51
|
Sá AL, Sampaio RV, da Costa Almeida NN, Sangalli JR, Brito KNL, Bressan FF, Rissino JD, do Socorro Damasceno Santos S, Meirelles FV, Ohashi OM, dos Santos Miranda M. Effect of POU5F1 Expression Level in Clonal Subpopulations of Bovine Fibroblasts Used as Nuclear Donors for Somatic Cell Nuclear Transfer. Cell Reprogram 2017; 19:294-301. [DOI: 10.1089/cell.2016.0063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- André Luiz Sá
- Laboratório de Fecundação In Vitro, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Rafael V. Sampaio
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | | | - Juliano Rodrigues Sangalli
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - Karynne Nazaré Lins Brito
- Laboratório de Fecundação In Vitro, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Fabiana Fernandes Bressan
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - Joirge Dores Rissino
- Laboratório de Citogenética, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | | | - Flavio Vieira Meirelles
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - Otávio Mitio Ohashi
- Laboratório de Fecundação In Vitro, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Moysés dos Santos Miranda
- Laboratório de Fecundação In Vitro, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| |
Collapse
|
52
|
Dias LD, Casali KR, Ghem C, da Silva MK, Sausen G, Palma PB, Covas DT, Kalil RAK, Schaan BD, Nardi NB, Markoski MM. Mesenchymal stem cells from sternum: the type of heart disease, ischemic or valvular, does not influence the cell culture establishment and growth kinetics. J Transl Med 2017; 15:161. [PMID: 28743269 PMCID: PMC5526254 DOI: 10.1186/s12967-017-1262-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/11/2017] [Indexed: 12/24/2022] Open
Abstract
Background In an attempt to increase the therapeutic potential for myocardial regeneration, there is a quest for new cell sources and types for cell therapy protocols. The pathophysiology of heart diseases may affect cellular characteristics and therapeutic results. Methods To study the proliferative and differentiation potential of mesenchymal stem cells (MSC), isolated from bone marrow (BM) of sternum, we made a comparative analysis between samples of patients with ischemic (IHD) or non-ischemic valvular (VHD) heart diseases. We included patients with IHD (n = 42) or VHD (n = 20), with average age of 60 years and no differences in cardiovascular risk factors. BM samples were collected (16.4 ± 6 mL) and submitted to centrifugation with Ficoll-Paque, yielding 4.5 ± 1.5 × 107 cells/mL. Results Morphology, immunophenotype and differentiation ability had proven that the cultivated sternal BM cells had MSC features. The colony forming unit-fibroblast (CFU-F) frequency was similar between groups (p = 0.510), but VHD samples showed positive correlation to plated cells vs. CFU-F number (r = 0.499, p = 0.049). The MSC culture was established in 29% of collected samples, achieved passage 9, without significant difference in expansion kinetics between groups (p > 0.05). Dyslipidemia and the use of statins was associated with culture establishment for IHD patients (p = 0.049 and p = 0.006, respectively). Conclusions Together, these results show that the sternum bone can be used as a source for MSC isolation, and that ischemic or valvular diseases do not influence the cellular yield, culture establishment or in vitro growth kinetics. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1262-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lucinara Dadda Dias
- Programa de Pós-graduação em Ciências da Saúde-Cardiologia, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Avenida Princesa Isabel, n° 370, 3° andar, Porto Alegre, RS, CEP: 90620-001, Brazil
| | | | - Carine Ghem
- Serviço de Patologia Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Melissa Kristocheck da Silva
- Programa de Pós-graduação em Ciências da Saúde-Cardiologia, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Avenida Princesa Isabel, n° 370, 3° andar, Porto Alegre, RS, CEP: 90620-001, Brazil
| | - Grasiele Sausen
- Programa de Pós-graduação em Ciências da Saúde-Cardiologia, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Avenida Princesa Isabel, n° 370, 3° andar, Porto Alegre, RS, CEP: 90620-001, Brazil
| | - Patrícia Bonini Palma
- Laboratório de Citometria de Fluxo, Centro Regional de Hemoterapia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto/Universidade de São Paulo, São Paulo, SP, Brazil
| | - Dimas Tadeu Covas
- Laboratório de Citometria de Fluxo, Centro Regional de Hemoterapia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto/Universidade de São Paulo, São Paulo, SP, Brazil
| | - Renato A K Kalil
- Programa de Pós-graduação em Ciências da Saúde-Cardiologia, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Avenida Princesa Isabel, n° 370, 3° andar, Porto Alegre, RS, CEP: 90620-001, Brazil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Beatriz D Schaan
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Nance Beyer Nardi
- Programa de Pós-graduação em Ciências da Saúde-Cardiologia, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Avenida Princesa Isabel, n° 370, 3° andar, Porto Alegre, RS, CEP: 90620-001, Brazil.,Laboratório de Células-Tronco e Engenharia de Tecidos, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Melissa Medeiros Markoski
- Programa de Pós-graduação em Ciências da Saúde-Cardiologia, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Avenida Princesa Isabel, n° 370, 3° andar, Porto Alegre, RS, CEP: 90620-001, Brazil. .,Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
53
|
Abstract
For decades, researchers have been fascinated by the strategy of using cell therapy for bone defects; some progress in the field has been made. Owing to its ample supply and easy access, skin, the largest organ in the body, has gained attention as a potential source of stem cells. Despite extensive applications in skin and nerve regeneration, an increasing number of reports indicate its potential use in bone tissue engineering and regeneration. Unfortunately, few review articles are available to outline current research efforts in skin-based osteogenesis. This review first summarizes the latest findings on stem cells or progenitors in skin and their niches and then discusses the strategies of skin cell-based osteogenesis. We hope this article elucidates this topic and generates new ideas for future studies.
Collapse
Affiliation(s)
- Tingliang Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA.,Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA.,Division of Exercise Physiology, West Virginia University, Morgantown, WV, USA.,Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
54
|
Dermal Fibroblasts Promote Alternative Macrophage Activation Improving Impaired Wound Healing. J Invest Dermatol 2017; 137:941-950. [DOI: 10.1016/j.jid.2016.11.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/28/2016] [Accepted: 11/27/2016] [Indexed: 02/06/2023]
|
55
|
Roson-Burgo B, Sanchez-Guijo F, Del Cañizo C, De Las Rivas J. Insights into the human mesenchymal stromal/stem cell identity through integrative transcriptomic profiling. BMC Genomics 2016; 17:944. [PMID: 27871224 PMCID: PMC5117530 DOI: 10.1186/s12864-016-3230-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 11/01/2016] [Indexed: 01/10/2023] Open
Abstract
Background Mesenchymal Stromal/Stem Cells (MSCs), isolated under the criteria established by the ISCT, still have a poorly characterized phenotype that is difficult to distinguish from similar cell populations. Although the field of transcriptomics and functional genomics has quickly grown in the last decade, a deep comparative analysis of human MSCs expression profiles in a meaningful cellular context has not been yet performed. There is also a need to find a well-defined MSCs gene-signature because many recent biomedical studies show that key cellular interaction processes (i.e. inmuno-modulation, cellular cross-talk, cellular maintenance, differentiation, epithelial-mesenchymal transition) are dependent on the mesenchymal stem cells within the stromal niche. Results In this work we define a core mesenchymal lineage signature of 489 genes based on a deep comparative analysis of multiple transcriptomic expression data series that comprise: (i) MSCs of different tissue origins; (ii) MSCs in different states of commitment; (iii) other related non-mesenchymal human cell types. The work integrates several public datasets, as well as de-novo produced microarray and RNA-Seq datasets. The results present tissue-specific signatures for adipose tissue, chorionic placenta, and bone marrow MSCs, as well as for dermal fibroblasts; providing a better definition of the relationship between fibroblasts and MSCs. Finally, novel CD marker patterns and cytokine-receptor profiles are unravelled, especially for BM-MSCs; with MCAM (CD146) revealed as a prevalent marker in this subtype of MSCs. Conclusions The improved biomolecular characterization and the released genome-wide expression signatures of human MSCs provide a comprehensive new resource that can drive further functional studies and redesigned cell therapy applications. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3230-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Beatriz Roson-Burgo
- Bioinformatics and Functional Genomics Group, Cancer Research Center (IBMCC, CSIC/USAL) and IBSAL, Consejo Superior de Investigaciones Cientificas (CSIC), Salamanca, Spain.,Hematology Department, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Fermin Sanchez-Guijo
- Hematology Department, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Consuelo Del Cañizo
- Hematology Department, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (IBMCC, CSIC/USAL) and IBSAL, Consejo Superior de Investigaciones Cientificas (CSIC), Salamanca, Spain.
| |
Collapse
|
56
|
Dermal-epidermal membrane systems by using human keratinocytes and mesenchymal stem cells isolated from dermis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 71:943-953. [PMID: 27987793 DOI: 10.1016/j.msec.2016.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/20/2016] [Accepted: 11/02/2016] [Indexed: 02/04/2023]
Abstract
Dermal-epidermal membrane systems were developed by co-culturing human keratinocytes with Skin derived Stem Cells (SSCs), which are Mesenchymal Stem Cells (MSCs) isolated from dermis, on biodegradable membranes of chitosan (CHT), polycaprolactone (PCL) and a polymeric blend of CHT and PCL. The membranes display physico-chemical, morphological, mechanical and biodegradation properties that could satisfy and fulfil specific requirements in skin tissue engineering. CHT membrane exhibits an optimal biodegradation rate for acute wounds; CHT-PCL for the chronic ones. On the other hand, PCL membrane in spite of its very slow biodegradation rate exhibits mechanical properties similar to in vivo dermis, a lower hydrophilic character, and a surface roughness, all properties that make it able to sustain cell adhesion and proliferation for in vitro skin models. Both CHT-PCL and PCL membranes guided epidermal and dermal differentiation of SSCs as pointed out by the expression of cytokeratins and the deposition of the ECM protein fibronectin, respectively. In the dermal-epidermal membrane systems, a more suitable microenvironment for the SSCs differentiation was promoted by the interactions and the mutual interplay with keratinocytes. Being skin tissue-biased stem cells committed to their specific final dermal and/or epidermal cell differentiation, SSCs are more suitable for skin tissue engineering than other adult MSCs with different origin. For this reason, they represent a useful autologous cell source for engineering skin substitutes for both in vivo and in vitro applications.
