-1 injected systemically together with MSCs markedly reduced muscle inflammation and fibrosis, and significantly improved muscle strength in dystrophic mice. Our results suggest that a combined treatment with IGF-1 and MSCs enhances efficiency of muscle repair and, therefore, should be further considered as a potential therapeutic approach in muscular dystrophies.
Collapse
|
38
|
Sung MS, Mun JY, Kwon O, Kwon KS, Oh DB. Efficient myogenic differentiation of human adipose-derived stem cells by the transduction of engineered MyoD protein. Biochem Biophys Res Commun 2013; 437:156-61. [DOI: 10.1016/j.bbrc.2013.06.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 06/17/2013] [Indexed: 01/17/2023]
|
39
|
Desiderio V, De Francesco F, Schiraldi C, De Rosa A, La Gatta A, Paino F, d'Aquino R, Ferraro GA, Tirino V, Papaccio G. Human Ng2+ adipose stem cells loaded in vivo on a new crosslinked hyaluronic acid-Lys scaffold fabricate a skeletal muscle tissue. J Cell Physiol 2013; 228:1762-73. [PMID: 23359523 DOI: 10.1002/jcp.24336] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/18/2013] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cell (MSC) therapy holds promise for treating diseases and tissue repair. Regeneration of skeletal muscle tissue that is lost during pathological muscle degeneration or after injuries is sustained by the production of new myofibers. Human Adipose stem cells (ASCs) have been reported to regenerate muscle fibers and reconstitute the pericytic cell pool after myogenic differentiation in vitro. Our aim was to evaluate the differentiation potential of constructs made from a new cross-linked hyaluronic acid (XHA) scaffold on which different sorted subpopulations of ASCs were loaded. Thirty days after engraftment in mice, we found that NG2(+) ASCs underwent a complete myogenic differentiation, fabricating a human skeletal muscle tissue, while NG2(-) ASCs merely formed a human adipose tissue. Myogenic differentiation was confirmed by the expression of MyoD, MF20, laminin, and lamin A/C by immunofluorescence and/or RT-PCR. In contrast, adipose differentiation was confirmed by the expression of adiponectin, Glut-4, and PPAR-γ. Both tissues formed expressed Class I HLA, confirming their human origin and excluding any contamination by murine cells. In conclusion, our study provides novel evidence that NG2(+) ASCs loaded on XHA scaffolds are able to fabricate a human skeletal muscle tissue in vivo without the need of a myogenic pre-differentiation step in vitro. We emphasize the translational significance of our findings for human skeletal muscle regeneration.
Collapse
Affiliation(s)
- Vincenzo Desiderio
- Dipartimento di Medicina Sperimentale, Sezione di Istologia ed Embriologia Medica, Tissue Engineering and Regenerative (TERM), Seconda Università degli Studi di Napoli, Napoli, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Evidence for crossing the blood barrier of adult rat brain by human adipose-derived mesenchymal stromal cells during a 6-month period of post-transplantation. Cytotherapy 2013; 15:951-60. [PMID: 23732047 DOI: 10.1016/j.jcyt.2013.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/02/2013] [Accepted: 03/11/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND AIMS Therapeutic promises of adult stem cells have been overshadowed by an elicited immune response, low maintenance of implanted cells or concerns regarding their migration to non-target sites. These problems might be lessened by the use of immune privilege cells and tissues for implantation. METHODS In this study, human adipose-derived mesenchymal stromal cells (hADMSCs) were stably transfected with a vector containing Turbo green fluorescent protein (GFP) and JRed, which allows tracing the cells after transplantation. Labeled hADMSCs were transplanted into the adult rat brain followed by assessment of their survival and migration during 6 months after transplantation. RESULTS Results indicate that there were no postsurgical complications, and the animals thrived after transplantation. The lesions of the surgical process were remarkable at the first weeks, and a high number of transplanted cells were accumulated around them. Cell populations declined over time as they partly migrated away from the injection sites; nonetheless, they were detectable at each examination time point. Although the cells could survive and remain at the injection site for up to 6 months, some of them drifted to spleen, which is an indication of their ability to cross the blood-brain barrier. CONCLUSIONS Despite the high survival rate of hADMSCs in the xenogenic condition, which is an ideal criterion in cell therapy, irregular migration tendency must be handled with caution.
