51
|
Musavi L, Brandacher G, Hoke A, Darrach H, Lee WPA, Kumar A, Lopez J. Muscle-derived stem cells: important players in peripheral nerve repair. Expert Opin Ther Targets 2018; 22:1009-1016. [PMID: 30347175 DOI: 10.1080/14728222.2018.1539706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Stem cell therapy for peripheral nerve repair is a rapidly evolving field in regenerative medicine. Although most studies to date have investigated stem cells originating from bone marrow or adipose, skeletal muscle has recently been recognized as an abundant and easily accessible source of stem cells. Muscle-derived stem cells (MDSCs) are a diverse population of multipotent cells with pronounced antioxidant and regenerative capacity. Areas covered: The current literature on the various roles MDSCs serve within the micro- and macro-environment of nerve injury. Furthermore, the exciting new research that is establishing MDSC-cellular therapy as an important therapeutic modality to improve peripheral nerve regeneration. Expert opinion: MDSCs are a promising therapeutic agent for the repair of peripheral nerves; MDSCs not only undergo gliogenesis and angiogenesis, but they also orchestrate larger pro-regenerative host responses. However, the isolation, transformation, and in-vivo behavior of MDSCs require further evaluation prior to clinical application.
Collapse
Affiliation(s)
- Leila Musavi
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - Gerald Brandacher
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - Ahmet Hoke
- b The Solomon H Snyder Department of Neuroscience , Johns Hopkins University , Baltimore , Maryland
| | - Halley Darrach
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - W P Andrew Lee
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - Anand Kumar
- c Department of Plastic & Reconstructive Surgery , Case Western Reserve University, Rainbow Babies Children's Hospital , Cleveland , OH , USA
| | - Joseph Lopez
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| |
Collapse
|
52
|
Kouroupis D, Sanjurjo-Rodriguez C, Jones E, Correa D. Mesenchymal Stem Cell Functionalization for Enhanced Therapeutic Applications. TISSUE ENGINEERING PART B-REVIEWS 2018; 25:55-77. [PMID: 30165783 DOI: 10.1089/ten.teb.2018.0118] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IMPACT STATEMENT Culture expansion of MSCs has detrimental effects on various cell characteristics and attributes (e.g., phenotypic changes and senescence), which, in addition to inherent interdonor variability, negatively impact the standardization and reproducibility of their therapeutic potential. The identification of innate distinct functional MSC subpopulations, as well as the description of ex vivo protocols aimed at maintaining phenotypes and enhancing specific functions have the potential to overcome these limitations. The incorporation of those approaches into cell-based therapy would significantly impact the field, as more reproducible clinical outcomes may be achieved.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- 1 Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, Florida.,2 Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Clara Sanjurjo-Rodriguez
- 3 Leeds Institute of Rheumatic and Musculoskeletal Disease, Saint James University Hospital, University of Leeds, Leeds, United Kingdom.,4 Department of Biomedical Sciences, Medicine and Physiotherapy, University of A Coruña, CIBER-BBN-Institute of Biomedical Research of A Coruña (INIBIC), A Coruña, Spain
| | - Elena Jones
- 3 Leeds Institute of Rheumatic and Musculoskeletal Disease, Saint James University Hospital, University of Leeds, Leeds, United Kingdom
| | - Diego Correa
- 1 Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, Florida.,2 Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
53
|
Álvarez V, Sánchez-Margallo FM, Macías-García B, Gómez-Serrano M, Jorge I, Vázquez J, Blázquez R, Casado JG. The immunomodulatory activity of extracellular vesicles derived from endometrial mesenchymal stem cells on CD4+ T cells is partially mediated by TGFbeta. J Tissue Eng Regen Med 2018; 12:2088-2098. [PMID: 30058282 DOI: 10.1002/term.2743] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/26/2022]
Abstract
Endometrial mesenchymal stem cells (endMSCs) reside in the basal and functional layer of human endometrium and participate in tissue remodelling, which is required for maintaining the regenerative capacity of the endometrium. The endMSCs are multipotent stem cells and exhibit immunomodulatory effects. This paper aimed to evaluate the regulatory effects of extracellular vesicles derived from endMSCs (EV-endMSCs) in the setting of T cell activation. In vitro stimulations of lymphocytes were performed in the presence of EV-endMSCs. These in vitro-stimulated lymphocytes were functionally and phenotypically characterized to distinguish CD4+ and CD8+ T cell differentiation subsets. Moreover, the inhibition of TGFβ was performed with neutralizing antibodies. The phenotype and nanoparticle tracking analysis of the EV-endMSCs demonstrated that they are similar in terms of size distribution to other mesenchymal stem cells-derived exosomes. The in vitro assays showed an immunomodulatory potential of these vesicles to counteract the differentiation of CD4+ T cells. The quantification of active TGFβ in EV-endMSCs was found to be very high when compared with extracellular vesicles-free concentrated supernatants. Finally, the neutralization of TGFβ significantly attenuated the immunomodulatory activity of EV-endMSCs. In summary, this is the first report demonstrating that EV-endMSCs exhibit a potent inhibitory effect against CD4+ T cell activation, which is partially mediated by TGFβ signalling.
Collapse
Affiliation(s)
- Verónica Álvarez
- Stem Cell Therapy Unit, Jesus Uson Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesus Uson Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Beatriz Macías-García
- Assisted Reproduction Unit, Jesus Uson Minimally Invasive Surgery Centre, Cáceres, Spain
| | - María Gómez-Serrano
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Inmaculada Jorge
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Rebeca Blázquez
- Stem Cell Therapy Unit, Jesus Uson Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Javier G Casado
- Stem Cell Therapy Unit, Jesus Uson Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
54
|
The Human Skin-Derived Precursors for Regenerative Medicine: Current State, Challenges, and Perspectives. Stem Cells Int 2018; 2018:8637812. [PMID: 30123295 PMCID: PMC6079335 DOI: 10.1155/2018/8637812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 02/05/2023] Open
Abstract
Skin-derived precursors (SKPs) are an adult stem cell source with self-renewal and multipotent differentiation. Although rodent SKPs have been discussed in detail in substantial studies, human SKPs (hSKPs) are rarely reported. Understanding the biological properties and possible mechanisms underlying hSKPs has important implications for regenerative medicine particularly clinical applications, as human-derived sources are more suitable for clinical transplantation. The finding that hSKPs derivatives, such as neural and mesodermal progeny, have both in vitro and in vivo function without any genetical modification makes hSKPs a trustable, secure, and accessible resource for cell-based therapy. Here, we provide an overview of hSKPs, describing their characteristics, originations and niches, and potential applications. A comparison between traditional and innovative culture methods used for hSKPs is also introduced. Furthermore, we discuss the challenges and the future perspectives towards the field of hSKPs. With this review, we hope to point out the current stage of hSKPs and highlight the problems that remain in this field.
Collapse
|
55
|
Masia F, Glen A, Stephens P, Langbein W, Borri P. Label-free quantitative chemical imaging and classification analysis of adipogenesis using mouse embryonic stem cells. JOURNAL OF BIOPHOTONICS 2018; 11:e201700219. [PMID: 29573183 DOI: 10.1002/jbio.201700219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/26/2018] [Indexed: 06/08/2023]
Abstract
Stem cells have received much attention recently for their potential utility in regenerative medicine. The identification of their differentiated progeny often requires complex staining procedures, and is challenging for intermediary stages which are a priori unknown. In this work, the ability of label-free quantitative coherent anti-Stokes Raman scattering (CARS) micro-spectroscopy to identify populations of intermediate cell states during the differentiation of murine embryonic stem cells into adipocytes is assessed. Cells were imaged at different days of differentiation by hyperspectral CARS, and images were analysed with an unsupervised factorization algorithm providing Raman-like spectra and spatially resolved maps of chemical components. Chemical decomposition combined with a statistical analysis of their spatial distributions provided a set of parameters that were used for classification analysis. The first 2 principal components of these parameters indicated 3 main groups, attributed to undifferentiated cells, cells differentiated into committed white pre-adipocytes, and differentiating cells exhibiting a distinct protein globular structure with adjacent lipid droplets. An unsupervised classification methodology was developed, separating undifferentiated cell from cells in other stages, using a novel method to estimate the optimal number of clusters. The proposed unsupervised classification pipeline of hyperspectral CARS data offers a promising new tool for automated cell sorting in lineage analysis.
Collapse
Affiliation(s)
- Francesco Masia
- School of Physics and Astronomy, Cardiff University, Cardiff, UK
| | - Adam Glen
- School of Dentistry, Cardiff University, Cardiff, UK
| | - Phil Stephens
- School of Dentistry, Cardiff University, Cardiff, UK
| | | | - Paola Borri
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
56
|
Behavior of multipotent stem cells isolated in mobilized peripheral blood from sheep after culture with human chondrogenic medium. Tissue Cell 2018; 52:116-123. [PMID: 29857820 DOI: 10.1016/j.tice.2018.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 05/08/2018] [Indexed: 01/30/2023]
Abstract
Today, regenerative medicine requires new sources of multipotent stem cells for their differentiation to chondrocytes using the mediums of differentiation available in the market. This study aimed to determine whether the Mesenchymal Stem Cells (MSCs) isolated from Mobilized Peripheral Blood (MPB) in sheep using the Granulocyte Colony-Stimulating Factor (G-CSF), have the ability of first acquire a fibroblast-like morphology after being forced out of the bone marrow niche by G-CSF and second, if the cells have the capacity to express collagen type-II α I in primary culture using a human commercial media of differentiation. Six Suffolk male sheep with age of 2 years were mobilized using G-CSF. One subcutaneous injection of 10 mcg per kilogram of bodyweight were administered every 24 h during three consecutive days. At day four, a sample of 20 mL of peripheral blood was harvested, afterwards, monocytes cells were separated by ficoll gradient. The mobilized MSCs were expanded in primary culture in DMEM medium supplemented with 10% adult sheep serum for three weeks and characterized by an antibody panel for surface markers: CD105, CD90, CD73, CD34, and CD45, before and after primary culture. Subsequently, an aliquot of cells in the first pass were cultured in a commercial human chondrogenic medium for three weeks. As a result, the percentage of surface markers for MSCs (CD105, CD90, CD73) in expanded cells in primary culture significantly increased, at the same time a decrease in the markers for hematopoietic cells (CD34 and CD45) was observed and the cells morphology was fibroblast-like. After three weeks of differentiation culture, the immunofluorescence analysis evidenced the expression of collagen-type-II. It was concluded that Mesenchymal Stem Cells isolated from mobilized peripheral blood in sheep have the ability to pre-differentiate into chondral like cells and express collagen type-II when are stimulated with a human commercial chondrogenic medium in monolayer culture.
