Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Journal of Experimental Hematology ; (6): 1044-1048, 2020.
Article in Chinese | WPRIM | ID: wpr-827164

ABSTRACT

Abstract  Hematopoietic stem cells are able to self-renewal and differentiate to all blood lineages. With the development of new technologies, recent studies have proposed the revised versions of hematopoiesis. In the classical model of hematopoietic differentiation, HSCs were located at the apex of hematopoietic hierarchy. During differentiation process, HSCs progressively lose self-renewal potential to be commited to progenitors with restricted differentiation potential. For instance, HSCs first give rise to multipotent progenitor cells, then produce bipotent and unipotent progenitors, and finally differentiate to mature blood cells. For the differentiation of megakaryocytes, common myeloid progenitors derived from HSCs give rise to megakaryocyte-erythrocyte progenitors and then develop to megakaryocytes. However, recent results show that megakaryocytes can be directly generated from HSCs without multipotent or bipotent phases. Alternatively, platelet-biased HSCs produce megakaryocyte progenitors. In this article, recent advances in the hematopoiesis and megakaryocyte differentiation pathway are reviewed.


Subject(s)
Cell Differentiation , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells , Megakaryocytes , Multipotent Stem Cells
2.
Biomolecules & Therapeutics ; : 25-33, 2019.
Article in English | WPRIM | ID: wpr-719411

ABSTRACT

Mesenchymal stem cells are classified as multipotent stem cells, due to their capability to transdifferentiate into various lineages that develop from mesoderm. Their popular appeal as cell-based therapy was initially based on the idea of their ability to restore tissue because of their differentiation potential in vitro; however, the lack of evidence of their differentiation to target cells in vivo led researchers to focus on their secreted trophic factors and their role as potential powerhouses on regulation of factors under different immunological environments and recover homeostasis. To date there are more than 800 clinical trials on humans related to MSCs as therapy, not to mention that in animals is actively being applied as therapeutic resource, though it has not been officially approved as one. But just as how results from clinical trials are important, so is to reveal the biological mechanisms involved on how these cells exert their healing properties to further enhance the application of MSCs on potential patients. In this review, we describe characteristics of MSCs, evaluate their benefits as tissue regenerative therapy and combination therapy, as well as their immunological properties, activation of MSCs that dictate their secreted factors, interactions with other immune cells, such as T cells and possible mechanisms and pathways involved in these interactions.


Subject(s)
Animals , Humans , Dinoprostone , Homeostasis , Immunomodulation , In Vitro Techniques , Mesenchymal Stem Cells , Mesoderm , Multipotent Stem Cells , Regeneration , Regenerative Medicine , T-Lymphocytes , Toll-Like Receptors
3.
Tissue Engineering and Regenerative Medicine ; (6): 771-779, 2018.
Article in English | WPRIM | ID: wpr-718788

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) are multipotent stem cells that can differentiate into several cell types. In addition, many studies have shown that MSCs modulate the immune response. However, little information is currently available regarding the maintenance of immunomodulatory characteristics of MSCs through passages. Therefore, we investigated and compared cytokine and gene expression levels from adipose (AD) and bone marrow (BM)-derived MSCs relevant to immune modulation from early to late passages. METHODS: MSC immunophenotype, growth characteristics, cytokine expressions, and gene expressions were analyzed. RESULTS: AD-MSCs and BM-MSCs had similar cell morphologies and surface marker expressions from passage 4 to passage 10. Cytokines secreted by AD-MSCs and BM-MSCs were similar from early to late passages. AD-MSCs and BM-MSCs showed similar immunomodulatory properties in terms of cytokine secretion levels. However, the gene expressions of tumor necrosis factor-stimulated gene (TSG)-6 and human leukocyte antigen (HLA)-G were decreased and gene expressions of galectin-1 and -3 were increased in both AD- and BM-MSCs with repeated passages. CONCLUSION: Our study showed that the immunophenotype and expression of immunomodulation-related cytokines of AD-MSCs and BM-MSCs immunomodulation through the passages were not significantly different, even though the gene expressions of both MSCs were different.