Collapse
|
41
|
Haddad-Mashadrizeh A, Bahrami AR, Matin MM, Edalatmanesh MA, Zomorodipour A, Gardaneh M, Farshchian M, Momeni-Moghaddam M. Human adipose-derived mesenchymal stem cells can survive and integrate into the adult rat eye following xenotransplantation. Xenotransplantation 2013; 20:165-76. [PMID: 23679842 DOI: 10.1111/xen.12033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/18/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Novel threads of discovery provide the basis for optimism for the development of a stem-cell-based strategy for the treatment of retinal blindness. Accordingly, achievement to suitable cell source with potential-to-long-term survival and appropriate differentiation can be an effective step in this direction. METHODS After derivation of human adipose-derived mesenchymal stem cells (HAD-MSCs), they were stably transfected with a vector containing Turbo-green fluorescent protein (GFP) and JRed to be able to trace them after transplantation. Labeled HAD-MSCs were transplanted into the intact adult rat eye and their survival, integration, and migration during 6 months post-transplantation were assessed. RESULTS The transplanted cells were traceable in the rat vitreous humor (VH) up until 90 days after transplantation, with gradual reduction in numbers, their adhesion and expansion capacity after recovery. These cells were also integrated into the ocular tissues. Nonetheless, some of the implanted cells succeeded to cross the blood-retina barrier (BRB) and accumulate in the spleen with time. CONCLUSIONS The survival of the HAD-MSCs for a period of 90 days in VH and even longer period of up to 6 months in other eye tissues makes them a promising source to be considered in regenerative medicine of eye diseases. However, the potency of crossing the BRB by the implanted cells suggests that use of HAD-MSCs must be handled with extreme caution.
Collapse
Affiliation(s)
- Aliakbar Haddad-Mashadrizeh
- Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Tissue engineering and ureter regeneration: is it possible? Int J Artif Organs 2013; 36:392-405. [PMID: 23645581 DOI: 10.5301/ijao.5000130] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2012] [Indexed: 12/11/2022]
Abstract
Large ureter damages are difficult to reconstruct. Current techniques are complicated, difficult to perform, and often associated with failures. The ureter has never been regenerated thus far. Therefore the use of tissue engineering techniques for ureter reconstruction and regeneration seems to be a promising way to resolve these problems. For proper ureter regeneration the following problems must be considered: the physiological aspects of the tissue, the type and shape of the scaffold, the type of cells, and the specific environment (urine).
This review presents tissue engineering achievements in the field of ureter regeneration focusing on the scaffold, the cells, and ureter healing.
Collapse
|
43
|
Indumathi S, Mishra R, Harikrishnan R, Rajkumar JS, Kantawala N, Dhanasekaran M. Lineage depletion of stromal vascular fractions isolated from human adipose tissue: a novel approach towards cell enrichment technology. Cytotechnology 2013; 66:219-28. [PMID: 23553017 DOI: 10.1007/s10616-013-9556-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 03/15/2013] [Indexed: 10/27/2022] Open
Abstract
The therapeutic rationale for tissue repair and regeneration using stem cells is at its infancy and needs advancement in understanding the role of individual component's innate capability. As stem cells of adipose tissue reside in a more heterogeneous population of stromal vascular fractions, cell separation or sorting becomes an eminent step towards revealing their unique properties. This study elucidates the comparative efficacy of lineage depleted adipose derived stromal vascular fraction (SVF) and their innate ability using magnetic activated cell sorter (MACS). To this end, isolated SVF from human adipose tissue was lineage depleted according to the manufacturer's instructions using specific antibody cocktail through MACS. The enriched lineage negative (lin-) and lineage positive (lin+) cell fractions were cultured, phenotypically characterized for the panel of cell surface markers using flowcytometry and subjected to osteoblastic and adipogenic differentiation. The expression profile obtained for lin- cells was CD34-/CD45-/HLADR-/CD49d-/CD140b-/CD31-/CD90+/CD105+/CD73+/CD54+/CD166+/CD117- when compared to Lin+ cells expressing CD34+/CD45+/HLADR-/CD49d-/CD140b+/CD31-/CD90+/CD105+/CD73+/CD54+/CD166+/CD117+ (CD-cluster of differentiation). These results, thus, advances our understanding on the inherent property of the individual cell population. Furthermore, both the fractions exhibited mesodermal lineage differentiation capacity. To conclude, this research pursuit rationalized the regenerative therapeutic applicability of both lin- and lin+ cultures of human adipose tissue for disorders of mesodermal, haematological and vascular origin.