Collapse
|
57
|
Santos AK, Vieira MS, Vasconcellos R, Goulart VAM, Kihara AH, Resende RR. Decoding cell signalling and regulation of oligodendrocyte differentiation. Semin Cell Dev Biol 2018; 95:54-73. [PMID: 29782926 DOI: 10.1016/j.semcdb.2018.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.
Collapse
Affiliation(s)
- A K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - R Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - V A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - A H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - R R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
58
|
Galkowski D, Ratajczak MZ, Kocki J, Darzynkiewicz Z. Of Cytometry, Stem Cells and Fountain of Youth. Stem Cell Rev Rep 2018; 13:465-481. [PMID: 28364326 DOI: 10.1007/s12015-017-9733-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Outlined are advances of cytometry applications to identify and sort stem cells, of laser scanning cytometry and ImageStream imaging instrumentation to further analyze morphometry of these cells, and of mass cytometry to classify a multitude of cellular markers in large cell populations. Reviewed are different types of stem cells, including potential candidates for cancer stem cells, with respect to their "stemness", and other characteristics. Appraised is further progress in identification and isolation of the "very small embryonic-like stem cells" (VSELs) and their autogenous transplantation for tissue repair and geroprotection. Also assessed is a function of hyaluronic acid, the major stem cells niche component, as a guardian and controller of stem cells. Briefly appraised are recent advances and challenges in the application of stem cells in regenerative medicine and oncology and their future role in different disciplines of medicine, including geriatrics.
Collapse
Affiliation(s)
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University in Lublin, 20-080, Lublin, Poland
| | - Zbigniew Darzynkiewicz
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, NY, 10095, USA.
| |
Collapse
|
59
|
Ge L, Yu D, Su R, Cao Y. [Effects of hypoxia-inducible factor 1α on hypoxic tolerance of human amniotic mesenchymal stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:264-269. [PMID: 29806273 PMCID: PMC8414270 DOI: 10.7507/1002-1892.201710104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/05/2018] [Indexed: 12/13/2022]
Abstract
Objective Under hypoxic conditions, the survival and apoptosis of human amniotic mesenchymal stem cells (hAMSCs) were observed by transient transfection of hypoxia-inducible factor 1α (HIF-1α) gene, to investigate the effect of HIF-1α on hypoxic tolerance of hAMSCs. Methods The hAMSCs were isolated and cultured from amniotic membrane tissue from voluntary donors who were treated with cesarean section. And the morphological observation by inverted phase contrast microscope and immunofluorescence detection of the expressions of stem cell markers OCT-4 and NANOG were performed to identify the cultured cells. The third generation hAMSCs were treated with 200 μmol/L CoCl 2, and transient transfection of plasmids were added according to the following grouping: group A was hAMSCs blank group; group B was pcDNA3.1 negative control group; group C was short hairpin RNA (shRNA) negative control group; group D was shRNA-HIF-1α interference group; group E was pcDNA3.1-HIF-1α over expression group. Cell survival rate of each group was measured by cell counting kit 8 (CCK-8) at 12, 24, 48 hours after hypoxia treatment. Flow cytometry was used to detect apoptosis rate of each group at 24 hours after hypoxia treatment. The expression levels of HIF-1α, vascular endothelial growth factor (VEGF), B-cell lymphoma 2 (Bcl-2), Bax, and cleaved Caspase-3 (C-Caspase-3) proteins were detected by Western blot at 24 hours after hypoxia treatment. Results CCK-8 assay showed that the cell survival rate of group D was significantly lower than those of groups A and C at all time points after hypoxia treatment; while the cell survival rate in group E was significantly increased than those in groups A and B, and the diffrences at 24 hours were significant ( P<0.05). In group E, the cell survival rate at 24 hours was significantly higher than those at 12 and 48 hours ( P<0.05). The results of flow cytometry showed that the apoptosis rate in group D was significantly higher than those in groups A and C ( P<0.05), and the apoptosis rate in group E was significantly lower than those in groups A and B ( P<0.05). Western blot showed that the expressions of HIF-1α, VEGF, and Bcl-2 proteins in group D were significantly decreased when compared with those in groups A and C, and the expressions of Bax and C-Caspase-3 proteins were significantly increased ( P<0.05). On the contrary, the expressions of HIF-1α, VEGF, and Bcl-2 proteins in group E were significantly higher than those in groups A and B, and the expressions of Bax and C-Caspase-3 proteins were significantly decreased ( P<0.05). Conclusion Overexpression of HIF-1α gene can significantly improve hAMSCs tolerance to hypoxia, the mechanism may be related to up-regulation of VEGF and Bcl-2 expressions, and down-regulation of Bax and C-Caspase-3 expressions.
Collapse
Affiliation(s)
- Lihao Ge
- Department of Orthopedics, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| | - Deshui Yu
- Jinzhou Medical University, Jinzhou Liaoning, 121001,
| | - Ruichao Su
- Department of Orthopedics, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| | - Yang Cao
- Department of Orthopedics, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| |
Collapse
|
60
|
Liu Y, Niu R, Yang F, Yan Y, Liang S, Sun Y, Shen P, Lin J. Biological characteristics of human menstrual blood-derived endometrial stem cells. J Cell Mol Med 2018; 22:1627-1639. [PMID: 29278305 PMCID: PMC5824373 DOI: 10.1111/jcmm.13437] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/01/2017] [Indexed: 12/20/2022] Open
Abstract
Successful isolation of human endometrial stem cells from menstrual blood, namely menstrual blood-derived endometrial stem cells (MenSCs), has provided enticing alternative seed cells for stem cell-based therapy. MenSCs are enriched in the self-regenerative tissue, endometrium, which shed along the periodic menstrual blood and thus their acquisition involves no physical invasiveness. However, the impact of the storage duration of menstrual blood prior to stem cell isolation, the age of the donor, the number of passages on the self-renewing of MenSCs, the paracrine production of biological factors in MenSCs and expression of adhesion molecules on MenSCs remain elusive. In this study, we confirmed that MenSCs reside in shedding endometrium, and documented that up to 3 days of storage at 4°C has little impact on MenSCs, while the age of the donor and the number of passages are negatively associated with proliferation capacity of MenSCs. Moreover, we found that MenSCs were actually immune-privileged and projected no risk of tumour formation. Also, we documented a lung- and liver-dominated, spleen- and kidney-involved organic distribution profile of MenSC 3 days after intravenous transfer into mice. At last, we suggested that MenSCs may have potentially therapeutic effects on diseases through paracrine effect and immunomodulation.
Collapse
Affiliation(s)
- Yanli Liu
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Rongcheng Niu
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Fen Yang
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Yan Yan
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Shengying Liang
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Yuliang Sun
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Ping Shen
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
- Deutsches Rheuma‐Forschungszentrum, a Leibniz InstituteBerlinGermany
| | - Juntang Lin
- Henan Key Laboratory of Medical Tissue RegenerationStem Cells & Biotherapy Engineering Research Center of HenanCollege of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
- Institute of Anatomy IJena University HospitalUniversity of Jena School of MedicineJenaGermany
| |
Collapse
|
61
|
Abstract
Purpose of Review The aim of the study is to provide an overview on the possibility of treating congenital disorders prenatally with mesenchymal stromal cells (MSCs). Recent Findings MSCs have multilineage potential and a low immunogenic profile and are immunomodulatory and more easy to expand in culture. Their ability to migrate, engraft and differentiate, or act via a paracrine effect on target tissues makes MSCs candidates for clinical therapies. Fetal and extra-fetal MSCs offer higher therapeutic potential compared to MSCs derived from adult sources. Summary MSCs may be safely transplanted prenatally via ultrasound-guided injection into the umbilical cord. Due to these characteristics, fetal MSCs are of great interest in the field of in utero stem cell transplantation for treatment of congenital disease.
Collapse
|
62
|
Corrêa NCR, Kuligovski C, Paschoal ACC, Abud APR, Rebelatto CLK, Leite LMB, Senegaglia AC, Dallagiovanna B, Aguiar AMD. Human adipose-derived stem cells (ADSC) and human periodontal ligament stem cells (PDLSC) as cellular substrates of a toxicity prediction assay. Regul Toxicol Pharmacol 2018; 92:75-82. [DOI: 10.1016/j.yrtph.2017.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023]
|
63
|
Fernandes M, Valente SG, Sabongi RG, Gomes dos Santos JB, Leite VM, Ulrich H, Nery AA, da Silva Fernandes MJ. Bone marrow-derived mesenchymal stem cells versus adipose-derived mesenchymal stem cells for peripheral nerve regeneration. Neural Regen Res 2018; 13:100-104. [PMID: 29451213 PMCID: PMC5840974 DOI: 10.4103/1673-5374.224378] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2017] [Indexed: 12/15/2022] Open
Abstract
Studies have confirmed that bone marrow-derived mesenchymal stem cells (MSCs) can be used for treatment of several nervous system diseases. However, isolation of bone marrow-derived MSCs (BMSCs) is an invasive and painful process and the yield is very low. Therefore, there is a need to search for other alterative stem cell sources. Adipose-derived MSCs (ADSCs) have phenotypic and gene expression profiles similar to those of BMSCs. The production of ADSCs is greater than that of BMSCs, and ADSCs proliferate faster than BMSCs. To compare the effects of venous grafts containing BMSCs or ADSCs on sciatic nerve injury, in this study, rats were randomly divided into four groups: sham (only sciatic nerve exposed), Matrigel (MG; sciatic nerve injury + intravenous transplantation of MG vehicle), ADSCs (sciatic nerve injury + intravenous MG containing ADSCs), and BMSCs (sciatic nerve injury + intravenous MG containing BMSCs) groups. Sciatic functional index was calculated to evaluate the function of injured sciatic nerve. Morphologic characteristics of nerves distal to the lesion were observed by toluidine blue staining. Spinal motor neurons labeled with Fluoro-Gold were quantitatively assessed. Compared with sham-operated rats, sciatic functional index was lower, the density of small-diameter fibers was significantly increased, and the number of motor neurons significantly decreased in rats with sciatic nerve injury. Neither ADSCs nor BMSCs significantly improved the sciatic nerve function of rats with sciatic nerve injury, increased fiber density, fiber diameters, axonal diameters, myelin sheath thickness, and G ratios (axonal diameter/fiber diameter ratios) in the sciatic nerve distal to the lesion site. There was no significant difference in the number of spinal motor neurons among ADSCs, BMSCs and MG groups. These results suggest that neither BMSCs nor ADSCs provide satisfactory results for peripheral nerve repair when using MG as the conductor for engraftment.