Subject(s)
Humans , Bone Marrow , Cytokines , Galectin 1 , Gene Expression , Immunomodulation , Leukocytes , Mesenchymal Stem Cells , Multipotent Stem Cells , Necrosis
4.
International Neurourology Journal ; : S34-S45, 2018.
Article in English | WPRIM | ID: wpr-740030

ABSTRACT

PURPOSE: To evaluate the therapeutic effect of human embryonic stem cell (hESC)-derived multipotent mesenchymal stem cells (M-MSCs) on ketamine-induced cystitis (KC) in rats. METHODS: To induce KC, 10-week-old female rats were injected with 25-mg/kg ketamine hydrochloride twice weekly for 12 weeks. In the sham group, phosphate buffered saline (PBS) was injected instead of ketamine. One week after the final injection of ketamine, the indicated doses (0.25, 0.5, and 1×106 cells) of M-MSCs (KC+M-MSC group) or PBS vehicle (KC group) were directly injected into the bladder wall. One week after M-MSC injection, the therapeutic outcomes were evaluated via cystometry, histological analyses, and measurement of gene expression. Next, we compared the efficacy of M-MSCs at a low dose (1×105 cells) to that of an identical dose of adult bone marrow (BM)-derived MSCs. RESULTS: Rats in the KC group exhibited increased voiding frequency and reduced bladder capacity compared to rats of the sham group. However, these parameters recovered after transplantation of M-MSCs at all doses tested. KC bladders exhibited markedly increased mast cell infiltration, apoptosis, and tissue fibrosis. Administration of M-MSCs significantly reversed these characteristic histological alterations. Gene expression analyses indicated that several genes associated with tissue fibrosis were markedly upregulated in KC bladders. However the expression of these genes was significantly suppressed by the administration of M-MSCs. Importantly, M-MSCs ameliorated bladder deterioration in KC rats after injection of a low dose (1×105) of cells, at which point BM-derived MSCs did not substantially improve bladder function. CONCLUSIONS: This study demonstrates for the first time the therapeutic efficacy of hESC-derived M-MSCs on KC in rats. M-MSCs restored bladder function more effectively than did BM-derived MSCs, protecting against abnormal changes including mast cell infiltration, apoptosis and fibrotic damage.


Subject(s)
Adult , Animals , Female , Humans , Rats , Apoptosis , Bone Marrow , Cystitis , Fibrosis , Gene Expression , Human Embryonic Stem Cells , Ketamine , Mast Cells , Mesenchymal Stem Cells , Multipotent Stem Cells , Pelvic Pain , Urinary Bladder
5.
Einstein (Säo Paulo) ; 16(4): eRB4587, 2018. graf
Article in English | LILACS | ID: biblio-975099

ABSTRACT

ABSTRACT The biology of stem cells is one of the most dynamic and promising fields of the biological sciences, since it is the basis for the development of organisms. Its biological complexity demands efforts from several lines of research aimed mainly at its therapeutic use. Nanotechnology has been emerging as a new field of study, which shows great potential in the treatment of various diseases. This new area of health has been called "Nanomedicine" or "Bionanotechnology", which can be applied in Medicine by transport and drug delivery systems, robotic tools to be used in diagnostic and surgical processes, nanobiomaterials, gene therapies, nanobiomedical devices, among others. Because stem cells and Nanotechnology are two areas of extremely promising science, a new field of study, called "stem cell Nanotechnology", has gradually emerged. In this, Nanotechnology is used to help the stem cells apply their therapeutic potential in the treatment, cure, and repair of the damaged tissues, in an effective and safe way. In this way, stem cell Nanotechnology has generated great interest, since it may result in significant contributions to Regenerative Medicine and tissue engineering. The present work aims to present the state-of-the-art regarding its therapeutic use in Human Medicine.


RESUMO A biologia das células-tronco é um dos campos mais dinâmicos e promissores das ciências biológicas, pois é a base do desenvolvimento dos organismos. Sua complexidade biológica demanda esforços de diversas linhas de pesquisa, visando principalmente à sua utilização terapêutica. A Nanotecnologia surge como um novo campo de estudo, o qual demonstra grande potencial no que tange ao tratamento de diversas doenças. Esta nova área da saúde vem sendo denominada "Nanomedicina" ou "Bionanotecnologia", a qual pode ser aplicada na Medicina por meio da utilização de sistemas de transporte e liberação de fármacos, ferramentas robóticas a serem utilizadas em processos de diagnóstico e cirurgia, nanobiomateriais, terapias gênicas, dispositivos nanobiomédicos, entre outros. Em razão das células-tronco e a Nanotecnologia serem duas áreas da ciência extremante promissoras, um novo campo de estudo, denominado "Nanotecnologia das células-tronco", surge gradativamente. Neste, a Nanotecnologia é utilizada de forma a auxiliar as células-tronco a exercerem seu potencial terapêutico no tratamento, na cura e na reparação dos tecidos lesionados, de forma eficaz e segura. A Nanotecnologia das células-tronco tem gerado grande interesse, podendo resultar em contribuições significativas na Medicina Regenerativa e na engenharia de tecidos. O presente trabalho teve por objetivo apresentar o estado da arte visando à sua utilização terapêutica na Medicina Humana.