Collapse
Affiliation(s)
- S Indumathi
- Department of Stem Cells, Lifeline Multispeciality Hospital, Chennai, 600 096, India
| | | | | | | | | | | |
Collapse
|
44
|
Human adipose tissue stem cells: relevance in the pathophysiology of obesity and metabolic diseases and therapeutic applications. Expert Rev Mol Med 2012; 14:e19. [PMID: 23302474 DOI: 10.1017/erm.2012.13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells are unique cells exhibiting self-renewing properties and the potential to differentiate into multiple specialised cell types. Totipotent or pluripotent stem cells are generally abundant in embryonic or fetal tissues, but the use of discarded embryos as sources of these cells raises challenging ethical problems. Adult stem cells can also differentiate into a wide variety of cell types. In particular, adult adipose tissue contains a pool of abundant and accessible multipotent stem cells, designated as adipose-derived stem cells (ASCs), that are able to replicate as undifferentiated cells, to develop as mature adipocytes and to differentiate into multiple other cell types along the mesenchymal lineage, including chondrocytes, myocytes and osteocytes, and also into cells of endodermal and neuroectodermal origin, including beta-cells and neurons, respectively. An impairment in the differentiation potential and biological functions of ASCs may contribute to the development of obesity and related comorbidities. In this review, we summarise different aspects of the ASCs with special reference to the isolation and characterisation of these cell populations, their relation to the biochemical features of the adipose tissue depot of origin and to the metabolic characteristics of the donor subject and discuss some prospective therapeutic applications.
Collapse
|
45
|
Local injections of adipose-derived mesenchymal stem cells modulate inflammation and increase angiogenesis ameliorating the dystrophic phenotype in dystrophin-deficient skeletal muscle. Stem Cell Rev Rep 2012; 8:363-74. [PMID: 21874281 DOI: 10.1007/s12015-011-9304-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The effects of adipose-derived mesenchymal stem cells (ADMSC) transplantation on degeneration, regeneration and skeletal muscle function were investigated in dystrophin-deficient mice (24-week-old). ADMSC transplantation improved muscle strength and, resistance to fatigue. An increase in fiber cross-sectional area and in the number of fibers with centralized nuclei and augment of myogenin content were observed. In ADMSC-treated muscles a decrease in muscle content of TNF-α, IL-6 and oxidative stress measured by Amplex(®) reagent were observed. The level of TGF-β1 was lowered whereas that of VEGF, IL-10 and IL-4 were increased by ADMSC treatment. An increase in markers of macrophage M1 (CD11 and F4-80) and a decrease in T lymphocyte marker (CD3) and arginase-1 were also observed in ADMSCs-treated dystrophic muscle. No change was observed in iNOS expression. Increased phosphorylation of Akt, p70S6k and 4E-BP1 was found in dystrophic muscles treated with ADMSC. These results suggest that ADMSC transplantation modulates inflammation and improves muscle tissue regeneration, ameliorating the dystrophic phenotype in dystrophin-deficient mice.