Collapse
Affiliation(s)
- Marcela Fernandes
- Division of Hand and Upper Limb Surgery, Department of Orthopedics and Traumatology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sandra Gomes Valente
- Division of Hand and Upper Limb Surgery, Department of Orthopedics and Traumatology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rodrigo Guerra Sabongi
- Division of Hand and Upper Limb Surgery, Department of Orthopedics and Traumatology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João Baptista Gomes dos Santos
- Division of Hand and Upper Limb Surgery, Department of Orthopedics and Traumatology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vilnei Mattioli Leite
- Division of Hand and Upper Limb Surgery, Department of Orthopedics and Traumatology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo, São Paulo, Brazil
| | - Arthur Andrade Nery
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo, São Paulo, Brazil
| | - Maria José da Silva Fernandes
- Division of Neurosciences, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
64
|
Nicotine-enhanced stemness and epithelial-mesenchymal transition of human umbilical cord mesenchymal stem cells promote tumor formation and growth in nude mice. Oncotarget 2017; 9:591-606. [PMID: 29416638 PMCID: PMC5787492 DOI: 10.18632/oncotarget.22712] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/01/2017] [Indexed: 12/12/2022] Open
Abstract
Cigarette smoking is a well-known risk factor in the development and progression of malignant diseases. Nicotine, the major constituent in cigarette smoke, has also shown negative effects on stem cells. Mesenchymal stem cells (MSCs) have been widely demonstrated to migrate into tumors and play key roles in cancer progression. However, the mechanisms by which nicotine impacts MSCs and tumorigenesis of lung cancer are still undetermined. In this study we investigated the effects of nicotine on human umbilical cord mesenchymal stem cells (hUC-MSCs) and the impacts of nicotine-treated hUC-MSCs on tumor formation and progression. We found that nicotine has a toxic effect on hUC-MSCs and changes the morphology, inhibits proliferation and promotes apoptosis of hUC-MSCs in a dose-dependent manner. Nicotine-treated hUC-MSCs produce higher level of IL-6. Moreover, nicotine promotes migration, stemness and epithelial-mesenchymal transition (EMT) of hUC-MSCs by inhibiting E-cadherin expression and upregulating mesenchymal markers such as N-cadherin and Vimentin, leading to the induction of stem cell markers Sox2, Nanog, Sall4, Oct4 and CD44. Migration and proliferation of non-small cell lung cancer A549 cells and breast cancer MCF-7 cells are promoted after their coculture with nicotine-treated hUC-MSCs in a cell-cell contact-independent manner. Furthermore, nicotine-treated hUC-MSCs promote tumor formation and growth of A549 cells in nude mice. These studies demonstrated that the enhanced stemness and EMT of hUC-MSCs induced by nicotine are critical for the development of tobacco-related cancers.
Collapse
|
65
|
Mei H, González S, Nakatsu MN, Baclagon ER, Chen FV, Deng SX. Human adipose-derived stem cells support the growth of limbal stem/progenitor cells. PLoS One 2017; 12:e0186238. [PMID: 29020119 PMCID: PMC5636133 DOI: 10.1371/journal.pone.0186238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/27/2017] [Indexed: 01/08/2023] Open
Abstract
The most efficient method to expand limbal stem cells (LSCs) in vitro for clinical transplantation is to culture single LSCs directly on growth-arrested mouse fibroblast 3T3 cells. To reduce possible xenobiotic contamination from 3T3s, primary human adipose-derived stem cells (ASCs) were examined as feeder cells to support the expansion of LSCs in vitro. To optimize the ASC-supported culture, freshly isolated limbal epithelial cells in the form of single cells (SC-ASC) or cell clusters (CC-ASC) were cultured using three different methods: LSCs seeded directly on feeder cells, a 3-dimensional (3D) culture system and a 3D culture system with fibrin (fibrin 3D). The expanded LSCs were examined at the end of a 2-week culture. The standard 3T3 culture served as control. Expansion of SC-ASC showed limited proliferation and exhibited differentiated morphology. CC-ASC generated epithelial cells with undifferentiated morphology in all culture methods, among which CC-ASC in 3D culture supported the highest cell doubling (cells doubled 9.0 times compared to cells doubled 4.9 times in control) while maintained the percentage of putative limbal stem/progenitor cells compared to the control. There were few cell-cell contacts between cultured LSCs and ASCs in 3D CC-ASC. In conclusion, ASCs support the growth of LSCs in the form of cell clusters but not in single cells. 3D CC-ASC could serve as a substitute for the standard 3T3 culture to expand LSCs.
Collapse
Affiliation(s)
- Hua Mei
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Sheyla González
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Martin N. Nakatsu
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Elfren R. Baclagon
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Felix V. Chen
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
- UCLA College of Letters and Science, University of California, Los Angeles, California, United States of America
| | - Sophie X. Deng
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
66
|
Uder C, Brückner S, Winkler S, Tautenhahn HM, Christ B. Mammalian MSC from selected species: Features and applications. Cytometry A 2017; 93:32-49. [PMID: 28906582 DOI: 10.1002/cyto.a.23239] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal/stem cells (MSC) are promising candidates for cellular therapy of different diseases in humans and in animals. Following the guidelines of the International Society for Cell Therapy, human MSC may be identified by expression of a specific panel of cell surface markers (CD105+, CD73+, CD90+, CD34-, CD14-, or CD11b-, CD79- or CD19-, HLA-DR-). In addition, multiple differentiation potential into at least the osteogenic, adipogenic, and chondrogenic lineage is a main criterion for MSC definition. Human MSC and MSC of a variety of mammals isolated from different tissues meet these criteria. In addition to the abovementioned, they express many more cell surface markers. Yet, these are not uniquely expressed by MSC. The gross phenotypic appearance like marker expression and differentiation potential is similar albeit not identical for MSC from different tissues and species. Similarly, MSC may feature different biological characteristics depending on the tissue source and the isolation and culture procedures. Their versatile biological qualities comprising immunomodulatory, anti-inflammatory, and proregenerative capacities rely largely on the migratory and secretory capabilities of MSC. They are attracted to sites of tissue lesion and secrete factors to promote self-repair of the injured tissue. This is a big perspective for clinical MSC applications in both veterinary and human medicine. Phase I/II clinical trials have been initiated to assess safety and feasibility of MSC therapies in acute and chronic disease settings. Yet, since the mode of MSC action in a specific disease environment is still unknown at large, it is mandatory to unravel the response of MSC from a given source onto a specific disease environment in suitable animal models prior to clinical applications. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Christiane Uder
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Sandra Brückner
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Sandra Winkler
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Hans-Michael Tautenhahn
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | | |
Collapse
|
67
|
Chen Z, Niu M, Sun M, Yuan Q, Yao C, Hou J, Wang H, Wen L, Fu H, Zhou F, Li Z, He Z. Transdifferentiation of human male germline stem cells to hepatocytes in vivo via the transplantation under renal capsules. Oncotarget 2017; 8:14576-14592. [PMID: 28107194 PMCID: PMC5362427 DOI: 10.18632/oncotarget.14713] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/11/2017] [Indexed: 12/29/2022] Open
Abstract
Here we proposed a new concept that human spermatogonial stem cells (SSCs) can transdifferentiate into hepatocytes in vivo. We first established liver injury model of mice by carbon tetrachloride to provide proper environment for human SSC transplantation. Liver mesenchymal cells were isolated from mice and identified phenotypically. Human SSC line was recombined with liver mesenchymal cells, and they were transplanted under renal capsules of nude mice with liver injury. The grafts expressed hepatocyte hallmarks, including ALB, AAT, CK18, and CYP1A2, whereas germ cell and SSC markers VASA and GPR125 were undetected in these cells, implicating that human SSCs were converted to hepatocytes. Furthermore, Western blots revealed high levels of PCNA, AFP, and ALB, indicating that human SSCs-derived hepatocytes had strong proliferation potential and features of hepatocytes. In addition, ALB–, CK8–, and CYP1A2– positive cells were detected in liver tissues of recipient mice. Significantly, no obvious lesion or teratomas was observed in several important organs and tissues of recipient mice, reflecting that transplantation of human SSCs was safe and feasible. Collectively, we have for the first time demonstrated that human SSCs can be transdifferentiated to hepatocyte in vivo. This study provides a novel approach for curing liver diseases using human SSC transplantation.
Collapse
Affiliation(s)
- Zheng Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Department of General Surgery, Suqian people's Hospital, The Affiliated Hospital of Xuzhou Medical University, Jiangsu 223800, China
| | - Minghui Niu
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Min Sun
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qingqing Yuan
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chencheng Yao
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jingmei Hou
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hong Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Liping Wen
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hongyong Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fan Zhou
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zheng Li
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Zuping He
- State Key Laboratory of Oncogenes and Related Genes, Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Institute of Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China.,Shanghai Key Laboratory of Assisted Reproduction and Reproductive Genetics, Shanghai 200127, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China
| |
Collapse
|
68
|
Nagaishi K, Mizue Y, Chikenji T, Otani M, Nakano M, Saijo Y, Tsuchida H, Ishioka S, Nishikawa A, Saito T, Fujimiya M. Umbilical cord extracts improve diabetic abnormalities in bone marrow-derived mesenchymal stem cells and increase their therapeutic effects on diabetic nephropathy. Sci Rep 2017; 7:8484. [PMID: 28814814 PMCID: PMC5559488 DOI: 10.1038/s41598-017-08921-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 07/20/2017] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSC) has been applied as the most valuable source of autologous cell transplantation for various diseases including diabetic complications. However, hyperglycemia may cause abnormalities in intrinsic BM-MSC which might lose sufficient therapeutic effects in diabetic patients. We demonstrated the functional abnormalities in BM-MSC derived from both type 1 and type 2 diabetes models in vitro, which resulted in loss of therapeutic effects in vivo in diabetic nephropathy (DN). Then, we developed a novel method to improve abnormalities in BM-MSC using human umbilical cord extracts, namely Wharton’s jelly extract supernatant (WJs). WJs is a cocktail of growth factors, extracellular matrixes and exosomes, which ameliorates proliferative capacity, motility, mitochondrial degeneration, endoplasmic reticular functions and exosome secretions in both type 1 and type 2 diabetes-derived BM-MSC (DM-MSC). Exosomes contained in WJs were a key factor for this activation, which exerted similar effects to complete WJs. DM-MSC activated by WJs ameliorated renal injury in both type 1 and type 2 DN. In this study, we developed a novel activating method using WJs to significantly increase the therapeutic effect of BM-MSC, which may allow effective autologous cell transplantation.