Collapse
|
57
|
He JG, Wang TL, Wang T, Xu H, Zhang Y, Dong JS. The Immunologic Properties of Bone Morphogenic Protein Receptor IB Positive Subpopulation before and after Osteogenic Differentiation in Mouse Dermis. PLoS One 2016; 11:e0161785. [PMID: 27552226 PMCID: PMC4995052 DOI: 10.1371/journal.pone.0161785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/11/2016] [Indexed: 11/30/2022] Open
Abstract
We have previously reported that human dermal bone morphogenic protein receptor (BMPR) IB positive subpopulation had a high osteogenic differentiation potential and may be a promising cell source for allogeneic bone tissue engineering. In this study, the immunologic properties of dermal BMPR-IB+ subpopulation before and after osteogenic differentiation were reported. The results confirmed that dermal BMPR-IB+ cells possessed a similar osteogenic differentiation potential with bone marrow mesenchymal stromal cells in a mouse model. Furthermore, the expression of immune rejection-related surface antigens such as major histocompatibility class II and co-stimulatory proteins (CD40, CD80, and CD86) were absent on dermal BMPRIB+ cells. Dermal BMPRIB+ cells elicited no proliferation of allogeneic splenocytes and suppressed the proliferation of stimulated immune cells. Interestingly, osteogenic differentiation in vitro had no adverse effect on the immunological features of these cells. Most importantly, inducible NO synthase (iNOS) was involved in immunoregulatory effects by undifferentiated BMPRIB+ fibroblasts, whereas indoleamine 2,3-dioxygenase (IDO) activity was related to mediating immunomodulatory function by osteogenic differentiated BMPRIB+ fibroblasts. In conclusion, dermal BMPRIB+ cells have a low immunogenicity and possess immunosuppressive capacity before and after osteogenic differentiation in vitro, which would facilitate the allotransplantation in the future. However, mechanisms mediating immunoregulatory property between undifferentiated and osteogenic differentiated BMPRIB+ fibroblasts may be different and need further investigation.
Collapse
Affiliation(s)
- Jin-Guang He
- Department of Plastic and Reconstructive Surgery, Shanghai ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ting-Liang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Tao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Hua Xu
- Department of Plastic and Reconstructive Surgery, Shanghai ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jia-Sheng Dong
- Department of Plastic and Reconstructive Surgery, Shanghai ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
58
|
Vezzani B, Pierantozzi E, Sorrentino V. Not All Pericytes Are Born Equal: Pericytes from Human Adult Tissues Present Different Differentiation Properties. Stem Cells Dev 2016; 25:1549-1558. [PMID: 27549576 DOI: 10.1089/scd.2016.0177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pericytes (PCs) have been recognized for a long time only as structural cells of the blood vessels. The identification of tight contacts with endothelial cells and the ability to interact with surrounding cells through paracrine signaling revealed additional functions of PCs in maintaining the homeostasis of the perivascular environment. PCs got the front page, in the late 1990s, after the identification and characterization of a new embryonic cell population, the mesoangioblasts, from which PCs present in the adult organism are thought to derive. From these studies, it was clear that PCs were also endowed with multipotent mesodermal abilities. Furthermore, their ability to cross the vascular wall and to reconstitute skeletal muscle tissue after systemic injection opened the way to a number of studies aimed to develop therapeutic protocols for a cell therapy of muscular dystrophy. This has resulted in a major effort to characterize pericytic cell populations from skeletal muscle and other adult tissues. Additional studies also addressed their relationship with other cells of the perivascular compartment and with mesenchymal stem cells. These data have provided initial evidence that PCs from different adult tissues might be endowed with distinctive differentiation abilities. This would suggest that the multipotent mesenchymal ability of PCs might be restrained within different tissues, likely depending on the specific cell renewal and repair requirements of each tissue. This review presents current knowledge on human PCs and highlights recent data on the differentiation properties of PCs isolated from different adult tissues.
Collapse
Affiliation(s)
- Bianca Vezzani
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| |
Collapse
|
59
|
Wang T, He J, Zhang Y, Shi W, Dong J, Pei M, Zhu L. A Selective Cell Population from Dermis Strengthens Bone Regeneration. Stem Cells Transl Med 2016; 6:306-315. [PMID: 28170187 PMCID: PMC5442747 DOI: 10.5966/sctm.2015-0426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/22/2016] [Indexed: 11/16/2022] Open
Abstract
Finding appropriate seed cells for bone tissue engineering remains a significant challenge. Considering that skin is the largest organ, we hypothesized that human bone morphogenetic protein receptor type IB (BmprIB)+ dermal cells could have enhanced osteogenic capacity in the healing of critical-sized calvarial defects in an immunodeficient mouse model. In this study, immunohistochemical staining revealed that BmprIB was expressed throughout reticular dermal cells; the positive expression rate of BmprIB was 3.5% ± 0.4% in freshly separated dermal cells, by flow cytometry. Furthermore, in vitro osteogenic capacity of BmprIB+ cells was confirmed by osteogenic-related staining and marker gene expression compared with unsorted dermal cells. In vivo osteogenic capacity was demonstrated by implantation of human BmprIB+ cell/coral constructs in the treatment of 4-mm diameter calvarial defects in an immunodeficient mouse model compared with implantation of unsorted cell/coral constructs and coral scaffold alone. These results indicate that the selective cell population BmprIB from human dermis is a promising osteogenic progenitor cell that can be a large-quantity and high-quality cell source for bone tissue engineering and regeneration. Stem Cells Translational Medicine 2017;6:306-315.
Collapse
Affiliation(s)
- Tingliang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jinguang He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wenjun Shi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiasheng Dong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, and Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia, USA
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
60
|
Lee SB, Shim S, Kim MJ, Shin HY, Jang WS, Lee SJ, Jin YW, Lee SS, Park S. Identification of a distinct subpopulation of fibroblasts from murine dermis: CD73(-) CD105(+) as potential marker of dermal fibroblasts subset with multipotency. Cell Biol Int 2016; 40:1008-16. [PMID: 27170595 DOI: 10.1002/cbin.10623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/07/2016] [Indexed: 01/22/2023]
Abstract
Skin dermis includes various types of multipotent stromal cells (MSCs) and a subpopulation of dermal fibroblasts that exhibit the ability to differentiate. However, characterization of this dermal fibroblast subtype remains less understood. In this study, we isolated dermal cells from the skin of newborn C57/B6 mice and investigated their characteristics. Isolated murine dermal cells exhibited a fibroblast phenotype as judged by accepted criteria including a lack of MSC-related antigens and the differentiation potential of MSCs, and the positive expression of fibroblast markers. A comparative analysis demonstrated that CD73(-) CD105(+) but not CD73(-) CD105(-) dermal fibroblasts exhibited some of the functional properties of MSCs. Furthermore, the multipotent phenotype of CD73(-) CD105(+) cells was diminished by treatment of CD105 siRNA and shRNA, indicating that CD105 expression was critical for the retention of differentiation potential of those cells. Overall, these results suggest that CD73(-) CD105(+) cells are a distinct subset of dermal fibroblasts with multipotency and that their surface antigens could help to classify this subpopulation. These cells may contribute to the regeneration of damaged tissue.