Collapse
|
46
|
Rodríguez-Jiménez FJ, Valdes-Sánchez T, Carrillo JM, Rubio M, Monleon-Prades M, García-Cruz DM, García M, Cugat R, Moreno-Manzano V. Platelet-rich plasma favors proliferation of canine adipose-derived mesenchymal stem cells in methacrylate-endcapped caprolactone porous scaffold niches. J Funct Biomater 2012; 3:556-68. [PMID: 24955632 PMCID: PMC4030998 DOI: 10.3390/jfb3030556] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 12/19/2022] Open
Abstract
Osteoarticular pathologies very often require an implementation therapy to favor regeneration processes of bone, cartilage and/or tendons. Clinical approaches performed on osteoarticular complications in dogs constitute an ideal model for human clinical translational applications. The adipose-derived mesenchymal stem cells (ASCs) have already been used to accelerate and facilitate the regenerative process. ASCs can be maintained in vitro and they can be differentiated to osteocytes or chondrocytes offering a good tool for cell replacement therapies in human and veterinary medicine. Although ACSs can be easily obtained from adipose tissue, the amplification process is usually performed by a time consuming process of successive passages. In this work, we use canine ASCs obtained by using a Bioreactor device under GMP cell culture conditions that produces a minimum of 30 million cells within 2 weeks. This method provides a rapid and aseptic method for production of sufficient stem cells with potential further use in clinical applications. We show that plasma rich in growth factors (PRGF) treatment positively contributes to viability and proliferation of canine ASCs into caprolactone 2-(methacryloyloxy) ethyl ester (CLMA) scaffolds. This biomaterial does not need additional modifications for cASCs attachment and proliferation. Here we propose a framework based on a combination of approaches that may contribute to increase the therapeutical capability of stem cells by the use of PRGF and compatible biomaterials for bone and connective tissue regeneration.
Collapse
Affiliation(s)
| | - Teresa Valdes-Sánchez
- Neuronal Regeneration Lab, Centro de Investigación Principe Felipe, València 46012, Spain.
| | - José M Carrillo
- Medicine and Surgery Department, CEU-Cardenal Herrera University, Moncada 46115, Spain.
| | - Mónica Rubio
- Medicine and Surgery Department, CEU-Cardenal Herrera University, Moncada 46115, Spain.
| | - Manuel Monleon-Prades
- Centre for Biomaterials and Tissue Enginering, Universitat Politècnica de València, València E-46022, Spain.
| | - Dunia Mercedes García-Cruz
- Centre for Biomaterials and Tissue Enginering, Universitat Politècnica de València, València E-46022, Spain.
| | | | - Ramón Cugat
- Fundación García Cugat, Barcelona 08006, Spain.
| | | |
Collapse
|
47
|
Grabowska I, Brzoska E, Gawrysiak A, Streminska W, Moraczewski J, Polanski Z, Hoser G, Kawiak J, Machaj EK, Pojda Z, Ciemerych MA. Restricted Myogenic Potential of Mesenchymal Stromal Cells Isolated from Umbilical Cord. Cell Transplant 2012; 21:1711-26. [DOI: 10.3727/096368912x640493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Nonhematopoietic cord blood cells and mesenchymal cells of umbilical cord Wharton's jelly have been shown to be able to differentiate into various cell types. Thus, as they are readily available and do not raise any ethical issues, these cells are considered to be a potential source of material that can be used in regenerative medicine. In our previous study, we tested the potential of whole mononucleated fraction of human umbilical cord blood cells and showed that they are able to participate in the regeneration of injured mouse skeletal muscle. In the current study, we focused at the umbilical cord mesenchymal stromal cells isolated from Wharton's jelly. We documented that limited fraction of these cells express markers of pluripotent and myogenic cells. Moreover, they are able to undergo myogenic differentiation in vitro, as proved by coculture with C2C12 myoblasts. They also colonize injured skeletal muscle and, with low frequency, participate in the formation of new muscle fibers. Pretreatment of Wharton's jelly mesenchymal stromal cells with SDF-1 has no impact on their incorporation into regenerating muscle fibers but significantly increased muscle mass. As a result, transplantation of mesenchymal stromal cells enhances the skeletal muscle regeneration.