Collapse
Affiliation(s)
- Kanna Nagaishi
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan. .,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan.
| | - Yuka Mizue
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan.,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| | - Takako Chikenji
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan.,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| | - Miho Otani
- Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| | - Masako Nakano
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan
| | - Yusaku Saijo
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan
| | - Hikaru Tsuchida
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan
| | - Shinichi Ishioka
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Akira Nishikawa
- Department of Gynecology and Obstetrics, NTT Sapporo Hospital, Sapporo, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Mineko Fujimiya
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan.,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
69
|
Majidinia M, Sadeghpour A, Yousefi B. The roles of signaling pathways in bone repair and regeneration. J Cell Physiol 2017; 233:2937-2948. [DOI: 10.1002/jcp.26042] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Maryam Majidinia
- Solid Tumor Research Center; Urmia University of Medical Sciences; Urmia Iran
| | - Alireza Sadeghpour
- Department of Orthopedic Surgery, School of Medicine and Shohada Educational Hospital; Tabriz University of Medical Sciences; Tabriz Iran
- Drug Applied Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Bahman Yousefi
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Molecular Targeting Therapy Research Group; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
- Stem cell and Regenerative Medicine Institute; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
70
|
Reyes B, Coca MI, Codinach M, López-Lucas MD, Del Mazo-Barbara A, Caminal M, Oliver-Vila I, Cabañas V, Lope-Piedrafita S, García-López J, Moraleda JM, Fontecha CG, Vives J. Assessment of biodistribution using mesenchymal stromal cells: Algorithm for study design and challenges in detection methodologies. Cytotherapy 2017; 19:1060-1069. [PMID: 28734679 DOI: 10.1016/j.jcyt.2017.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/01/2017] [Accepted: 06/16/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS Biodistribution of candidate cell-based therapeutics is a critical safety concern that must be addressed in the preclinical development program. We aimed to design a decision tree based on a series of studies included in actual dossiers approved by competent regulatory authorities, noting that the design, execution and interpretation of pharmacokinetics studies using this type of therapy is not straightforward and presents a challenge for both developers and regulators. METHODS Eight studies were evaluated for the definition of a decision tree, in which mesenchymal stromal cells (MSCs) were administered to mouse, rat and sheep models using diverse routes (local or systemic), cell labeling (chemical or genetic) and detection methodologies (polymerase chain reaction [PCR], immunohistochemistry [IHC], fluorescence bioimaging, and magnetic resonance imaging [MRI]). Moreover, labeling and detection methodologies were compared in terms of cost, throughput, speed, sensitivity and specificity. RESULTS A decision tree was defined based on the model chosen: (i) small immunodeficient animals receiving heterologous MSC products for assessing biodistribution and other safety aspects and (ii) large animals receiving homologous labeled products; this contributed to gathering data not only on biodistribution but also on pharmacodynamics. PCR emerged as the most convenient technique despite the loss of spatial information on cell distribution that can be further assessed by IHC. DISCUSSION This work contributes to the standardization in the design of biodistribution studies by improving methods for accurate assessment of safety. The evaluation of different animal models and screening of target organs through a combination of techniques is a cost-effective and timely strategy.
Collapse
Affiliation(s)
- Blanca Reyes
- Servei de Teràpia Cellular, Banc de Sang i Teixits, Barcelona, Spain
| | - Maria Isabel Coca
- Servei de Teràpia Cellular, Banc de Sang i Teixits, Barcelona, Spain
| | | | - María Dolores López-Lucas
- Unidad de Terapia Celular y Trasplante Hematopoyético, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB, Murcia, Spain
| | | | - Marta Caminal
- Servei de Teràpia Cellular, Banc de Sang i Teixits, Barcelona, Spain
| | - Irene Oliver-Vila
- Servei de Teràpia Cellular, Banc de Sang i Teixits, Barcelona, Spain
| | - Valentín Cabañas
- Unidad de Terapia Celular y Trasplante Hematopoyético, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB, Murcia, Spain
| | - Silvia Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Joan García-López
- Servei de Teràpia Cellular, Banc de Sang i Teixits, Barcelona, Spain; Chair of Transfusion Medicine and Cellular and Tissue Therapies, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - José M Moraleda
- Unidad de Terapia Celular y Trasplante Hematopoyético, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB, Murcia, Spain
| | - Cesar G Fontecha
- Reconstructive Surgery of the Locomotor System, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cellular, Banc de Sang i Teixits, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain; Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
71
|
Tonelli FMP, Lacerda SMSN, Tonelli FCP, Costa GMJ, de França LR, Resende RR. Progress and biotechnological prospects in fish transgenesis. Biotechnol Adv 2017; 35:832-844. [PMID: 28602961 DOI: 10.1016/j.biotechadv.2017.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/04/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022]
Abstract
The history of transgenesis is marked by milestones such as the development of cellular transdifferentiation, recombinant DNA, genetic modification of target cells, and finally, the generation of simpler genetically modified organisms (e.g. bacteria and mice). The first transgenic fish was developed in 1984, and since then, continuing technological advancements to improve gene transfer have led to more rapid, accurate, and efficient generation of transgenic animals. Among the established methods are microinjection, electroporation, lipofection, viral vectors, and gene targeting. Here, we review the history of animal transgenesis, with an emphasis on fish, in conjunction with major developments in genetic engineering over the past few decades. Importantly, spermatogonial stem cell modification and transplantation are two common techniques capable of revolutionizing the generation of transgenic fish. Furthermore, we discuss recent progress and future biotechnological prospects of fish transgenesis, which has strong applications for the aquaculture industry. Indeed, some transgenic fish are already available in the current market, validating continued efforts to improve economically important species with biotechnological advancements.
Collapse
Affiliation(s)
- Fernanda M P Tonelli
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Instituto Nanocell, Divinópolis, MG, Brazil
| | - Samyra M S N Lacerda
- Laboratório de Biologia Celular, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávia C P Tonelli
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Guilherme M J Costa
- Laboratório de Biologia Celular, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Renato de França
- Laboratório de Biologia Celular, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Instituto Nacional de Pesquisas da Amazônia (INPA), Manaus, AM, Brazil.
| | - Rodrigo R Resende
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Instituto Nanocell, Divinópolis, MG, Brazil.
| |
Collapse
|
72
|
Zhou J, Cui H, Lu H, Xu Z, Feng W, Chen L, Jin X, Yang X, Qi Z. Muscle-derived stem cells in peripheral nerve regeneration: reality or illusion? Regen Med 2017. [PMID: 28621200 DOI: 10.2217/rme-2016-0165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Owing to the complicated and time-consuming regenerative process, the repair of injured peripheral nerves depends largely on ongoing stem-cell therapy. Decades ago, researchers successfully isolated and identified muscle-derived stem cells (MDSCs) and discovered their potential for multidifferentiation. MDSCs play an important role in trauma repair associated with neuromuscular and vascular injury by simultaneously promoting tissue regrowth via direct differentiation and systematic secretion under physiological conditions. However, the isolation, culture, induction and application of MDSCs require further methodological analysis before clinical application. In this review, we comprehensively discuss the challenges associated with neural regeneration and reviewed the progress of stem cell based regenerative medicine, in an effort to realize the potential of MDSCs in nerve regeneration.