Collapse
Affiliation(s)
- Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Min-Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Hye-Yun Shin
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Young-Woo Jin
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Seung-Sook Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, 75 Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| |
Collapse
|
61
|
Jiang Y, Shi Y, He J, Zhang Z, Zhou G, Zhang W, Cao Y, Liu W. Enhanced tenogenic differentiation and tendon-like tissue formation by tenomodulin overexpression in murine mesenchymal stem cells. J Tissue Eng Regen Med 2016; 11:2525-2536. [PMID: 27098985 DOI: 10.1002/term.2150] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/12/2015] [Accepted: 12/22/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Yongkang Jiang
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
| | - Yuan Shi
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
| | - Jing He
- Department of Anatomy and Neurobiology; Tongji University School of Medicine; Shanghai People's Republic of China
| | - Zhiyong Zhang
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
- National Tissue Engineering Centre of China; Shanghai People's Republic of China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
- National Tissue Engineering Centre of China; Shanghai People's Republic of China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
- National Tissue Engineering Centre of China; Shanghai People's Republic of China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
- National Tissue Engineering Centre of China; Shanghai People's Republic of China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery shanghai 9th People's Hospital; People's Republic of China
- National Tissue Engineering Centre of China; Shanghai People's Republic of China
| |
Collapse
|
62
|
Kwon H, Paschos NK, Hu JC, Athanasiou K. Articular cartilage tissue engineering: the role of signaling molecules. Cell Mol Life Sci 2016; 73:1173-94. [PMID: 26811234 PMCID: PMC5435375 DOI: 10.1007/s00018-015-2115-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/23/2015] [Accepted: 12/10/2015] [Indexed: 02/08/2023]
Abstract
Effective early disease modifying options for osteoarthritis remain lacking. Tissue engineering approach to generate cartilage in vitro has emerged as a promising option for articular cartilage repair and regeneration. Signaling molecules and matrix modifying agents, derived from knowledge of cartilage development and homeostasis, have been used as biochemical stimuli toward cartilage tissue engineering and have led to improvements in the functionality of engineered cartilage. Clinical translation of neocartilage faces challenges, such as phenotypic instability of the engineered cartilage, poor integration, inflammation, and catabolic factors in the arthritic environment; these can all contribute to failure of implanted neocartilage. A comprehensive understanding of signaling molecules involved in osteoarthritis pathogenesis and their actions on engineered cartilage will be crucial. Thus, while it is important to continue deriving inspiration from cartilage development and homeostasis, it has become increasingly necessary to incorporate knowledge from osteoarthritis pathogenesis into cartilage tissue engineering.
Collapse
Affiliation(s)
- Heenam Kwon
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Nikolaos K Paschos
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kyriacos Athanasiou
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA.
| |
Collapse
|
63
|
Hiraoka C, Toki F, Shiraishi K, Sayama K, Nishimura EK, Miura H, Higashiyama S, Nanba D. Two clonal types of human skin fibroblasts with different potentials for proliferation and tissue remodeling ability. J Dermatol Sci 2016; 82:84-94. [PMID: 26867959 DOI: 10.1016/j.jdermsci.2016.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/05/2016] [Accepted: 01/25/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Skin fibroblast heterogeneity is of growing interest due to its relevance in not only skin development but also cutaneous wound healing. However, the characterization of human dermal fibroblasts at a clonal level has not been accomplished and their functional heterogeneity remains poorly understood. OBJECTIVE The aim of this study was to define the clonal heterogeneity of human dermal fibroblasts. METHODS Isolated human dermal fibroblasts were clonally expanded and categorized by comprehensive phenotypic and gene expression profiling. RESULTS Single fibroblasts were significantly multiplied and efficiently cloned without chromosomal abnormalities under hypoxic conditions. Individual clones were heterogeneous in their proliferative capacity, and gene expression profiling revealed differences in the expression of genes involved in extracellular matrix synthesis and degradation. Each cloned fibroblast also had different abilities in terms of collagen remodeling. All phenotypic and gene expression data were analyzed with Spearman's rank correlation, and fibroblasts were categorized into at least two functional clonal types. One was highly proliferative, while the other was less proliferative but had the ability to remodel the tissue architecture. The proliferative clones were predominant in infants, but decreased with physiological aging. CONCLUSION This study provides strong evidence for the functional heterogeneity of human dermal fibroblasts at a clonal level, which has implications regarding skin repair and aging.
Collapse
Affiliation(s)
- Chihiro Hiraoka
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan; Department of Bone and Joint Surgery, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan
| | - Fujio Toki
- Department of Biochemistry and Molecular Genetics, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan; Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ken Shiraishi
- Department of Dermatology, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan
| | - Koji Sayama
- Department of Dermatology, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan
| | - Emi K Nishimura
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hiromasa Miura
- Department of Bone and Joint Surgery, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan
| | - Daisuke Nanba
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Graduate School of Medicine, Ehime University, Toon, Ehime 791-0295, Japan; Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
64
|
Mesenchymal Stromal Cells and Tissue-Specific Progenitor Cells: Their Role in Tissue Homeostasis. Stem Cells Int 2015; 2016:4285215. [PMID: 26823669 PMCID: PMC4707334 DOI: 10.1155/2016/4285215] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/15/2015] [Indexed: 02/07/2023] Open
Abstract
Multipotent mesenchymal stromal/stem cells (MSCs) reside in many human organs and comprise heterogeneous population of cells with self-renewal ability. These cells can be isolated from different tissues, and their morphology, immunophenotype, and differentiation potential are dependent on their tissue of origin. Each organ contains specific population of stromal cells which maintain regeneration process of the tissue where they reside, but some of them have much more wide plasticity and differentiate into multiple cells lineage. MSCs isolated from adult human tissues are ideal candidates for tissue regeneration and tissue engineering. However, MSCs do not only contribute to structurally tissue repair but also MSC possess strong immunomodulatory and anti-inflammatory properties and may influence in tissue repair by modulation of local environment. This paper is presenting an overview of the current knowledge of biology of tissue-resident mesenchymal stromal and progenitor cells (originated from bone marrow, liver, skeletal muscle, skin, heart, and lung) associated with tissue regeneration and tissue homeostasis.
Collapse
|
65
|
Uchida H, Morita T, Niizuma K, Kushida Y, Kuroda Y, Wakao S, Sakata H, Matsuzaka Y, Mushiake H, Tominaga T, Borlongan CV, Dezawa M. Transplantation of Unique Subpopulation of Fibroblasts, Muse Cells, Ameliorates Experimental Stroke Possibly via Robust Neuronal Differentiation. Stem Cells 2015; 34:160-73. [DOI: 10.1002/stem.2206] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/30/2015] [Accepted: 07/12/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Hiroki Uchida
- Department of Stem Cell Biology and Histology
- Department of Neurosurgery
| | - Takahiro Morita
- Department of Stem Cell Biology and Histology
- Department of Neurosurgery
| | | | | | | | | | | | - Yoshiya Matsuzaka
- Department of Physiology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Hajime Mushiake
- Department of Physiology; Tohoku University Graduate School of Medicine; Sendai Japan
| | | | - Cesario V. Borlongan
- Department of Neurosurgery and Brain Repair; University of South Florida College of Medicine; Tampa Florida USA
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology
- Department of Anatomy and Anthropology
| |
Collapse
|
66
|
Alkaline phosphatase expression/activity and multilineage differentiation potential are the differences between fibroblasts and orbital fat-derived stem cells--a study in animal serum-free culture conditions. Stem Cell Rev Rep 2015; 10:697-711. [PMID: 24913281 DOI: 10.1007/s12015-014-9529-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human orbital fat tissues are a potential source to isolate stem cells for the development of regenerative medicine therapies. For future safe clinical application of these cells, it is critical to establish animal component-free culture conditions as well as to clearly define the stem cell population characteristics differentiating them from other cell types, such as fibroblasts. Therefore, the present study aimed to compare phenotypic and functional characteristics of orbital fat-derived stem cells (OFSCs) and fibroblasts resident in the eyelid skin in donor-matched samples grown in culture medium supplemented with pooled allogeneic human serum (HS) replacing fetal bovine serum (FBS). We first investigated the proliferative effects of OFSCs on HS, and then we compared the alkaline phosphatase (AP) expression and activity, immunophenotypic profile, and in vitro multilineage differentiation potential of OFSCs side-by-side with fibroblasts. The results showed that HS enhanced OFSCs proliferation without compromising their immunophenotype, AP activity, and osteogenic, adipogenic, and chondrogenic differentiation capacities. In contrast to OFSCs, the fibroblasts did not exhibit AP expression and activity and did not have multilineage differentiation potential. The results enabled us to successfully distinguish OFSCs from fibroblasts populations, suggesting that AP expression/activity and multilineage differentiation assays can be used reliably to discriminate mesenchymal stem cells from fibroblasts. Our findings also support the feasibility of pooled allogeneic HS as a safer and more effective alternative to FBS for clinical applications.
Collapse
|
67
|
Gaspar D, Spanoudes K, Holladay C, Pandit A, Zeugolis D. Progress in cell-based therapies for tendon repair. Adv Drug Deliv Rev 2015; 84:240-56. [PMID: 25543005 DOI: 10.1016/j.addr.2014.11.023] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/08/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
The last decade has seen significant developments in cell therapies, based on permanently differentiated, reprogrammed or engineered stem cells, for tendon injuries and degenerative conditions. In vitro studies assess the influence of biophysical, biochemical and biological signals on tenogenic phenotype maintenance and/or differentiation towards tenogenic lineage. However, the ideal culture environment has yet to be identified due to the lack of standardised experimental setup and readout system. Bone marrow mesenchymal stem cells and tenocytes/dermal fibroblasts appear to be the cell populations of choice for clinical translation in equine and human patients respectively based on circumstantial, rather than on hard evidence. Collaborative, inter- and multi-disciplinary efforts are expected to provide clinically relevant and commercially viable cell-based therapies for tendon repair and regeneration in the years to come.