Subject(s)
Humans , Nanotechnology/methods , Multipotent Stem Cells , Regenerative Medicine/methods , Adult Stem Cells , Bone Diseases/therapy , Cardiovascular Diseases/therapy , Tissue Engineering/methods , Nanostructures/therapeutic use , Nanomedicine/methods , Neoplasms/therapy , Nervous System Diseases/therapy
6.
Blood Research ; : 138-144, 2018.
Article in English | WPRIM | ID: wpr-714930

ABSTRACT

BACKGROUND: Targeted therapy has revolutionized the management of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL); however, relapse still occurs because of the presence of quiescent stem cells, termed leukemia propagating cells (LPCs). This study aimed to assess the phenotypic diversity of LPCs in adult patients with Ph+ B-Acute ALL (B-ALL) and to assess its prognostic impact. METHODS: Seventy adults with newly diagnosed Ph+ B-ALL were recruited at the Mansoura Oncology Center. Multiparameter flow cytometry studies of mononuclear blast cells for cluster of differentiation (CD)34, CD38, and CD58 were performed. RESULTS: Seventeen patients had blasts with the pattern of LPCs (CD34+CD38−CD58−), while 53 cases had other diverse phenotypic patterns. The rate of complete response was significantly lower in patients with the LPC phenotype (47% vs. 81%, P=0.006). The median time to achieve a complete response was prolonged in patients with the CD34+CD38−CD58− phenotype (48 vs. 32 days, P=0.016). The three-year overall survival was significantly lower in patients with the CD34+CD38−CD58− phenotype (37% vs. 55% respectively, P=0.028). Multivariate analysis showed that the CD34+CD38− CD58− phenotype was an independent risk factor for overall survival. CONCLUSION: The presence of CD34+CD38−CD58− LPCs at diagnosis allows rapid identification of higher risk patients. Risk stratification of these patients is needed to further guide therapy and develop effective LPCs-targeted therapy to improve treatment outcome.


Subject(s)
Adult , Humans , Diagnosis , Flow Cytometry , Leukemia , Multipotent Stem Cells , Multivariate Analysis , Phenotype , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Prognosis , Recurrence , Risk Factors , Stem Cells , Treatment Outcome
7.
Journal of Peking University(Health Sciences) ; (6): 1014-1021, 2018.
Article in Chinese | WPRIM | ID: wpr-941739

ABSTRACT

OBJECTIVE@#Systemic lupus erythematosus (SLE) is an autoimmune disease with multi-organ involvement and several typical autoantibodies. Mesenchymal stem cells (MSC) are multipotent stem cells with low immunogenicity that can differentiate into various kinds of cells, such as bone, cartilage, fat and skin tissue. MSC have immunomodulatory and reparative properties through interactions with immune cells. MSC have been used in the treatment of refractory SLE and lupus nephritis patients for more than ten years. Most clinical studies were self-controlled studies and only a few were randomized controlled trials. The objective of this study was to use meta-analysis method to evaluate the efficacy and safety of MSC treatment in SLE patients.@*METHODS@#The PubMed, Cochrane Library, Wanfang and VIP databases were searched for published randomized controlled trials and self-controlled studies before June 1, 2018. The search terms included the Chinese and English versions of mesenchymal stem cells, Mesenchymal Stromal Cells [Mesh], systemic lupus erythematosus, lupus, Lupus Erythematosus, Systemic [Mesh]. Two authors independently screened the literatures, assessed the quality of the studies and collected data according to the inclusion and exclusion criteria. The endpoints were the SLE disease activity index, 24 h urine protein and complement C3. Meta-analysis was performed with the Revman 5.3 software according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standard.@*RESULTS@#Eight studies involving 213 patients were included and three of the studies were randomized controlled trials with 66 patients involved. The MSC group showed that the SLE disease activity index decreased significantly [standard mean difference (SMD)=-1.76, 95% confidence interval (CI): -2.00 to -1.51, P<0.001), the 24 h urine protein decreased significantly (SMD=-1.74, 95%CI: -2.46 to -1.03, P<0.001), as well as the complement C3 increased significantly (SMD=1.28, 95%CI: 0.93 to 1.62, P<0.001). Four studies reported adverse events including fever, diarrhea and headache during the infusion.@*CONCLUSION@#Current evidences showed that MSC could improve the disease activity, proteinuria and hypocomplementemia in SLE patients. Large scale and high-quality randomized controlled trials are required to validate the efficacy and safety of MSC treatment in SLE patients.