Collapse
Affiliation(s)
- Jing Zhou
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Haiyan Cui
- Department of Plastic & Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Haibin Lu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Zhuqiu Xu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Weifeng Feng
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Lulu Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Xiaolei Jin
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Xiaonan Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Zuoliang Qi
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
73
|
Shi X, Zhang W, Yin L, Chilian WM, Krieger J, Zhang P. Vascular precursor cells in tissue injury repair. Transl Res 2017; 184:77-100. [PMID: 28284670 PMCID: PMC5429880 DOI: 10.1016/j.trsl.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
Abstract
Vascular precursor cells include stem cells and progenitor cells giving rise to all mature cell types in the wall of blood vessels. When tissue injury occurs, local hypoxia and inflammation result in the generation of vasculogenic mediators which orchestrate migration of vascular precursor cells from their niche environment to the site of tissue injury. The intricate crosstalk among signaling pathways coordinates vascular precursor cell proliferation and differentiation during neovascularization. Establishment of normal blood perfusion plays an essential role in the effective repair of the injured tissue. In recent years, studies on molecular mechanisms underlying the regulation of vascular precursor cell function have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches to treat chronic wounds and ischemic diseases in vital organ systems. Verification of safety and establishment of specific guidelines for the clinical application of vascular precursor cell-based therapy remain major challenges in the field.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Weihong Zhang
- Department of Basic Medicine, School of Nursing, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Liya Yin
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Jessica Krieger
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
74
|
Quiescent adult stem cells in murine teeth are regulated by Shh signaling. Cell Tissue Res 2017; 369:497-512. [DOI: 10.1007/s00441-017-2632-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 04/22/2017] [Indexed: 12/17/2022]
|
75
|
Bayraktar S, Jungbluth P, Deenen R, Grassmann J, Schneppendahl J, Eschbach D, Scholz A, Windolf J, Suschek CV, Grotheer V. Molecular- and microarray-based analysis of diversity among resting and osteogenically induced porcine mesenchymal stromal cells of several tissue origin. J Tissue Eng Regen Med 2017; 12:114-128. [PMID: 27966263 PMCID: PMC5811815 DOI: 10.1002/term.2375] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 11/12/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022]
Abstract
Mesenchymal stromal cells (MSCs) play a pivotal role in modern therapeutic approaches in bone‐healing disorders. Although bone marrow‐derived MSCs are most frequently used, the knowledge that many other adult tissues represent promising sources for potent MSCs has gained acceptance. In the present study, the osteogenic differentiation potential of porcine skin fibroblasts (FBs), as well as bone marrow‐ (BMSCs), adipose tissue‐ (ASCs) and dental pulp‐derived stromal cells (DSCs) were evaluated. However, additional application of BMP‐2 significantly elevated the delayed osteogenic differentiation capacity of ASC and FB cultures, and in DSC cultures the supplementation of platelet‐rich plasma increased osteogenic differentiation potential to a comparable level of the good differentiable BMSCs. Furthermore, microarray gene expression performed in an exemplary manner for ASCs and BMSCs revealed that ASCs and BMSCs use different gene expression patterns for osteogenic differentiation under standard media conditions, as diverse MSCs are imprinted dependent from their tissue niche. However, after increasing the differentiation potential of ASCs to a comparable level as shown in BMSCs, a small subset of identical key molecules was used to differentiate in the osteogenic lineage. Until now, the importance of identified genes seems to be underestimated for osteogenic differentiation. Apparently, the regulation of transmembrane protein 229A, interleukin‐33 and the fibroblast growth factor receptor‐2 in the early phase of osteogenic differentiation is needed for optimum results. Based on these results, bone regeneration strategies of MSCs have to be adjusted, and in vivo studies on the osteogenic capacities of the different types of MCSs are warranted. Copyright © 2016 The Authors Tissue Engineering and Regenerative Medicine published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Samet Bayraktar
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Pascal Jungbluth
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - René Deenen
- Biological and Medical Research Center (BMFZ), Genomics and Transcriptomics Laboratory (GTL), Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jan Grassmann
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Johannes Schneppendahl
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Daphne Eschbach
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Giessen and Marburg, Location Marburg, 35033, Marburg, Germany
| | - Armin Scholz
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Windolf
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christoph V Suschek
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Vera Grotheer
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
76
|
From skeletal muscle to stem cells: an innovative and minimally-invasive process for multiple species. Sci Rep 2017; 7:696. [PMID: 28386120 PMCID: PMC5429713 DOI: 10.1038/s41598-017-00803-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 03/14/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow and adipose tissue represent the two most commonly exploited sources of adult mesenchymal stem cells for musculoskeletal applications. Unfortunately the sampling of bone marrow and fat tissue is invasive and does not always lead to a sufficient number of cells. The present study describes a novel sampling method based on microbiopsy of skeletal muscle in man, pigs, dogs and horses. The process includes explant of the sample, Percoll density gradient for isolation and subsequent culture of the cells. We further characterized the cells and identified their clonogenic and immunomodulatory capacities, their immune-phenotyping behavior and their capability to differentiate into chondroblasts, osteoblasts and adipocytes. In conclusion, this report describes a novel and easy-to-use technique of skeletal muscle-derived mesenchymal stem cell harvest, culture, characterization. This technique is transposable to a multitude of different animal species.
Collapse
|
77
|
Pachón-Peña G, Donnelly C, Ruiz-Cañada C, Katz A, Fernández-Veledo S, Vendrell J, Sackstein R. A Glycovariant of Human CD44 is Characteristically Expressed on Human Mesenchymal Stem Cells. Stem Cells 2017; 35:1080-1092. [PMID: 27888602 DOI: 10.1002/stem.2549] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/28/2016] [Accepted: 11/07/2016] [Indexed: 12/26/2022]
Abstract
The clinical effectiveness of systemically administered human mesenchymal stem cells (hMSCs) depends on their capacity to engage vascular endothelium. hMSCs derived from bone marrow (BM-hMSCs) natively lack endothelial binding capacity, but express a CD44 glycovariant containing N-linked sialyllactosamines that can be α(1,3)-fucosylated using fucosyltransferase-VI (FTVI) to enforce sLeX decorations, thereby creating hematopoietic cell E-/L-selectin ligand (HCELL). HCELL expression programs potent shear-resistant adhesion of circulating cells to endothelial beds expressing E-selectin. An alternative source of hMSCs is adipose tissue (A-hMSCs), and we assessed whether A-hMSCs bind E-selectin and/or possess sialyllactosamine-decorated CD44 accessible to α(1,3)-fucosylation. Similar to BM-hMSCs, we found that A-hMSCs natively lack E-selectin ligands, but FTVI-mediated cell surface α(1,3)-fucosylation induces sLeX expression and robust E-selectin binding secondary to conversion of CD44 into HCELL. Moreover, treatment with the α(1,3)-fucosyltransferase-FTVII also generated expression of HCELL on both BM-hMSCs and A-hMSCs, with sLeX decorations created on N-linked glycans of the "standard" CD44 (CD44s) isoform. The finding that hMSCs from both source tissues each lack native E-selectin ligand expression prompted examination of the expression of glycosyltransferases that direct lactosaminyl glycan synthesis. These studies reveal that both types of hMSCs conspicuously lack transcripts encoding α(1,3)-fucosyltransferases, but equally express glycosyltransferases critical to creation of sialyllactosamines. Collectively, these data indicate that assembly of a sialyllactosaminyl-decorated CD44s glycovariant is a conserved feature of hMSCs derived from adipose tissue and marrow, thus identifying a CD44 glycosignature of these cells and supporting the applicability of cell surface α(1,3)-fucosylation in programming migration of systemically administered A-hMSCs to sites of tissue injury/inflammation. Stem Cells 2017;35:1080-1092.
Collapse
Affiliation(s)
- Gisela Pachón-Peña
- Department of Dermatology, and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Conor Donnelly
- Department of Dermatology, and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Catalina Ruiz-Cañada
- Department of Dermatology, and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Adam Katz
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Sonia Fernández-Veledo
- Hospital Universitario de Tarragona Joan XXIII. Institut d'Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Vendrell
- Hospital Universitario de Tarragona Joan XXIII. Institut d'Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Robert Sackstein
- Department of Dermatology, and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
78
|
Wongsupa N, Nuntanaranont T, Kamolmattayakul S, Thuaksuban N. Biological characteristic effects of human dental pulp stem cells on poly-ε-caprolactone-biphasic calcium phosphate fabricated scaffolds using modified melt stretching and multilayer deposition. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:25. [PMID: 28070691 DOI: 10.1007/s10856-016-5833-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/08/2016] [Indexed: 06/06/2023]
Abstract
Craniofacial bone defects such as alveolar cleft affect the esthetics and functions that need bone reconstruction. The advanced techniques of biomaterials combined with stem cells have been a challenging role for maxillofacial surgeons and scientists. PCL-coated biphasic calcium phosphate (PCL-BCP) scaffolds were created with the modified melt stretching and multilayer deposition (mMSMD) technique and merged with human dental pulp stem cells (hDPSCs) to fulfill the component of tissue engineering for bone substitution. In the present study, the objective was to test the biocompatibility and biofunctionalities that included cell proliferation, cell viability, alkaline phosphatase activity, osteocalcin, alizarin red staining for mineralization, and histological analysis. The results showed that mMSMD PCL-BCP scaffolds were suitable for hDPSCs viability since the cells attached and spread onto the scaffold. Furthermore, the constructs of induced hDPSCs and scaffolds performed ALP activity and produced osteocalcin and mineralized nodules. The results indicated that mMSMD PCL-BCP scaffolds with hDPSCs showed promise in bone regeneration for treatment of osseous defects.
Collapse
Affiliation(s)
- Natkrita Wongsupa
- Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai, 90112, Songkhla, Thailand
| | - Thongchai Nuntanaranont
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, 90112, Songkhla, Thailand.
| | - Suttatip Kamolmattayakul
- Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai, 90112, Songkhla, Thailand
| | - Nuttawut Thuaksuban
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, 90112, Songkhla, Thailand
| |
Collapse
|
79
|
Aharony I, Michowiz S, Goldenberg-Cohen N. The promise of stem cell-based therapeutics in ophthalmology. Neural Regen Res 2017; 12:173-180. [PMID: 28400789 PMCID: PMC5361491 DOI: 10.4103/1673-5374.200793] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The promising role of cellular therapies in the preservation and restoration of visual function has prompted intensive efforts to characterize embryonic, adult, and induced pluripotent stem cells for regenerative purposes. Three main approaches to the use of stem cells have been described: sustained drug delivery, immunomodulation, and differentiation into various ocular structures. Studies of the differentiation capacity of all three types of stem cells into epithelial, neural, glial and vascular phenotypes have reached proof-of-concept in culture, but the correction of vision is still in the early developmental stages, and the requirements for effective in vivo implementation are still unclear. We present an overview of some of the preclinical findings on stem-cell rescue and regeneration of the cornea and retina in acute injury and degenerative disorders.
Collapse
Affiliation(s)
- Israel Aharony
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shalom Michowiz
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Neurosurgery, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Nitza Goldenberg-Cohen
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Ophthalmology, Bnai Zion Medical Center, Haifa, Israel
| |
Collapse
|
80
|
Shioda M, Muneta T, Tsuji K, Mizuno M, Komori K, Koga H, Sekiya I. TNFα promotes proliferation of human synovial MSCs while maintaining chondrogenic potential. PLoS One 2017; 12:e0177771. [PMID: 28542363 PMCID: PMC5461123 DOI: 10.1371/journal.pone.0177771] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 05/03/2017] [Indexed: 12/21/2022] Open
Abstract
Synovial mesenchymal stem cells (MSCs) are a candidate cell source for cartilage and meniscus regeneration. If we can proliferate synovial MSCs more effectively, we can expand clinical applications to patients with large cartilage and meniscus lesions. TNFα is a pleiotropic cytokine that can affect the growth and differentiation of cells in the body. The purpose of this study was to examine the effect of TNFα on proliferation, chondrogenesis, and other properties of human synovial MSCs. Passage 1 human synovial MSCs from 2 donors were cultured with 2.5 x 10-12~10-7 g/ml, 10 fold dilution series of TNFα for 14 days, then the cell number and colony number was counted. The effect of the optimum dose of TNFα on proliferation was also examined in synovial MSCs from 6 donors. Chondrogenic potential of synovial MSCs pretreated with TNFα was evaluated in 6 donors. The expressions of 12 surface antigens were also examined in 3 donors.2.5 ng/ml and higher concentration of TNFα significantly increased cell number/dish and cell number/colony in both donors. The effect of 25 ng/ml TNFα was confirmed in all 6 donors. There was no significant difference in the weight, or amount of glycosaminoglycan and DNA of the cartilage pellets between the MSCs untreated and MSCs pretreated with 25 ng/ml TNFα. TNFα decreased expression rate of CD 105 and 140b in all 3 donors. TNFα promoted proliferation of synovial MSCs with increase of cell number/ colony. Pretreatment with TNFα did not affect chondrogenesis of synovial MSCs. However, TNFα affected some properties of synovial MSCs.