Collapse
Affiliation(s)
- Diana Gaspar
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Kyriakos Spanoudes
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Carolyn Holladay
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Dimitrios Zeugolis
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
68
|
Johnstone SA, Liley M, Dalby MJ, Barnett SC. Comparison of human olfactory and skeletal MSCs using osteogenic nanotopography to demonstrate bone-specific bioactivity of the surfaces. Acta Biomater 2015; 13:266-76. [PMID: 25463488 DOI: 10.1016/j.actbio.2014.11.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/15/2014] [Accepted: 11/13/2014] [Indexed: 01/14/2023]
Abstract
Recently we identified a novel population of mesenchymal stem cells (MSCs) from human olfactory mucosa (OM-MSCs), a tissue which promotes neurogenesis throughout life, and demonstrated that they promoted CNS myelination to a greater extent than bone marrow-derived (BM)-MSCs. Previous data demonstrated that nanotopographies with a degree of disorder induce BM-MSC osteogenic differentiation. Thus, using biomaterials as non-chemical tools, we investigated if MSCs from a completely different cellular niche could be induced to differentiate similarly to nanoscale cues alone. Both MSCs differentiated into bone when cultured on nanotopographically embossed polycaprolactone (PCL) with a disordered pattern and heights but not on a "smooth" non-embossed PCL control substrate, but OM-MSC changes were at lower expression levels. Both MSCs showed similar increases in differentiation markers at the protein and mRNA level when plated on the two patterned surfaces. Thus, topographical cues from substrates with disordered patterns can up-regulate several MSC resident genes in both BM-MSCs and OM-MSCs. Moreover, antibody purified BM-MSCs had similar properties to non-purified BM-MSCs. These data suggest that MSCs from a neural cellular niche express similar bone-induced cues to BM-MSCs, suggesting that MSCs that inherently support nervous tissue can differentiate along the bone lineage in a similar manner to MSCs from a skeletal environment.
Collapse
|
69
|
Mohamad-Fauzi N, Ross PJ, Maga EA, Murray JD. Impact of source tissue and ex vivo expansion on the characterization of goat mesenchymal stem cells. J Anim Sci Biotechnol 2015; 6:1. [PMID: 25838897 PMCID: PMC4382838 DOI: 10.1186/2049-1891-6-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 12/18/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND There is considerable interest in using goats as models for genetically engineering dairy animals and also for using stem cells as therapeutics for bone and cartilage repair. Mesenchymal stem cells (MSCs) have been isolated and characterized from various species, but are poorly characterized in goats. RESULTS Goat MSCs isolated from bone marrow (BM-MSCs) and adipose tissue (ASCs) have the ability to undergo osteogenic, adipogenic and chondrogenic differentiation. Cytochemical staining and gene expression analysis show that ASCs have a greater capacity for adipogenic differentiation compared to BM-MSCs and fibroblasts. Different methods of inducing adipogenesis also affect the extent and profile of adipogenic differentiation in MSCs. Goat fibroblasts were not capable of osteogenesis, hence distinguishing them from the MSCs. Goat MSCs and fibroblasts express CD90, CD105, CD73 but not CD45, and exhibit cytoplasmic localization of OCT4 protein. Goat MSCs can be stably transfected by Nucleofection, but, as evidenced by colony-forming efficiency (CFE), yield significantly different levels of progenitor cells that are robust enough to proliferate into colonies of integrants following G418 selection. BM-MSCs expanded over increasing passages in vitro maintained karyotypic stability up to 20 passages in culture, exhibited an increase in adipogenic differentiation and CFE, but showed altered morphology and amenability to genetic modification by selection. CONCLUSIONS Our findings provide characterization information on goat MSCs, and show that there can be significant differences between MSCs isolated from different tissues and from within the same tissue. Fibroblasts do not exhibit trilineage differentiation potential at the same capacity as MSCs, making it a more reliable method for distinguishing MSCs from fibroblasts, compared to cell surface marker expression.
Collapse
Affiliation(s)
- Nuradilla Mohamad-Fauzi
- Department of Animal Science, University of California, Davis, California 95616 USA ; Institute of Ocean and Earth Sciences, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Pablo J Ross
- Department of Animal Science, University of California, Davis, California 95616 USA
| | - Elizabeth A Maga
- Department of Animal Science, University of California, Davis, California 95616 USA
| | - James D Murray
- Department of Animal Science, University of California, Davis, California 95616 USA ; Department of Population Health and Reproduction, University of California, Davis, California 95616 USA
| |
Collapse
|
70
|
Lv FJ, Tuan RS, Cheung KMC, Leung VYL. Concise review: the surface markers and identity of human mesenchymal stem cells. Stem Cells 2015; 32:1408-19. [PMID: 24578244 DOI: 10.1002/stem.1681] [Citation(s) in RCA: 742] [Impact Index Per Article: 74.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 02/09/2014] [Indexed: 12/13/2022]
Abstract
The concept of mesenchymal stem cells (MSCs) is becoming increasingly obscure due to the recent findings of heterogeneous populations with different levels of stemness within MSCs isolated by traditional plastic adherence. MSCs were originally identified in bone marrow and later detected in many other tissues. Currently, no cloning based on single surface marker is capable of isolating cells that satisfy the minimal criteria of MSCs from various tissue environments. Markers that associate with the stemness of MSCs await to be elucidated. A number of candidate MSC surface markers or markers possibly related to their stemness have been brought forward so far, including Stro-1, SSEA-4, CD271, and CD146, yet there is a large difference in their expression in various sources of MSCs. The exact identity of MSCs in vivo is not yet clear, although reports have suggested they may have a fibroblastic or pericytic origin. In this review, we revisit the reported expression of surface molecules in MSCs from various sources, aiming to assess their potential as MSC markers and define the critical panel for future investigation. We also discuss the relationship of MSCs to fibroblasts and pericytes in an attempt to shed light on their identity in vivo.
Collapse
Affiliation(s)
- Feng-Juan Lv
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China; Stem Cell & Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong SAR, People's Republic of China; Center for Reproduction, Development and Growth, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | |
Collapse
|
71
|
Chen F, Zhang X, Bi D, Xia L, Lin Y, Zhang W, Liu W, Cao Y. RETRACTED ARTICLE: Screening research on membrane protein of dermal stem/progenitor cells with different differentiation potential. Cytotechnology 2015; 67:175. [PMID: 23475157 PMCID: PMC4294848 DOI: 10.1007/s10616-012-9475-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 06/08/2012] [Indexed: 10/27/2022] Open
Affiliation(s)
- Fuguo Chen
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Xiaoping Zhang
- />Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 People’s Republic of China
| | - Dan Bi
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Linlin Xia
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Yang Lin
- />Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 People’s Republic of China
| | - Wenjie Zhang
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Wei Liu
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Yilin Cao
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| |
Collapse
|
72
|
Levin S, Pevsner-Fischer M, Kagan S, Lifshitz H, Weinstock A, Gataulin D, Friedlander G, Zipori D. Divergent levels of LBP and TGFβ1 in murine MSCs lead to heterogenic response to TLR and proinflammatory cytokine activation. Stem Cell Rev Rep 2014; 10:376-88. [PMID: 24664302 DOI: 10.1007/s12015-014-9498-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The outstanding heterogeneity of stem cell populations is a major obstacle on the way to their clinical application. It is therefore paramount to identify the molecular mechanisms that underlay this heterogeneity. Individually derived bone marrow mesenchymal stromal cells (MSCs) preparations, studied here, diverged markedly in various properties, despite of being all tripotent in their differentiation potential. Microarray analysis showed that MSC diversity is evident also in highly variable gene expression patterns. Differentially expressed genes were significantly enriched in toll-like receptors (TLRs) and differentiation pathways. Marked differences were observed in LPS binding protein (LBP) and transforming growth factor (TGF)β1 expression. These differences correlated with MSC functionality. Therefore, the possible contribution of these molecules to MSC diversity was examined. In the TLR signaling pathway, LBP levels predicted the ability of specific MSCs to secrete interleukin (IL)-6 in response to LPS. A relatively higher expression of TGFβ1 endowed MSCs with a capacity to respond to IL-1β by reduced osteogenic differentiation. This study thus demonstrates major diversity within MSC isolates, which appears early on following derivation and persists following long-term culture. MSC heterogeneity results from highly variable transcriptome. Differential expression of LBP and TGFβ1, along with other genes, in different MSC preparations, produces the variable responses to external stimuli.