Subject(s)
Humans , Lupus Erythematosus, Systemic/therapy , Lupus Nephritis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Multipotent Stem Cells , Proteinuria/therapy , Randomized Controlled Trials as Topic
8.
Experimental & Molecular Medicine ; : e315-2017.
Article in English | WPRIM | ID: wpr-212087

ABSTRACT

Spermatogonial stem cells (SSCs) are germline stem cells located along the basement membrane of seminiferous tubules in testes. Recently, SSCs were shown to be reprogrammed into multipotent SSCs (mSSCs). However, both the key factors and biological networks underlying this reprogramming remain elusive. Here, we present transcriptional regulatory networks (TRNs) that control cellular processes related to the SSC-to-mSSC reprogramming. Previously, we established intermediate SSCs (iSSCs) undergoing the transition to mSSCs and generated gene expression profiles of SSCs, iSSCs and mSSCs. By comparing these profiles, we identified 2643 genes that were up-regulated during the reprogramming process and 15 key transcription factors (TFs) that regulate these genes. Using the TF-target relationships, we developed TRNs describing how these TFs regulate three pluripotency-related processes (cell proliferation, stem cell maintenance and epigenetic regulation) during the reprogramming. The TRNs showed that 4 of the 15 TFs (Oct4/Pou5f1, Cux1, Zfp143 and E2f4) regulated cell proliferation during the early stages of reprogramming, whereas 11 TFs (Oct4/Pou5f1, Foxm1, Cux1, Zfp143, Trp53, E2f4, Esrrb, Nfyb, Nanog, Sox2 and Klf4) regulated the three pluripotency-related processes during the late stages of reprogramming. Our TRNs provide a model for the temporally coordinated transcriptional regulation of pluripotency-related processes during the SSC-to-mSSC reprogramming, which can be further tested in detailed functional studies.


Subject(s)
Basement Membrane , Cell Proliferation , Epigenomics , Multipotent Stem Cells , Seminiferous Tubules , Stem Cells , Testis , Transcription Factors , Transcriptome
9.
Tissue Engineering and Regenerative Medicine ; (6): 113-121, 2017.
Article in English | WPRIM | ID: wpr-649881

ABSTRACT

Mesenchymal stem cells (MSCs) represent a heterogeneous group of multipotent stem cells that could be found in various somatic tissues. MSCs are defined by molecular and functional features including spindle-shape morphology, adherence to plastic surfaces, expression of specific surface markers and differentiation potential to chondrocytes, adipocytes and osteocytes. The surface markers were proposed to affect the differentiation potential of MSCs by a limited number of studies. Endoglin (CD105) is defined to be a significant marker for osteogenic and chondrogenic differentiation ability of MSCs. Low CD105 expression is associated with increased osteogenic potential while high CD105 expression is correlated with strong chondrogenic potential. Myrtucommulone-A (MC-A) is an active compound with various biological effects on different cell types but its effect on MSC differentiation has not been described yet. In the present study we aimed at investigating the longterm effects of MC-A on hMSCs. MC-A-treatment reduced CD105 expression in distinct human mesenchymal stem cell (hMSC) lines and gave rise to CD105(low) population but did not change CD44, CD90 or CD73 expression. The decrease in CD105 expression reduced the chondrogenic potential of hMSCs subsequently while adipogenic or osteogenic differentiation was not affected dramatically. MC-A-treatment also suppressed the NF-κB p65 activation which might be responsible for the reduced chondrogenic potential. Our findings suggest thatMC-Acould be used to enrichCD105(low)hMSCs without the need for cell sorting or changing culture conditions which could be utilised in targeted differentiation studies.