Collapse
Affiliation(s)
- Mikio Shioda
- Department of Joint Surgery and Sports Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Muneta
- Department of Joint Surgery and Sports Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuru Mizuno
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichiro Komori
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
81
|
|
82
|
Gugliandolo A, Bramanti P, Mazzon E. Mesenchymal stem cell therapy in Parkinson's disease animal models. Curr Res Transl Med 2016; 65:51-60. [PMID: 28466824 DOI: 10.1016/j.retram.2016.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/14/2016] [Accepted: 10/14/2016] [Indexed: 12/14/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra, and as a consequence, by decreased dopamine levels in the striatum. Currently available therapies are not able to stop or reverse the progression of the disease. A novel therapeutic approach is based on cell therapy with stem cells, in order to replace degenerated neurons. Among stem cells, mesenchymal stem cells seemed the most promising thanks to their capacities to differentiate toward dopaminergic neurons and to release neurotrophic factors. Indeed, mesenchymal stem cells are able to produce different molecules with immunomodulatory, neuroprotective, angiogenic, chemotactic effects and that stimulate differentiation of resident stem cells. Mesenchymal stem cells were isolated for the first time from bone marrow, but can be collected also from adipose tissue, umbilical cord and other tissues. In this review, we focused our attention on mesenchymal stem cells derived from different sources and their application in Parkinson's disease animal models.
Collapse
Affiliation(s)
- A Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - P Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - E Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
83
|
Vasconcellos R, Alvarenga ÉC, Parreira RC, Lima SS, Resende RR. Exploring the cell signalling in hepatocyte differentiation. Cell Signal 2016; 28:1773-88. [DOI: 10.1016/j.cellsig.2016.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 02/08/2023]
|
84
|
Iordache F, Constantinescu A, Andrei E, Amuzescu B, Halitzchi F, Savu L, Maniu H. Electrophysiology, immunophenotype, and gene expression characterization of senescent and cryopreserved human amniotic fluid stem cells. J Physiol Sci 2016; 66:463-476. [PMID: 27053101 PMCID: PMC10717473 DOI: 10.1007/s12576-016-0441-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/12/2016] [Indexed: 01/16/2023]
Abstract
We characterized human amniotic fluid stem cells (AFSC) in senescent cultures (6 weeks) versus cryopreserved cells using whole-cell patch-clamp, immunophenotyping, and differential gene expression profiling for senescence genes. We evidenced five ion current components (outward rectifier, A-type, inward rectifier, and big conductance Ca2+-dependent K+ currents, fast voltage-dependent Na+ currents). Senescent AFSC showed reduced expression of CD90, CD44, CD133, over 500-fold increase of interferon gamma and telomerase reverse transcriptase genes, increased cycle-dependent kinase 4 inhibitors, p53-binding protein 1, and decreased calreticulin and CD44. HLA-ABC immune expression was similar, and HLA-DR expression very low in both cell types. A subset of cryopreserved AFSC featured large inward rectifier K+ currents, voltage-dependent Na+ currents, and neural progenitor markers evidenced by immunophenotyping and RT-PCR. In all AFSC, in both culture conditions, at patch rupture the outward currents were very low, and they increased progressively over several minutes upon cytoplasm dialysis with pipette solution.
Collapse
Affiliation(s)
- Florin Iordache
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| | - Andrei Constantinescu
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| | - Eugen Andrei
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| | - Bogdan Amuzescu
- Department Biophysics and Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095, Bucharest, Romania.
| | - Ferdinand Halitzchi
- Department Biophysics and Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095, Bucharest, Romania
| | - Lorand Savu
- Genetic Lab SRL, Cpt. Av. N. Drossu Str. 9, 012071, Bucharest, Romania
| | - Horia Maniu
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| |
Collapse
|
85
|
Bone Marrow Mesenchymal Stem Cells Enhance the Differentiation of Human Switched Memory B Lymphocytes into Plasma Cells in Serum-Free Medium. J Immunol Res 2016; 2016:7801781. [PMID: 27872867 PMCID: PMC5107863 DOI: 10.1155/2016/7801781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 12/21/2022] Open
Abstract
The differentiation of human B lymphocytes into plasma cells is one of the most stirring questions with regard to adaptive immunity. However, the terminal differentiation and survival of plasma cells are still topics with much to be discovered, especially when targeting switched memory B lymphocytes. Plasma cells can migrate to the bone marrow in response to a CXCL12 gradient and survive for several years while secreting antibodies. In this study, we aimed to get closer to niches favoring plasma cell survival. We tested low oxygen concentrations and coculture with mesenchymal stem cells (MSC) from human bone marrow. Besides, all cultures were performed using an animal protein-free medium. Overall, our model enables the generation of high proportions of CD38+CD138+CD31+ plasma cells (≥50%) when CD40-activated switched memory B lymphocytes were cultured in direct contact with mesenchymal stem cells. In these cultures, the secretion of CXCL12 and TGF-β, usually found in the bone marrow, was linked to the presence of MSC. The level of oxygen appeared less impactful than the contact with MSC. This study shows for the first time that expanded switched memory B lymphocytes can be differentiated into plasma cells using exclusively a serum-free medium.
Collapse
|
86
|
Al Delfi IR, Sheard JJ, Wood CR, Vernallis A, Innes JF, Myint P, Johnson WEB. Canine mesenchymal stem cells are neurotrophic and angiogenic: An in vitro assessment of their paracrine activity. Vet J 2016; 217:10-17. [PMID: 27810198 DOI: 10.1016/j.tvjl.2016.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/10/2016] [Accepted: 09/13/2016] [Indexed: 12/27/2022]
Abstract
Mesenchymal stem cells (MSCs) have been used in cell replacement therapies for connective tissue damage, but also can stimulate wound healing through paracrine activity. In order to further understand the potential use of MSCs to treat dogs with neurological disorders, this study examined the paracrine action of adipose-derived canine MSCs on neuronal and endothelial cell models. The culture-expanded MSCs exhibited a MSC phenotype according to plastic adherence, cell morphology, CD profiling and differentiation potential along mesenchymal lineages. Treating the SH-SY5Y neuronal cell line with serum-free MSC culture-conditioned medium (MSC CM) significantly increased SH-SY5Y cell proliferation (P <0.01), neurite outgrowth (P = 0.0055) and immunopositivity for the neuronal marker βIII-tubulin (P = 0.0002). Treatment of the EA.hy926 endothelial cell line with MSC CM significantly increased the rate of wound closure in endothelial cell scratch wound assays (P = 0.0409), which was associated with significantly increased endothelial cell proliferation (P <0.05) and migration (P = 0.0001). Furthermore, canine MSC CM induced endothelial tubule formation in EA.hy926 cells in a soluble basement membrane matrix. Hence, this study has demonstrated that adipose-derived canine MSC CM stimulated neuronal and endothelial cells probably through the paracrine activity of MSC-secreted factors. This supports the use of canine MSC transplants or their secreted products in the clinical treatment of dogs with neurological disorders and provides some insight into possible mechanisms of action.
Collapse
Affiliation(s)
- I R Al Delfi
- Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - J J Sheard
- Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - C R Wood
- Department of Biological Sciences, Faculty of Medicine, Dentistry and Life Sciences, University of Chester, Parkgate Road, Chester, Cheshire CH1 4BJ, UK
| | - A Vernallis
- Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - J F Innes
- Veterinary Tissue Bank Ltd, Brynkinalt Business Centre, Wrexham LL14 5NS, UK
| | - P Myint
- Veterinary Tissue Bank Ltd, Brynkinalt Business Centre, Wrexham LL14 5NS, UK
| | - W E B Johnson
- Department of Biological Sciences, Faculty of Medicine, Dentistry and Life Sciences, University of Chester, Parkgate Road, Chester, Cheshire CH1 4BJ, UK.
| |
Collapse
|
87
|
Nitkin CR, Bonfield TL. Concise Review: Mesenchymal Stem Cell Therapy for Pediatric Disease: Perspectives on Success and Potential Improvements. Stem Cells Transl Med 2016; 6:539-565. [PMID: 28191766 PMCID: PMC5442806 DOI: 10.5966/sctm.2015-0427] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/04/2016] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a potentially revolutionary therapy for a wide variety of pediatric diseases, but the optimal cell-based therapeutics for such diversity have not yet been specified. The published clinical trials for pediatric pulmonary, cardiac, orthopedic, endocrine, neurologic, and hematologic diseases provide evidence that MSCs are indeed efficacious, but the significant heterogeneity in therapeutic approaches between studies raises new questions. The purpose of this review is to stimulate new preclinical and clinical trials to investigate these factors. First, we discuss recent clinical trials for pediatric diseases studying MSCs obtained from bone marrow, umbilical cord and umbilical cord blood, placenta, amniotic fluid, and adipose tissue. We then identify factors, some unique to pediatrics, which must be examined to optimize therapeutic efficacy, including route of administration, dose, timing of administration, the role of ex vivo differentiation, cell culture techniques, donor factors, host factors, and the immunologic implications of allogeneic therapy. Finally, we discuss some of the practicalities of bringing cell-based therapy into the clinic, including regulatory and manufacturing considerations. The aim of this review is to inform future studies seeking to maximize therapeutic efficacy for each disease and for each patient. Stem Cells Translational Medicine 2017;6:539-565.