Collapse
Affiliation(s)
- Sarit Levin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel, 76100
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Chen Z, Wang Y, Shi C. Therapeutic Implications of Newly Identified Stem Cell Populations From the Skin Dermis. Cell Transplant 2014; 24:1405-22. [PMID: 24972091 DOI: 10.3727/096368914x682431] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Skin, the largest organ of the body, is a promising reservoir for adult stem cells. The epidermal stem cells and hair follicle stem cells have been well studied for their important roles in homeostasis, regeneration, and repair of the epidermis and appendages for decades. However, stem cells residing in dermis were not identified until the year 2001, when a variety of stem cell subpopulations have been isolated and identified from the dermis of mammalian skin such as neural crest stem cells, mesenchymal stem cell-like dermal stem cells, and dermal hematopoietic cells. These stem cell subpopulations exhibited capabilities of self-renewing, multipotent differentiating, and immunosuppressive properties. Hence, the dermis-derived stem cells showed extensive potential applications in regenerative medicine, especially for wound healing/tissue repair, neural repair, and hematopoietic recovery. Here we summarized current research on the stem cell subpopulations derived from the dermis and aimed to provide a comprehensive review on their isolation, specific markers, differentiation capacity, and the functional activities in homeostasis, regeneration, and tissue repair.
Collapse
Affiliation(s)
- Zelin Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
74
|
Gago-Lopez N, Awaji O, Zhang Y, Ko C, Nsair A, Liem D, Stempien-Otero A, MacLellan W. THY-1 receptor expression differentiates cardiosphere-derived cells with divergent cardiogenic differentiation potential. Stem Cell Reports 2014; 2:576-91. [PMID: 24936447 PMCID: PMC4050474 DOI: 10.1016/j.stemcr.2014.03.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 01/26/2023] Open
Abstract
Despite over a decade of intense research, the identity and differentiation potential of human adult cardiac progenitor cells (aCPC) remains controversial. Cardiospheres have been proposed as a means to expand aCPCs in vitro, but the identity of the progenitor cell within these 3D structures is unknown. We show that clones derived from cardiospheres could be subdivided based on expression of thymocyte differentiation antigen 1 (THY-1/CD90) into two distinct populations that exhibit divergent cardiac differentiation potential. One population, which is CD90(+), expressed markers consistent with a mesenchymal/myofibroblast cell. The second clone type was CD90(-) and could form mature, functional myocytes with sarcomeres albeit at a very low rate. These two populations of cardiogenic clones displayed distinct cell surface markers and unique transcriptomes. Our study suggests that a rare aCPC exists in cardiospheres along with a mesenchymal/myofibroblast cell, which demonstrates incomplete cardiac myocyte differentiation.
Collapse
Affiliation(s)
- Nuria Gago-Lopez
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98195-6422, USA
- Department of Medicine and Physiology, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Obinna Awaji
- Department of Medicine and Physiology, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Yiqiang Zhang
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98195-6422, USA
| | - Christopher Ko
- Department of Medicine and Physiology, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Ali Nsair
- Department of Medicine and Physiology, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - David Liem
- Department of Medicine and Physiology, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - April Stempien-Otero
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98195-6422, USA
| | - W. Robb MacLellan
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98195-6422, USA
- Department of Medicine and Physiology, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| |
Collapse
|
75
|
Kan L, Kessler JA. Evaluation of the cellular origins of heterotopic ossification. Orthopedics 2014; 37:329-40. [PMID: 24810815 DOI: 10.3928/01477447-20140430-07] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 11/22/2013] [Indexed: 02/03/2023]
Abstract
Heterotopic ossification (HO), acquired or hereditary, is featured by the formation of bone outside of the normal skeleton. Typical acquired HO is a common, debilitating condition associated with traumatic events. Cardiovascular calcification, an atypical form of acquired HO, is prevalent and associated with high rates of cardiovascular mortality. Hereditary HO syndromes, such as fibrodysplasia ossificans progressiva and progressive osseous heteroplasia, are rare, progressive, life-threatening disorders. The cellular origins of HO remain elusive. Some bona fide contributing cell populations have been found through genetic lineage tracing and other experiments in vivo, and various other candidate populations have been proposed. Nevertheless, because of the difficulties in establishing cellular phenotypes in vivo and other confounding factors, the true identities of these populations are still uncertain. This review critically evaluates the accumulating data in the field. The major focus is on the candidate populations that may give rise to osteochondrogenic lineage cells directly, not the populations that may contribute to HO indirectly. This issue is important not solely because of the clinical implications, but also because it highlights the basic biological processes that govern bone formation.
Collapse
|
76
|
Golpour M, Fattahi S, Niaki HA, Hadipoor A, Abedian Z, Ahangarian GR, Parsian H, Mosapour A, Khorasani HR, Vaziri HR, Bijani A, Mostafazadeh A. Starved human fibroblasts secrete acidic proteins inducing post re-feeding proliferation and in vitro cell migration: a potential tool for wound healing. Biol Cell 2014; 106:139-50. [PMID: 24612410 DOI: 10.1111/boc.201300063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/18/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND INFORMATION There are several reports indicating that starved fibroblasts show higher proliferation rates when re-fed with foetal bovine serum. We have evidence demonstrating that this phenomenon is related to secretory proteins which may be beneficial to wound healing. RESULTS After re-feeding, 16 and 72 h serum-starved fibroblasts showed the highest and lowest proliferation rates, 1.59 and 0.51-fold difference compared to the non-starved control, respectively (P < 0.05). However, the latest value could be normalised by incubating cells with 16 h-starved fibroblast cell culture supernatant (16-SFS), prior to re-feeding. A strong correlation was found between total protein level in starved fibroblast culture supernatants and post re-feeding proliferation rates (r(2) = 0.90, P < 0.001). Two-dimensional gel electrophoresis analysis of 16-SFS confirmed the presence of proteins with relative molecular weights of 10-120 kDa and pI ranging from 4 to 6. A significant difference in calcium influx course was found between 16-SFS and the negative control (Dulbecco's Modified Eagle Medium) (P < 0.05). There was no significant difference in Ca(2+) concentrations after 1 h between non-starved controls and 16-SFS-treated fibroblasts. The scratch test demonstrated that the 16-SFS is able to induce fibroblast migration. CONCLUSIONS We concluded that human starved fibroblasts secrete proteins that are able to induce post re-feeding cell proliferation and fibroblasts migration, probably through the induction of a sustained calcium influx. This is worth being considered as a potential tool for wound healing.
Collapse
Affiliation(s)
- Monireh Golpour
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran; University of Guilan, Rasht, Iran
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Feisst V, Brooks AES, Chen CJJ, Dunbar PR. Characterization of mesenchymal progenitor cell populations directly derived from human dermis. Stem Cells Dev 2014; 23:631-42. [PMID: 24325341 DOI: 10.1089/scd.2013.0207] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cell (MSC) and progenitor cell (MPC) populations in human dermis remain poorly characterized, despite their importance to wound repair and the pathogenesis of many skin diseases. To identify MSC/MPC populations in human dermis we developed an 11-marker flow cytometry technique that enabled sorting of mesenchymal cell populations for functional assays, using adipose-derived stem cells (ASCs) from human adipose tissue as a positive control. Two populations of dermal cells had similar phenotypes to ASCs: both were CD34(+) CD73(+) CD105(-)/low, and lacked expression of c-kit (CD117) and hematopoietic or vascular markers (CD31, CD45, CD146, and HLA-DR). However, whereas ASCs were CD36(+/-) CD90(+), dermal mesenchymal progenitor cells (DMPCs) were split between a dominant CD36(-) CD90(+) population (DMPC1) and a small CD36(+) CD90(-) population (DMPC2). Both these populations were capable of differentiating into adipocytes, but only DMPC1 localized to a perivascular location, similar to that reported for ASCs. Re-gating of the flow cytometry data revealed that both DMPC1 and DMPC2 were part of CD45(-) CD73(+) CD146(-) populations with variable expression of CD34. This suggests that CD34 may not be a stable marker of DMPC populations in human dermis, consistent with data from MSCs in human bone marrow, and with the loss of CD34 we observed from both ASCs and DMPCs on cell culture. These data enable future study of DMPCs in health and disease, and may also explain why some mesenchymal cell lines derived from human dermis exhibit characteristics of MSCs.
Collapse
Affiliation(s)
- Vaughan Feisst
- School of Biological Sciences and Maurice Wilkins Centre, University of Auckland , Auckland, New Zealand
| | | | | | | |
Collapse
|
78
|
Hong S, Alapure BV, Lu Y, Tian H, Wang Q. Immunohistological localization of endogenous unlabeled stem cells in wounded skin. J Histochem Cytochem 2014; 62:276-85. [PMID: 24399040 DOI: 10.1369/0022155414520710] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Various types of endogenous stem cells (SCs) participate in wound healing in the skin at different anatomical locations. SCs need to be identified through multiple markers, and this is usually performed using flow cytometry. However, immunohistological identification of endogenous stem cells in the skin at different anatomical locations by co-staining multiple SC markers has been seldom explored. We examined the immunohistological localization of four major types of SCs in wounded skin by co-staining for their multiple markers. Hematopoietic SCs were co-stained for Sca1 and CD45; mesenchymal SCs for Sca1, CD29, and CD106; adipose SCs for CD34, CD90, and CD105; and endothelial progenitor cells and their differentiated counterparts were co-stained for CD34, Tie2, and von Willebrand factor. We found Sca1(+)CD45(+) SCs in the epidermis, dermis and hypodermis of wounded skin. Sca1(+)CD29(+) and Sca1(+)CD106(+) mesenchymal SCs, CD34(+)CD105(+), CD34(+)CD90(+), and CD90(+)CD105(+) adipose SCs, as well as CD34(+)Tie2(+) endothelial progenitor cells were also located in the epidermis, dermis, and hypodermis. This study demonstrates the feasibility of using immunohistological staining to determine the location of SCs in wounded skin and the intracellular distribution of their molecular markers.