Collapse
Affiliation(s)
- Iwona Grabowska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Agnieszka Gawrysiak
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Wladyslawa Streminska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Jerzy Moraczewski
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Zbigniew Polanski
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grazyna Hoser
- Department of Clinical Cytology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Jerzy Kawiak
- Department of Clinical Cytology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Eugeniusz K. Machaj
- Department of Cellular Engineering, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Zygmunt Pojda
- Department of Cellular Engineering, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Maria A. Ciemerych
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
48
|
Haghighipour N, Heidarian S, Shokrgozar MA, Amirizadeh N. Differential effects of cyclic uniaxial stretch on human mesenchymal stem cell into skeletal muscle cell. Cell Biol Int 2012; 36:669-675. [PMID: 22681392 DOI: 10.1042/cbi20110400] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Both fetal and adult skeletal muscle cells are continually being subjected to biomechanical forces. Biomechanical stimulation during cell growth affects proliferation, differentiation and maturation of skeletal muscle cells. Bone marrow-derived hMSCs [human MSCs (mesenchymal stem cells)] can differentiate into a variety of cell types, including skeletal muscle cells that are potentially a source for muscle regeneration. Our investigations involved a 10% cyclic uniaxial strain at 1 Hz being applied to hMSCs grown on collagen-coated silicon membranes with or without IGF-I (insulin-like growth factor-I) for 24 h. Results obtained from morphological studies confirmed the rearrangement of cells after loading. Comparison of MyoD and MyoG mRNA levels between test groups showed that mechanical loading alone can initiate myogenic differentiation. Furthermore, comparison of Myf5, MyoD, MyoG and Myf6 mRNA levels between test groups showed that a combination of mechanical loading and growth factor results in the highest expression of myogenic genes. These results indicate that cyclic strain may be useful in myogenic differentiation of stem cells, and can accelerate the differentiation of hMSCs into MSCs in the presence of growth factor.
Collapse
Affiliation(s)
- Nooshin Haghighipour
- National Cell Bank of Iran, Pasteur Institute of Iran, National Cell Bank of Iran, Tehran, Iran
| | | | | | | |
Collapse
|
49
|
Jazedje T, Bueno DF, Almada BVP, Caetano H, Czeresnia CE, Perin PM, Halpern S, Maluf M, Evangelista LP, Nisenbaum MG, Martins MT, Passos-Bueno MR, Zatz M. Human fallopian tube mesenchymal stromal cells enhance bone regeneration in a xenotransplanted model. Stem Cell Rev Rep 2012; 8:355-62. [PMID: 21744049 PMCID: PMC3362709 DOI: 10.1007/s12015-011-9297-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We have recently reported that human fallopian tubes, which are discarded during surgical procedures of women submitted to sterilization or hysterectomies, are a rich source of human fallopian tube mesenchymal stromal cells (htMSCs). It has been previously shown that human mesenchymal stromal cells may be useful in enhancing the speed of bone regeneration. This prompted us to investigate whether htMSCs might be useful for the treatment of osteoporosis or other bone diseases, since they present a pronounced capacity for osteogenic differentiation in vitro. Based on this prior knowledge, our aim was to evaluate, in vivo, the osteogenic capacity of htMSCs to regenerate bone through an already described xenotransplantation model: nonimmunosuppressed (NIS) rats with cranial defects. htMSCs were obtained from five 30-50 years old healthy women and characterized by flow cytometry and for their multipotenciality in vitro capacity (osteogenic, chondrogenic and adipogenic differentiations). Two symmetric full-thickness cranial defects on each parietal region of seven NIS rats were performed. The left side (LS) of six animals was covered with CellCeram (Scaffdex)-a bioabsorbable ceramic composite scaffold that contains 60% hydroxyapatite and 40% β-tricalciumphosphate-only, and the right side (RS) with the CellCeram and htMSCs (10(6) cells/scaffold). The animals were euthanized at 30, 60 and 90 days postoperatively and cranial tissue samples were taken for histological analysis. After 90 days we observed neobone formation in both sides. However, in animals euthanized 30 and 60 days after the procedure, a mature bone was observed only on the side with htMSCs. PCR and immunofluorescence analysis confirmed the presence of human DNA and thus that human cells were not rejected, which further supports the imunomodulatory property of htMSCs. In conclusion, htMSCs can be used successfully to enhance bone regeneration in vivo, opening a new field for future treatments of osteoporosis and bone reconstruction.