Collapse
Affiliation(s)
- Christopher R. Nitkin
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Tracey L. Bonfield
- Division of Pulmonology, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
88
|
Herzig MC, Cap AP. Challenges in translating mesenchymal stem cell therapies for trauma and critical care. Transfusion 2016; 56:20S-5S. [PMID: 27079318 DOI: 10.1111/trf.13566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maryanne C Herzig
- Coagulation and Blood Research Program, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas
| | - Andrew P Cap
- Coagulation and Blood Research Program, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas
| |
Collapse
|
89
|
Osteogenic signaling on silk-based matrices. Biomaterials 2016; 97:133-53. [DOI: 10.1016/j.biomaterials.2016.04.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/25/2016] [Accepted: 04/20/2016] [Indexed: 12/11/2022]
|
90
|
De Francesco F, Romano M, Zarantonello L, Ruffolo C, Neri D, Bassi N, Giordano A, Zanus G, Ferraro GA, Cillo U. The role of adipose stem cells in inflammatory bowel disease: From biology to novel therapeutic strategies. Cancer Biol Ther 2016; 17:889-98. [PMID: 27414952 DOI: 10.1080/15384047.2016.1210741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel diseases are an increasing phenomenon in western countries and in growing populations. The physiopathology of these conditions is linked to intestinal stem cells homeostasis and regenerative potential in a chronic inflammatory microenvironment. Patients with IBD present an increased risk of developing colorectal cancer (CRC), or colitis associated cancer (CAC). Conventional treatment for IBD target the inflammatory process (and include anti-inflammatory and immunosuppressive drugs) with biological agents emerging as a therapeutic approach for non-responders to traditional therapy. Conventional treatment provides scarce results and present severe complications. The intestinal environment may host incoming stem cells, able to engraft in the epithelial damaged sites and differentiate. Therefore, stem cell therapies represent an emerging alternative in inflammatory bowel diseases, with current investigations on the use of haematopoietic and mesenchymal stem cells, in particular adipose stem cells, apparently fundamental as regenerators and as immune-modulators. Here, we discuss stem cells in intestinal homeostasis and as therapeutic agents for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Francesco De Francesco
- a Multidisciplinary Department of Medical-Surgery and Dental Specialties , School of Medicine and Surgery, Second University of Naples , Italy
| | - Maurizio Romano
- a Multidisciplinary Department of Medical-Surgery and Dental Specialties , School of Medicine and Surgery, Second University of Naples , Italy
| | - Laura Zarantonello
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| | - Cesare Ruffolo
- c Department of Surgery , Regional Center for hpb surgery, Regional Hospital of Treviso , TV , Italy
| | - Daniele Neri
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| | - Nicolò Bassi
- c Department of Surgery , Regional Center for hpb surgery, Regional Hospital of Treviso , TV , Italy
| | - Antonio Giordano
- d Department of Medicine , Surgery and Neuroscience, University of Siena , Siena , Italy.,e Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University , Philadelphia , PA , USA
| | - Giacomo Zanus
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| | - Giuseppe A Ferraro
- a Multidisciplinary Department of Medical-Surgery and Dental Specialties , School of Medicine and Surgery, Second University of Naples , Italy
| | - Umberto Cillo
- b Department of Surgery , Oncology and Gastroenterology, Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital , Padua , Italy
| |
Collapse
|
91
|
Bajek A, Olkowska J, Walentowicz-Sadłecka M, Walentowicz P, Sadłecki P, Grabiec M, Bodnar M, Marszałek A, Dębski R, Porowińska D, Czarnecka J, Kaźmierski Ł, Drewa T. High Quality Independent From a Donor: Human Amniotic Fluid Derived Stem Cells-A Practical Analysis Based on 165 Clinical Cases. J Cell Biochem 2016; 118:116-126. [DOI: 10.1002/jcb.25618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/03/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Anna Bajek
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
| | - Joanna Olkowska
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
| | | | - Paweł Walentowicz
- Department of Obstetrics and Gynecology; Nicolaus Copernicus University; Bydgoszcz 85-168 Poland
| | - Paweł Sadłecki
- Department of Obstetrics and Gynecology; Nicolaus Copernicus University; Bydgoszcz 85-168 Poland
| | - Marek Grabiec
- Department of Obstetrics and Gynecology; Nicolaus Copernicus University; Bydgoszcz 85-168 Poland
| | - Magdalena Bodnar
- Department of Clinical Pathomorphology; Nicolaus Copernicus University; Bydgoszcz 85-094 Poland
| | - Andrzej Marszałek
- Department of Clinical Pathomorphology; Nicolaus Copernicus University; Bydgoszcz 85-094 Poland
| | - Robert Dębski
- Department of Experimental Oncology; Nicolaus Copernicus University; Bydgoszcz 85-094 Poland
| | - Dorota Porowińska
- Department of Biochemistry; Nicolaus Copernicus University; Toruń 87-100 Poland
| | - Joanna Czarnecka
- Department of Biochemistry; Nicolaus Copernicus University; Toruń 87-100 Poland
| | - Łukasz Kaźmierski
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
| | - Tomasz Drewa
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
- Department of Urology; Nicolaus Copernicus Hospital; Toruń 87-100 Poland
| |
Collapse
|
92
|
Maria ATJ, Maumus M, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Adipose-Derived Mesenchymal Stem Cells in Autoimmune Disorders: State of the Art and Perspectives for Systemic Sclerosis. Clin Rev Allergy Immunol 2016; 52:234-259. [DOI: 10.1007/s12016-016-8552-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
93
|
Human Bone Marrow Stromal Cells: A Reliable, Challenging Tool for In Vitro Osteogenesis and Bone Tissue Engineering Approaches. Stem Cells Int 2016; 2016:7842191. [PMID: 27293446 PMCID: PMC4879246 DOI: 10.1155/2016/7842191] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/21/2016] [Indexed: 12/18/2022] Open
Abstract
Adult human bone marrow stromal cells (hBMSC) are important for many scientific purposes because of their multipotency, availability, and relatively easy handling. They are frequently used to study osteogenesis in vitro. Most commonly, hBMSC are isolated from bone marrow aspirates collected in clinical routine and cultured under the "aspect plastic adherence" without any further selection. Owing to the random donor population, they show a broad heterogeneity. Here, the osteogenic differentiation potential of 531 hBMSC was analyzed. The data were supplied to correlation analysis involving donor age, gender, and body mass index. hBMSC preparations were characterized as follows: (a) how many passages the osteogenic characteristics are stable in and (b) the influence of supplements and culture duration on osteogenic parameters (tissue nonspecific alkaline phosphatase (TNAP), octamer binding transcription factor 4, core-binding factor alpha-1, parathyroid hormone receptor, bone gla protein, and peroxisome proliferator-activated protein γ). The results show that no strong prediction could be made from donor data to the osteogenic differentiation potential; only the ratio of induced TNAP to endogenous TNAP could be a reliable criterion. The results give evidence that hBMSC cultures are stable until passage 7 without substantial loss of differentiation potential and that established differentiation protocols lead to osteoblast-like cells but not to fully authentic osteoblasts.
Collapse
|
94
|
Pierantozzi E, Vezzani B, Badin M, Curina C, Severi FM, Petraglia F, Randazzo D, Rossi D, Sorrentino V. Tissue-Specific Cultured Human Pericytes: Perivascular Cells from Smooth Muscle Tissue Have Restricted Mesodermal Differentiation Ability. Stem Cells Dev 2016; 25:674-86. [PMID: 26956507 DOI: 10.1089/scd.2015.0336] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Microvascular pericytes (PCs) are considered the adult counterpart of the embryonic mesoangioblasts, which represent a multipotent cell population that resides in the dorsal aorta of the developing embryo. Although PCs have been isolated from several adult organs and tissues, it is still controversial whether PCs from different tissues exhibit distinct differentiation potentials. To address this point, we investigated the differentiation potentials of isogenic human cultured PCs isolated from skeletal (sk-hPCs) and smooth muscle tissues (sm-hPCs). We found that both sk-hPCs and sm-hPCs expressed known pericytic markers and did not express endothelial, hematopoietic, and myogenic markers. Both sk-hPCs and sm-hPCs were able to differentiate into smooth muscle cells. In contrast, sk-hPCs, but not sm-hPCs, differentiated in skeletal muscle cells and osteocytes. Given the reported ability of the Notch pathway to regulate skeletal muscle and osteogenic differentiation, sk-hPCs and sm-hPCs were treated with N-[N-(3,5- difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), a known inhibitor of Notch signaling. DAPT treatment, as assessed by histological and molecular analysis, enhanced myogenic differentiation and abolished osteogenic potential of sk-hPCs. In contrast, DAPT treatment did not affect either myogenic or osteogenic differentiation of sm-hPCs. In summary, these results indicate that, despite being isolated from the same anatomical niche, cultured PCs from skeletal muscle and smooth muscle tissues display distinct differentiation abilities.
Collapse
Affiliation(s)
| | - Bianca Vezzani
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Margherita Badin
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Carlo Curina
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Filiberto Maria Severi
- 2 Division of Obstetrics and Gynecology, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Felice Petraglia
- 2 Division of Obstetrics and Gynecology, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Davide Randazzo
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Daniela Rossi
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | | |
Collapse
|
95
|
Human adipose mesenchymal stem cells as potent anti-fibrosis therapy for systemic sclerosis. J Autoimmun 2016; 70:31-9. [PMID: 27052182 DOI: 10.1016/j.jaut.2016.03.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Displaying immunosuppressive and trophic properties, mesenchymal stem/stromal cells (MSC) are being evaluated as promising therapeutic options in a variety of autoimmune and degenerative diseases. Although benefits may be expected in systemic sclerosis (SSc), a rare autoimmune disease with fibrosis-related mortality, MSC have yet to be evaluated in this specific condition. While autologous approaches could be inappropriate because of functional alterations in MSC from patients, the objective of the present study was to evaluate allogeneic and xenogeneic MSC in the HOCl-induced model of diffuse SSc. We also questioned the source of human MSC and compared bone marrow- (hBM-MSC) and adipose-derived MSC (hASC). METHODS HOCl-challenged BALB/c mice received intravenous injection of BM-MSC from syngeneic BALB/c or allogeneic C57BL/6 mice, and xenogeneic hBM-MSC or hASC (3 donors each). Skin thickness was measured during the experiment. At euthanasia, histology, immunostaining, collagen determination and RT-qPCR were performed in skin and lungs. RESULTS Xenogeneic hBM-MSC were as effective as allogeneic or syngeneic BM-MSC in decreasing skin thickness, expression of Col1, Col3, α-Sma transcripts, and collagen content in skin and lungs. This anti-fibrotic effect was not associated with MSC migration to injured skin or with long-term MSC survival. Interestingly, compared with hBM-MSC, hASC were significantly more efficient in reducing skin fibrosis, which was related to a stronger reduction of TNFα, IL1β, and enhanced ratio of Mmp1/Timp1 in skin and lung tissues. CONCLUSIONS Using primary cells isolated from 3 murine and 6 human individuals, this preclinical study demonstrated similar therapeutic effects using allogeneic or xenogeneic BM-MSC while ASC exerted potent anti-inflammatory and remodeling properties. This sets the proof-of-concept prompting to evaluate the therapeutic efficacy of allogeneic ASC in SSc patients.