Collapse
Affiliation(s)
- Song Hong
- Center of Neuroscience Excellence, Louisiana State University Health Science Center, New Orleans, Louisiana
| | | | | | | | | |
Collapse
|
79
|
Human dermal stem/progenitor cell-derived conditioned medium ameliorates ultraviolet a-induced damage of normal human dermal fibroblasts. PLoS One 2013; 8:e67604. [PMID: 23874431 PMCID: PMC3708938 DOI: 10.1371/journal.pone.0067604] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/07/2013] [Indexed: 12/31/2022] Open
Abstract
Adult skin stem cells are considered an attractive cell resource for therapeutic potential in aged skin. We previously reported that multipotent human dermal stem/progenitor cells (hDSPCs) can be enriched from (normal human dermal fibroblasts (NHDFs) using collagen type IV. However, the beneficial effects of hDSPCs on aged skin remain to be elucidated. In the present study, we analyzed the growth factors secreted from hDSPCs in conditioned medium (CM) derived from hDSPCs (hDSPC-CM) and found that hDSPCs secreted higher levels of bFGF, IGFBP-1, IGFBP-2, HGF, VEGF and IGF-1 compared with non-hDSPCs. We then investigated whether hDSPC-CM has an effect on ultraviolet A (UVA)-irradiated NHDFs. Real-time RT-PCR analysis revealed that the treatment of UVA-irradiated NHDFs with hDSPC-CM significantly antagonized the UVA-induced up-regulation of the MMP1 and the UVA-induced down-regulation of the collagen types I, IV and V and TIMP1 mRNA expressions. Furthermore, a scratch wound healing assay showed that hDSPC-CM enhanced the migratory properties of UVA-irradiated NHDFs. hDSPC-CM also significantly reduced the number of the early and late apoptotic cell population in UVA-irradiated NHDFs. Taken together, these data suggest that hDSPC-CM can exert some beneficial effects on aged skin and may be used as a therapeutic agent to improve skin regeneration and wound healing.
Collapse
|
80
|
Brohem CA, de Carvalho CM, Radoski CL, Santi FC, Baptista MC, Swinka BB, de A. Urban C, de Araujo LRR, Graf RM, Feferman IHS, Lorencini M. Comparison between fibroblasts and mesenchymal stem cells derived from dermal and adipose tissue. Int J Cosmet Sci 2013; 35:448-57. [DOI: 10.1111/ics.12064] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/28/2013] [Indexed: 12/11/2022]
Affiliation(s)
- C. A. Brohem
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | | | - C. L. Radoski
- Department of Biotechnology; Positivo University; Curitiba; Paraná; Brazil
| | - F. C. Santi
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - M. C. Baptista
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - B. B. Swinka
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - C. de A. Urban
- Department of Biotechnology; Positivo University; Curitiba; Paraná; Brazil
| | | | - R. M. Graf
- Department of Plastic Surgery; Federal University of Paraná; Curitiba; Paraná; Brazil
| | - I. H. S. Feferman
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - M. Lorencini
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| |
Collapse
|
81
|
Bussmann BM, Reiche S, Marí-Buyé N, Castells-Sala C, Meisel HJ, Semino CE. Chondrogenic potential of human dermal fibroblasts in a contractile, soft, self-assembling, peptide hydrogel. J Tissue Eng Regen Med 2013; 10:E54-62. [PMID: 23737099 DOI: 10.1002/term.1766] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 02/14/2013] [Accepted: 04/13/2013] [Indexed: 12/30/2022]
Abstract
The present paper describes a simple approach to obtain three-dimensional (3D) cartilage constructs using human normal dermal fibroblasts (hNDFs) cultured in a self-assembling peptide nanofibre scaffold. During the first days of culture, the 3D constructs underwent morphological changes consisting of a substantial contraction process that ended in a small compact structure. During this process the system became sensitive to induction with standard chondrogenic medium, evidenced by the expression of specific markers of mature cartilage. First, it was detected that the samples become highly stained with toluidine blue dye over time (40-50 days), indicating that the system produced significantly high amounts of glycosaminoglycans. By quantitative PCR, it was confirmed that the system significantly upregulated the expression of the proteoglycan aggrecan, a good indicator of cartilage commitment. Moreover, collagen type II was upregulated at protein level, confirming that the system differentiated to a chondrocyte-like construct. Additionally, during the first days of culture in control medium analysed hNDFs proliferation capacity in this 3D system was analysed. This platform could be used in the future to obtain an autologous source of cells from a simple patient skin biopsy, which could be easily translated into a low-cost and effective regenerative therapy.
Collapse
Affiliation(s)
- Bianca M Bussmann
- Translational Centre for Regenerative Medicine (TRM-Leipzig), Universität Leipzig, Germany
| | - Sven Reiche
- Translational Centre for Regenerative Medicine (TRM-Leipzig), Universität Leipzig, Germany
| | - Núria Marí-Buyé
- Department of Bioengineering, Tissue Engineering Laboratory, Institut Químic de Sarrià -Universitat Ramon Llull, Barcelona, Spain
| | - Cristina Castells-Sala
- Department of Bioengineering, Tissue Engineering Laboratory, Institut Químic de Sarrià -Universitat Ramon Llull, Barcelona, Spain
| | - Hans Jörg Meisel
- Department of Neurosurgery, BG Hospital Bergmannstrost, Halle, Germany
| | - Carlos E Semino
- Translational Centre for Regenerative Medicine (TRM-Leipzig), Universität Leipzig, Germany.,Department of Bioengineering, Tissue Engineering Laboratory, Institut Químic de Sarrià -Universitat Ramon Llull, Barcelona, Spain
| |
Collapse
|
82
|
Calloni R, Cordero EAA, Henriques JAP, Bonatto D. Reviewing and updating the major molecular markers for stem cells. Stem Cells Dev 2013; 22:1455-76. [PMID: 23336433 PMCID: PMC3629778 DOI: 10.1089/scd.2012.0637] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Stem cells (SC) are able to self-renew and to differentiate into many types of committed cells, making SCs interesting for cellular therapy. However, the pool of SCs in vivo and in vitro consists of a mix of cells at several stages of differentiation, making it difficult to obtain a homogeneous population of SCs for research. Therefore, it is important to isolate and characterize unambiguous molecular markers that can be applied to SCs. Here, we review classical and new candidate molecular markers that have been established to show a molecular profile for human embryonic stem cells (hESCs), mesenchymal stem cells (MSCs), and hematopoietic stem cells (HSCs). The commonly cited markers for embryonic ESCs are Nanog, Oct-4, Sox-2, Rex-1, Dnmt3b, Lin-28, Tdgf1, FoxD3, Tert, Utf-1, Gal, Cx43, Gdf3, Gtcm1, Terf1, Terf2, Lefty A, and Lefty B. MSCs are primarily identified by the expression of CD13, CD29, CD44, CD49e, CD54, CD71, CD73, CD90, CD105, CD106, CD166, and HLA-ABC and lack CD14, CD31, CD34, CD45, CD62E, CD62L, CD62P, and HLA-DR expression. HSCs are mainly isolated based on the expression of CD34, but the combination of this marker with CD133 and CD90, together with a lack of CD38 and other lineage markers, provides the most homogeneous pool of SCs. Here, we present new and alternative markers for SCs, along with microRNA profiles, for these cells.
Collapse
Affiliation(s)
- Raquel Calloni
- Departamento de Biologia Molecular e Biotecnologia, Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | | | | | | |
Collapse
|
83
|
Khosrotehrani K. Mesenchymal stem cell therapy in skin: why and what for? Exp Dermatol 2013; 22:307-10. [DOI: 10.1111/exd.12141] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2013] [Indexed: 12/24/2022]
Affiliation(s)
- Kiarash Khosrotehrani
- UQ Centre for Clinical Research; The University of Queensland; Brisbane QLD Australia
| |
Collapse
|
84
|
Hilmi ABM, Halim AS, Hassan A, Lim CK, Noorsal K, Zainol I. In vitro characterization of a chitosan skin regenerating template as a scaffold for cells cultivation. SPRINGERPLUS 2013; 2:79. [PMID: 23503998 PMCID: PMC3597272 DOI: 10.1186/2193-1801-2-79] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 01/20/2013] [Indexed: 12/11/2022]
Abstract
Chitosan is a marine-derived product that has been widely used in clinical applications, especially in skin reconstruction. The mammalian scaffolds derived from bovine and porcine material have many limitations, for example, prion transmission and religious concerns. Therefore, we created a chitosan skin regenerating template (SRT) and investigated the behavior of fibroblast cell-scaffold constructs. Primary human dermal fibroblasts (HDF) were isolated and then characterized using vimentin and versican. HDF were seeded into chitosan SRT at a density of 3×106 cells/cm2 for fourteen days. Histological analysis and live cells imaging revealed that the cell-chitosan constructs within interconnected porous chitosan showed significant interaction between the cells as well as between the cells and the chitosan. Scanning electron microscopy (SEM) analysis revealed cells spreading and covering the pores. As the pore sizes of the chitosan SRT range between 40–140 μm, an average porosity is about 93 ± 12.57% and water uptake ratio of chitosan SRT is 536.02 ± 14.29%, it is a supportive template for fibroblast attachment and has potential in applications as a dermal substitute.