Collapse
Affiliation(s)
- Tatiana Jazedje
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Daniela F. Bueno
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Bruno V. P. Almada
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Heloisa Caetano
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | | | - Paulo M. Perin
- CEERH Specialized Center for Human Reproduction, São Paulo, Brazil
| | | | - Mariangela Maluf
- CEERH Specialized Center for Human Reproduction, São Paulo, Brazil
| | | | | | - Marília T. Martins
- Department of Oral Pathology, University of São Paulo, São Paulo, Brazil
| | - Maria R. Passos-Bueno
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mayana Zatz
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
50
|
Mattioli M, Gloria A, Turriani M, Berardinelli P, Russo V, Nardinocchi D, Curini V, Baratta M, Martignani E, Barboni B. Osteo-regenerative potential of ovarian granulosa cells: an in vitro and in vivo study. Theriogenology 2012; 77:1425-37. [PMID: 22284224 DOI: 10.1016/j.theriogenology.2011.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 11/05/2011] [Accepted: 11/13/2011] [Indexed: 11/26/2022]
Abstract
Granulosa cells (GC) express stemness markers and can differentiate into cell types not present within the follicles. Given that follicles at different stages of development populate the ovary, we undertook this research in the pig model to identify the stage of follicle, growing or luteinizing, from which GC with the best regenerative potential can be retrieved. Growing follicles were isolated from prepubertal gilts 50 h after equine chorionic gonadotropin (eCG) (1,200 IU) administration. Luteinizing follicles were obtained from prepubertal gilts treated with eCG (1,200 IU) followed, 60 h later, by hCG (500 IU). The follicles were isolated 30 h after hCG. The GC isolated from growing (GGC) and from luteinizing (LGC) follicles were expanded in vitro for three passages and exposed to osteogenic medium to trigger differentiation. The GC incorporated in PLGA scaffolds were cultured in osteogenic medium for 2 wks and then implanted subcutaneously in the dorsal region of SCID mice to assess their osteogenic potential in vivo. In addition to the typical granulosa cells characteristics (inhibin, progesterone and estrogen production and FSH receptors), GGC and LGC showed a diffused expression of the stemness markers Sox2, Nanog and TERT immediately after isolation. Expansion caused in both cell types a rapid disappearance of granulosa cell characters while it did not modify stemness marker expression. Osteogenic medium induced a marked extracellular matrix mineralization and alkaline phosphatase activation in LGC, clearly detectable after two wks, while the process was much lighter in GGC, where it became evident after 3 wks. Osteocalcin and Runx2 expressions were upregulated and stemness markers downregulated by osteogenic medium. The GC loaded implants, retrieved 8 wks after transplantation, had viable GC surrounding the several nodules of calcifications recorded. Similar effects were induced by GGC and LGC while calcification nodules were not recorded when scaffolds without cells were implanted. These data confirm that GC, expanded in vitro undergo progressive de-differentiation retaining their plasticity and demonstrate that both GGC and LGC have osteogenic potential, luteinizing cells being more efficient. Transplanted in SCID mice, GC participate in new bone formation, thus confirming their therapeutic potential.
Collapse
Affiliation(s)
- M Mattioli
- Department of Comparative Biomedical Sciences, University of Teramo, 64100 Teramo, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|