Collapse
|
96
|
Shen Y, Zuo S, Wang Y, Shi H, Yan S, Chen D, Xiao B, Zhang J, Gong Y, Shi M, Tang J, Kong D, Lu L, Yu Y, Zhou B, Duan SZ, Schneider C, Funk CD, Yu Y. Thromboxane Governs the Differentiation of Adipose-Derived Stromal Cells Toward Endothelial Cells In Vitro and In Vivo. Circ Res 2016; 118:1194-207. [PMID: 26957525 DOI: 10.1161/circresaha.115.307853] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 03/08/2016] [Indexed: 12/30/2022]
Abstract
RATIONALE Autologous adipose-derived stromal cells (ASCs) offer great promise as angiogenic cell therapy for ischemic diseases. Because of their limited self-renewal capacity and pluripotentiality, the therapeutic efficacy of ASCs is still relatively low. Thromboxane has been shown to play an important role in the maintenance of vascular homeostasis. However, little is known about the effects of thromboxane on ASC-mediated angiogenesis. OBJECTIVE To explore the role of the thromboxane-prostanoid receptor (TP) in mediating the angiogenic capacity of ASCs in vivo. METHODS AND RESULTS ASCs were prepared from mouse epididymal fat pads and induced to differentiate into endothelial cells (ECs) by vascular endothelial growth factor. Cyclooxygenase-2 expression, thromboxane production, and TP expression were upregulated in ASCs on vascular endothelial growth factor treatment. Genetic deletion or pharmacological inhibition of TP in mouse or human ASCs accelerated EC differentiation and increased tube formation in vitro, enhanced angiogenesis in in vivo Matrigel plugs and ischemic mouse hindlimbs. TP deficiency resulted in a significant cellular accumulation of β-catenin by suppression of calpain-mediated degradation in ASCs. Knockdown of β-catenin completely abrogated the enhanced EC differentiation of TP-deficient ASCs, whereas inhibition of calpain reversed the suppressed angiogenic capacity of TP re-expressed ASCs. Moreover, TP was coupled with Gαq to induce calpain-mediated suppression of β-catenin signaling through calcium influx in ASCs. CONCLUSION Thromboxane restrained EC differentiation of ASCs through TP-mediated repression of the calpain-dependent β-catenin signaling pathway. These results indicate that TP inhibition could be a promising strategy for therapy utilizing ASCs in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Yujun Shen
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Shengkai Zuo
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Yuanyang Wang
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Hongfei Shi
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Shuai Yan
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Di Chen
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Bing Xiao
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Jian Zhang
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Yanjun Gong
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Maohua Shi
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Juan Tang
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Deping Kong
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Luheng Lu
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Yu Yu
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Bin Zhou
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Sheng-Zhong Duan
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Claudio Schneider
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Colin D Funk
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Ying Yu
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.).
| |
Collapse
|
97
|
Hanhan S, Ejzenberg A, Goren K, Saba F, Suki Y, Sharon S, Shilo D, Waxman J, Spitzer E, Shahar R, Atkins A, Liebergall M, Blumenfeld A, Deutsch D, Haze A. Skeletal ligament healing using the recombinant human amelogenin protein. J Cell Mol Med 2016; 20:815-24. [PMID: 26917487 PMCID: PMC4831364 DOI: 10.1111/jcmm.12762] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/22/2015] [Indexed: 12/27/2022] Open
Abstract
Injuries to ligaments are common, painful and debilitating, causing joint instability and impaired protective proprioception sensation around the joint. Healing of torn ligaments usually fails to take place, and surgical replacement or reconstruction is required. Previously, we showed that in vivo application of the recombinant human amelogenin protein (rHAM+) resulted in enhanced healing of the tooth‐supporting tissues. The aim of this study was to evaluate whether amelogenin might also enhance repair of skeletal ligaments. The rat knee medial collateral ligament (MCL) was chosen to prove the concept. Full thickness tear was created and various concentrations of rHAM+, dissolved in propylene glycol alginate (PGA) carrier, were applied to the transected MCL. 12 weeks after transection, the mechanical properties, structure and composition of transected ligaments treated with 0.5 μg/μl rHAM+ were similar to the normal un‐transected ligaments, and were much stronger, stiffer and organized than control ligaments, treated with PGA only. Furthermore, the proprioceptive free nerve endings, in the 0.5 μg/μl rHAM+ treated group, were parallel to the collagen fibres similar to their arrangement in normal ligament, while in the control ligaments the free nerve endings were entrapped in the scar tissue at different directions, not parallel to the axis of the force. Four days after transection, treatment with 0.5 μg/μl rHAM+ increased the amount of cells expressing mesenchymal stem cell markers at the injured site. In conclusion application of rHAM+ dose dependently induced mechanical, structural and sensory healing of torn skeletal ligament. Initially the process involved recruitment and proliferation of cells expressing mesenchymal stem cell markers.
Collapse
Affiliation(s)
- Salem Hanhan
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Ayala Ejzenberg
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Koby Goren
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Faris Saba
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Yarden Suki
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Shay Sharon
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Dekel Shilo
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Jacob Waxman
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Elad Spitzer
- Orthopaedic Department, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Ron Shahar
- Faculty of Agriculture, Kort School of Veterinary Medicine, Hebrew University, Rehovot, Israel
| | - Ayelet Atkins
- Faculty of Agriculture, Kort School of Veterinary Medicine, Hebrew University, Rehovot, Israel
| | - Meir Liebergall
- Orthopaedic Department, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Anat Blumenfeld
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Dan Deutsch
- Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Amir Haze
- Orthopaedic Department, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
98
|
Dias CC, Nogueira-Pedro A, Tokuyama PY, Martins MNC, Segreto HRC, Buri MV, Miranda A, Paredes-Gamero EJ. A synthetic fragment of leptin increase hematopoietic stem cell population and improve its engraftment ability. J Cell Biochem 2016; 116:1334-40. [PMID: 25735790 DOI: 10.1002/jcb.25090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/16/2015] [Indexed: 02/06/2023]
Abstract
Several studies have shown the important actions of cytokine leptin that regulates food intake and energy expenditure. Additionally, the ability to modulate hematopoiesis has also been demonstrated. Previous reports have shown that some synthetic sequences of leptin molecules can activate leptin receptor. Herein, decapeptides encompassing amino acids from positions 98 to 122 of the leptin molecule were constructed to evaluate their effects on hematopoiesis. Among them, the synthetic peptide Lep(110-119)-NH2 (LEP F) was the only peptide that possessed the ability to increase the percentage of hematopoietic stem cells (HSC). Moreover, LEP F also produced an increase of granulocyte/macrophage colony-forming units and activated leptin receptor. Furthermore, LEP F also improves the grafting of HSC in bone marrow, but did not accelerate the recovery of bone marrow after ablation with 5-fluorouracil. These results show that LEP F is a positive modulator of the in vivo expansion of HSC and could be useful in bone marrow transplantation.
Collapse
Affiliation(s)
- Carolina C Dias
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | - Amanda Nogueira-Pedro
- Departmento de Biofísica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | - Paula Yumi Tokuyama
- Departmento de Biofísica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | - Marta N C Martins
- Departmento de Biofísica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | - Helena Regina Comodo Segreto
- Departamento de Oncologia Clínica e Experimental, Universidade Federal de São Paulo, R. Napoleão de Barros, 715, São Paulo, Brazil
| | - Marcus V Buri
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | - Antonio Miranda
- Departmento de Biofísica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | - Edgar J Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo, SP, 04044-020, Brazil.,Centro Interdisciplinar de Investigação Bioquímica, Universidade de Mogi das Cruzes, Mogi das Cruzes, SP, Brazil
| |
Collapse
|
99
|
Uzbas F, May ID, Parisi AM, Thompson SK, Kaya A, Perkins AD, Memili E. Molecular physiognomies and applications of adipose-derived stem cells. Stem Cell Rev Rep 2016; 11:298-308. [PMID: 25504377 DOI: 10.1007/s12015-014-9578-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adipose-derived stromal/stem cells (ASC) are multipotent with abilities to differentiate into multiple lineages including connective tissue and neural cells. Despite unlimited opportunity and needs for human and veterinary regenerative medicine, applications of adipose-derived stromal/stem cells are at present very limited. Furthermore, the fundamental biological factors regulating stemness in ASC and their stable differentiation into other tissue cells are not fully understood. The objective of this review was to provide an update on the current knowledge of the nature and isolation, molecular and epigenetic determinants of the potency, and applications of adipose-derived stromal/stem cells, as well as challenges and future directions. The first quarter of the review focuses on the nature of ASC, namely their definition, origin, isolation and sorting methods and multilineage differentiation potential, often with a comparison to mesenchymal stem cells of bone marrow. Due to the indisputable role of epigenetic regulation on cell identities, epigenetic modifications (DNA methylation, chromatin remodeling and microRNAs) are described broadly in stem cells but with a focus on ASC. The final sections provide insights into the current and potential applications of ASC in human and veterinary regenerative medicine.
Collapse
Affiliation(s)
- F Uzbas
- Helmholtz Zentrum München, Institute of Stem Cell Research, Neuherberg, München, 85764, Germany
| | | | | | | | | | | | | |
Collapse
|
100
|
Rodrigues F, Pezzi A, Laureano Á, Valim V, Zambonato B, Dahmer A, Baggio L, Sehn F, Wilke I, L. da Silva MA, Amorin B. Adipocyte Derived Mesenchymal Stromal Cell and Platelet Lysate: Ideal Cell and Supplement for the Treatment of Immune-Inflammatory Diseases? Cell 2016. [DOI: 10.4236/cellbio.2016.52002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|