Collapse
|
85
|
Zhang X, Wang Y, Gao Y, Liu X, Bai T, Li M, Li L, Chi G, Xu H, Liu F, Liu JY, Li Y. Maintenance of high proliferation and multipotent potential of human hair follicle-derived mesenchymal stem cells by growth factors. Int J Mol Med 2013; 31:913-21. [PMID: 23403715 DOI: 10.3892/ijmm.2013.1272] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/04/2013] [Indexed: 12/27/2022] Open
Abstract
Cell therapy and cell-based tissue engineering is becoming increasingly important in regenerative medicine. Stem cells that are characterized by self-renewal, high proliferation and multiple differentiation potentials have attracted attention in cell-based regenerative medicine. Maintaining the aforementioned characteristics of stem cells is the first key step in cell-based regenerative medicine. Basic fibroblast growth factor (bFGF) is a well-known growth factor that efficiently maintains the self-renewal, high proliferation and multilineage differentiation potential of stem cells. Whether or not other growth factors, such as acidic fibroblast growth factor (aFGF) and epidermal growth factor (EGF) have similar effects has yet to be fully elucidated. Human hair follicle-derived mesenchymal stem cells (HF-MSCs) were obtained by organ culture. They exhibited surface markers of bone marrow mesenchymal stem cells as shown by positive staining for CD44, CD73, CD90 and CD105, and they also displayed trilineage differentiation potentials into adipocytes, chondrocytes and osteoblasts by cytochemistry and qRT-PCR. Flow cytometry analysis showed that up to 70% of HF-MSCs cultured in the presence of aFGF, bFGF or EGF stayed at the G0/G1 phase. Proliferation analysis showed that both bFGF and EGF at as low as 1 ng/ml and aFGF at above 5 ng/ml levels significantly increased the proliferation of HF-MSCs by cell counting. Consistent with proliferation analysis, immunofluorescence staining showed that more than 95% of HF-MSCs cultured in the presence of aFGF, bFGF and EGF were positively stained for proliferating cell nuclear antigen. HF-MSCs cultured in the presence of aFGF, bFGF or EGF retained marked trilineage differentiation potentials. By contrast, HF-MSCs cultured in the absence of bFGF, aFGF and EGF lost multipotency.
Collapse
Affiliation(s)
- Xueyan Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Li X, Zhou ZP, Hu L, Zhang WJ, Li W. Apoptotic cell death induced by 5-aminolaevulinic acid-mediated photodynamic therapy of hypertrophic scar-derived fibroblasts. J DERMATOL TREAT 2013; 25:428-33. [DOI: 10.3109/09546634.2012.697987] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
87
|
Differentiation of human dermal fibroblasts towards endothelial cells. Differentiation 2013; 85:67-77. [DOI: 10.1016/j.diff.2013.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 12/11/2012] [Accepted: 01/23/2013] [Indexed: 01/20/2023]
|
88
|
de la Puente P, Ludeña D, López M, Ramos J, Iglesias J. Differentiation within autologous fibrin scaffolds of porcine dermal cells with the mesenchymal stem cell phenotype. Exp Cell Res 2013; 319:144-52. [DOI: 10.1016/j.yexcr.2012.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 10/17/2012] [Accepted: 10/20/2012] [Indexed: 10/27/2022]
|
89
|
Shim JH, Lee TR, Shin DW. Enrichment and characterization of human dermal stem/progenitor cells by intracellular granularity. Stem Cells Dev 2013; 22:1264-74. [PMID: 23336432 DOI: 10.1089/scd.2012.0244] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adult stem cells from the dermis would be an attractive cell source for therapeutic purposes as well as studying the process of skin aging. Several studies have reported that human dermal stem/progenitor cells (hDSPCs) with multipotent properties exist within the dermis of adult human skin. However, these cells have not been well characterized, because methods for their isolation or enrichment have not yet been optimized. In the present study, we enriched high side scatter (SSC(high))-hDSPCs from normal human dermal fibroblasts using a structural characteristic, intracellular granularity, as a sorting parameter. The SSC(high)-hDSPCs had high in vitro proliferation properties and expressed high levels of SOX2 and S100B, similar to previously identified mouse SOX2+ hair follicle dermal stem cells. The SSC(high)-hDSPCs could differentiate into not only mesodermal cell types, for example, adipocytes, chondrocytes, and osteoblasts, but also neuroectodermal cell types, such as neural cells. In addition, the SSC(high)-hDSPCs exhibited no significant differences in the expression of nestin, vimentin, SNAI2, TWIST1, versican, and CORIN compared with non-hDSPCs. These cells are therefore different from the previously identified multipotent fibroblasts and skin-derived progenitors. In this study, we suggest that hDSPCs can be enriched by using characteristic of their high intracellular granularity, and these SSC(high)-hDSPCs exhibit high in vitro proliferation and differentiation potentials.
Collapse
Affiliation(s)
- Joong Hyun Shim
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Republic of Korea
| | | | | |
Collapse
|
90
|
Llombart-Bosch A, Monteagudo C, Santa Cruz DJ. Introduction. Semin Diagn Pathol 2013; 30:1-3. [PMID: 23327725 DOI: 10.1053/j.semdp.2012.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
91
|
Zong Z, Li N, Ran X, Su Y, Shen Y, Shi CM, Cheng TM. Isolation and characterization of two kinds of stem cells from the same human skin back sample with therapeutic potential in spinal cord injury. PLoS One 2012; 7:e50222. [PMID: 23226248 PMCID: PMC3511430 DOI: 10.1371/journal.pone.0050222] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 10/22/2012] [Indexed: 01/22/2023] Open
Abstract
Backgrounds and Objective Spinal cord injury remains to be a challenge to clinicians and it is attractive to employ autologous adult stem cell transplantation in its treatment, however, how to harvest cells with therapeutic potential easily and how to get enough number of cells for transplantation are challenging issues. In the present study, we aimed to isolate skin-derived precursors (SKPs) and dermal multipotent stem cells (dMSCs) simultaneously from single human skin samples from patients with paraplegia. Methods Dissociated cells were initially generated from the dermal layer of skin samples from patients with paraplegia and cultured in SKPs proliferation medium. Four hours later, many cells adhered to the base of the flask. The suspended cells were then transferred to another flask for further culture as SKPs, while the adherent cells were cultured in dMSCs proliferation medium. Twenty-four hours later, the adherent cells were harvested and single-cell colonies were generated using serial dilution method. [3H]thymidine incorporation assay, microchemotaxis Transwell chambers assay, RT-PCR and fluorescent immunocytochemistry were employed to examine the characterizations of the isolated cells. Results SKPs and dMSCs were isolated simultaneously from a single skin sample. SKPs and dMSCs differed in several respects, including in terms of intermediate protein expression, proliferation capacities, and differentiation tendencies towards mesodermal and neural progenies. However, both SKPs and dMSCs showed high rates of differentiation into neurons and Schwann cells under appropriate inducing conditions. dMSCs isolated by this method showed no overt differences from dMSCs isolated by routine methods. Conclusions Two kinds of stem cells, namely SKPs and dMSCs, can be isolated simultaneously from individual human skin sample from paraplegia patients. Both of them show ability to differentiate into neural cells under proper inducing conditions, indicating their potential for the treatment of spinal cord injury patients by autologous cell transplantation.
Collapse
Affiliation(s)
- Zhaowen Zong
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, China.
| | | | | | | | | | | | | |
Collapse
|
92
|
Wakao S, Kuroda Y, Ogura F, Shigemoto T, Dezawa M. Regenerative Effects of Mesenchymal Stem Cells: Contribution of Muse Cells, a Novel Pluripotent Stem Cell Type that Resides in Mesenchymal Cells. Cells 2012; 1:1045-60. [PMID: 24710542 PMCID: PMC3901150 DOI: 10.3390/cells1041045] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/01/2012] [Accepted: 11/05/2012] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are easily accessible and safe for regenerative medicine. MSCs exert trophic, immunomodulatory, anti-apoptotic, and tissue regeneration effects in a variety of tissues and organs, but their entity remains an enigma. Because MSCs are generally harvested from mesenchymal tissues, such as bone marrow, adipose tissue, or umbilical cord as adherent cells, MSCs comprise crude cell populations and are heterogeneous. The specific cells responsible for each effect have not been clarified. The most interesting property of MSCs is that, despite being adult stem cells that belong to the mesenchymal tissue lineage, they are able to differentiate into a broad spectrum of cells beyond the boundary of mesodermal lineage cells into ectodermal or endodermal lineages, and repair tissues. The broad spectrum of differentiation ability and tissue-repairing effects of MSCs might be mediated in part by the presence of a novel pluripotent stem cell type recently found in adult human mesenchymal tissues, termed multilineage-differentiating stress enduring (Muse) cells. Here we review recently updated studies of the regenerative effects of MSCs and discuss their potential in regenerative medicine.
Collapse
Affiliation(s)
- Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Yasumasa Kuroda
- Department of Anatomy and Anthropology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Fumitaka Ogura
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Taeko Shigemoto
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| |
Collapse
|
93
|
Skin-derived multipotent stromal cells--an archrival for mesenchymal stem cells. Cell Tissue Res 2012; 350:1-12. [PMID: 22885940 DOI: 10.1007/s00441-012-1471-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/21/2012] [Indexed: 12/13/2022]
Abstract
Progenitor stem cells have been identified, isolated and characterized in numerous tissues and organs. However, their therapeutic potential and the use of these stem cells remain elusive except for a few progenitor cells from bone marrow, umbilical cord blood, eyes and dental pulp. The use of bone marrow-derived hematopoietic stem cells (HSC) or mesenchymal stem cells (MSCs) is restricted due to their extreme invasive procedures, low differentiation potential with age and rejection. Thus, we need a clinical grade alternative to progenitor stem cells with a high potential to differentiate, naïve and is relatively easy in in vitro propagation. In this review, we summarize cell populations of adherent and floating spheres derived from different origins of skin, or correctly foreskin, by enzymatic digestion compared with established MSCs. The morphology, phenotype, differentiation capability and immunosuppressive property of the adherent cell populations are comparable with MSCs. Serum-free cultured floating spheres have limited mesodermal but higher neurogenic differentation potential, analogous to neural crest stem cells. Both the populations confirmed their plethora potential in in vitro. Together, it may be noted that the skin-derived adherent cell populations and floating cells can be good alternative sources of progenitor cells especially in cosmetic, plastic and sports regenerative medicine.
Collapse
|
94
|
Enrichment and characterization of human dermal stem/progenitor cells using collagen type IV. J Dermatol Sci 2012; 67:202-5. [PMID: 22784784 DOI: 10.1016/j.jdermsci.2012.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 05/24/2012] [Accepted: 06/18/2012] [Indexed: 01/06/2023]
|
95
|
Wakao S, Kitada M, Dezawa M. The elite and stochastic model for iPS cell generation: Multilineage-differentiating stress enduring (Muse) cells are readily reprogrammable into iPS cells. Cytometry A 2012; 83:18-26. [DOI: 10.1002/cyto.a.22069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/02/2012] [Accepted: 04/16/2012] [Indexed: 01/25/2023]
|
96
|
Kim B, Yoon BS, Moon JH, Kim J, Jun EK, Lee JH, Kim JS, Baik CS, Kim A, Whang KY, You S. Differentiation of human labia minora dermis-derived fibroblasts into insulin-producing cells. Exp Mol Med 2012; 44:26-35. [PMID: 22020533 DOI: 10.3858/emm.2012.44.1.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recent evidence has suggested that human skin fibroblasts may represent a novel source of therapeutic stem cells. In this study, we report a 3-stage method to induce the differentiation of skin fibroblasts into insulin- producing cells (IPCs). In stage 1, we establish the isolation, expansion and characterization of mesenchymal stem cells from human labia minora dermis- derived fibroblasts (hLMDFs) (stage 1: MSC expansion). hLMDFs express the typical mesenchymal stem cell marker proteins and can differentiate into adipocytes, osteoblasts, chondrocytes or muscle cells. In stage 2, DMEM/F12 serum-free medium with ITS mix (insulin, transferrin, and selenite) is used to induce differentiation of hLMDFs into endoderm-like cells, as determined by the expression of the endoderm markers Sox17, Foxa2, and PDX1 (stage 2: mesenchymal-endoderm transition). In stage 3, cells in the mesenchymal- endoderm transition stage are treated with nicotinamide in order to further differentiate into self-assembled, 3-dimensional islet cell-like clusters that express multiple genes related to pancreatic β-cell development and function (stage 3: IPC). We also found that the transplantation of IPCs can normalize blood glucose levels and rescue glucose homeostasis in streptozotocin- induced diabetic mice. These results indicate that hLMDFs have the capacity to differentiate into functionally competent IPCs and represent a potential cell-based treatment for diabetes mellitus.
Collapse
Affiliation(s)
- Bona Kim
- Laboratory of Cell Function Regulation College of Life Sciences and Biotechnology Korea University Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Chang H, Knothe Tate ML. Concise review: the periosteum: tapping into a reservoir of clinically useful progenitor cells. Stem Cells Transl Med 2012. [PMID: 23197852 DOI: 10.5966/sctm.2011-0056] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Elucidation of the periosteum and its regenerative potential has become a hot topic in orthopedics. Yet few review articles address the unique features of periosteum-derived cells, particularly in light of translational therapies and engineering solutions inspired by the periosteum's remarkable regenerative capacity. This review strives to define periosteum-derived cells in light of cumulative research in the field; in addition, it addresses clinical translation of current insights, hurdles to advancement, and open questions in the field. First, we examine the periosteal niche and its inhabitant cells and the key characteristics of these cells in the context of mesenchymal stem cells and their relevance for clinical translation. We compare periosteum-derived cells with those derived from the marrow niche in in vivo studies, addressing commonalities as well as features unique to periosteum cells that make them potentially ideal candidates for clinical application. Thereafter, we review the differentiation and tissue-building properties of periosteum cells in vitro, evaluating their efficacy in comparison with marrow-derived cells. Finally, we address a new concept of banking periosteum and periosteum-derived cells as a novel alternative to currently available autogenic umbilical blood and perinatal tissue sources of stem cells for today's population of aging adults who were "born too early" to bank their own perinatal tissues. Elucidating similarities and differences inherent to multipotent cells from distinct tissue niches and their differentiation and tissue regeneration capacities will facilitate the use of such cells and their translation to regenerative medicine.
Collapse
Affiliation(s)
- Hana Chang
- Departments of Biomedical Engineering and Mechanical & Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
98
|
Short exposure to collagenase and coculture with mouse embryonic pancreas improve human dermal fibroblast culture. Biotechnol Appl Biochem 2012; 59:254-61. [DOI: 10.1002/bab.1020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 04/02/2012] [Indexed: 12/21/2022]
|
99
|
Orbay H, Tobita M, Mizuno H. Mesenchymal stem cells isolated from adipose and other tissues: basic biological properties and clinical applications. Stem Cells Int 2012; 2012:461718. [PMID: 22666271 PMCID: PMC3361347 DOI: 10.1155/2012/461718] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 03/02/2012] [Indexed: 01/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that were initially isolated from bone marrow. However, subsequent research has shown that other adult tissues also contain MSCs. MSCs originate from mesenchyme, which is embryonic tissue derived from the mesoderm. These cells actively proliferate, giving rise to new cells in some tissues, but remain quiescent in others. MSCs are capable of differentiating into multiple cell types including adipocytes, chondrocytes, osteocytes, and cardiomyocytes. Isolation and induction of these cells could provide a new therapeutic tool for replacing damaged or lost adult tissues. However, the biological properties and use of stem cells in a clinical setting must be well established before significant clinical benefits are obtained. This paper summarizes data on the biological properties of MSCs and discusses current and potential clinical applications.
Collapse
Affiliation(s)
- Hakan Orbay
- Department of Plastic and Reconstructive Surgery, Nippon Medical School, Tokyo 113-0022, Japan
| | - Morikuni Tobita
- Department of Dentistry and Oral Surgery, Self Defense Force Hospital, Yokosuka 237-0071, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo 1138421, Japan
| |
Collapse
|
100
|
Lee JP, Cha HJ, Lee KS, Lee KK, Son JH, Kim KN, Lee DK, An S. Phytosphingosine-1-phosphate represses the hydrogen peroxide-induced activation of c-Jun N-terminal kinase in human dermal fibroblasts through the phosphatidylinositol 3-kinase/Akt pathway. Arch Dermatol Res 2012; 304:673-8. [PMID: 22566145 DOI: 10.1007/s00403-012-1241-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 10/28/2022]
Abstract
Dermal fibroblasts are differentiated mesenchymal cells that regulate the extracellular matrix through the production of dermis components. Dermal fibroblasts can be damaged by reactive oxygen species induced by ultraviolet rays and chemicals. In addition to its effects on the dermis, oxidative stress poses a major threat to organisms and is believed to play an essential role in many disease processes. In this study, we show that human dermal fibroblasts (HDFs) express sphingosine-1-phosphate (S1P) receptors S1P(1), S1P(2), and S1P(3). In addition, cell viability of HDFs is increased by phytosphingosine-1-phosphate (PhS1P) via regulation of the Jun N-terminal kinase (JNK)/Akt pathway. Interestingly, regulation of the JNK/Akt pathway by PhS1P attenuated H(2)O(2)-induced cell growth arrest. Together, our data indicate that PhS1P attenuates H(2)O(2)-induced growth arrest through regulation of the signal molecules Akt and JNK, and suggest that PhS1P may have value as an anti-aging material in cosmetics and medicine.
Collapse
Affiliation(s)
- Jeong Pyo Lee
- Coreana Cosmetics Co., Ltd., Seonggeo-eup, Seobuk-gu, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|