Subject(s)
Humans , Adipocytes , Chondrocytes , Mesenchymal Stem Cells , Multipotent Stem Cells , Osteocytes , Plastics
10.
The Korean Journal of Physiology and Pharmacology ; : 153-160, 2017.
Article in English | WPRIM | ID: wpr-728584

ABSTRACT

In this study, we aim to determine the in vivo effect of human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs) on neuropathic pain, using three, principal peripheral neuropathic pain models. Four weeks after hUCB-MSC transplantation, we observed significant antinociceptive effect in hUCB-MSC–transplanted rats compared to that in the vehicle-treated control. Spinal cord cells positive for c-fos, CGRP, p-ERK, p-p 38, MMP-9 and MMP 2 were significantly decreased in only CCI model of hUCB-MSCs-grafted rats, while spinal cord cells positive for CGRP, p-ERK and MMP-2 significantly decreased in SNL model of hUCB-MSCs-grafted rats and spinal cord cells positive for CGRP and MMP-2 significantly decreased in SNI model of hUCB-MSCs-grafted rats, compared to the control 4 weeks or 8weeks after transplantation (p<0.05). However, cells positive for TIMP-2, an endogenous tissue inhibitor of MMP-2, were significantly increased in SNL and SNI models of hUCB-MSCs-grafted rats. Taken together, subcutaneous injection of hUCB-MSCs may have an antinociceptive effect via modulation of pain signaling during pain signal processing within the nervous system, especially for CCI model. Thus, subcutaneous administration of hUCB-MSCs might be beneficial for improving those patients suffering from neuropathic pain by decreasing neuropathic pain activation factors, while increasing neuropathic pain inhibition factor.


Subject(s)
Animals , Humans , Rats , Cord Blood Stem Cell Transplantation , Injections, Subcutaneous , Multipotent Stem Cells , Nervous System , Neuralgia , Spinal Cord , Tissue Inhibitor of Metalloproteinase-2 , Umbilical Cord
11.
International Journal of Stem Cells ; : 184-192, 2017.
Article in English | WPRIM | ID: wpr-80752

ABSTRACT

BACKGROUND AND OBJECTIVES: Mesenchymal stem cells (MSCs) are multipotent stem cells that can be isolated from umbilical cords and are therapeutically used because of their ability to differentiate into various types of cells, in addition to their immunosuppressive and anti-inflammatory properties. Fetal bovine serum (FBS), considered as the standard additive when isolating and culturing MSCs, has a major limitation related to its animal origin. Here, we employed a simple and economically efficient protocol to isolate MSCs from human umbilical cord tissues without using digestive enzymes and replacing FBS with umbilical cord blood serum (CBS). METHODS AND RESULTS: MSCs were isolated by culturing umbilical cord pieces in CBS or FBS supplemented media. Expansion and proliferation kinetics of cells isolated by explant method in the presence of either FBS or CBS were measured, with morphology and multi-differentiation potential of expanded cells characterized by flow cytometry, RT-PCR, and immunofluorescence. MSCs maintained morphology, immunophenotyping, multi-differentiation potential, and self-renewal ability, with better proliferation rates for cells cultured in CBS compared to FBS supplement media. CONCLUSIONS: We here present a simple, reliable and efficient method to isolate MSCs from umbilical cord tissues, where cells maintained proliferation, differentiation potential and immunophenotyping properties and could be efficiently expanded for clinical applications.


Subject(s)
Animals , Humans , Fetal Blood , Flow Cytometry , Fluorescent Antibody Technique , Immunophenotyping , Kinetics , Mesenchymal Stem Cells , Methods , Multipotent Stem Cells , Umbilical Cord
12.
Pesqui. vet. bras ; 36(supl.1): 79-88, June 2016. tab, graf
Article in English | LILACS, VETINDEX | ID: lil-798013

ABSTRACT

The mammalian Wharton's jelly of umbilical cord (WJUC) is a promising source of multipotent cells, providing advantages due to ethical implications, ease of collection and the absence of teratomas in pre-clinical trials. Ovine multipotent cells have already been isolated from various tissues, however there are no reports using umbilical cords in this species. This study aimed to investigate the best medium to transport the umbilical cord, to isolate and maintain ovine WJUC cells and to compare in vitro growth and mesodermal differentiation potential. Eight ovine umbilical cords were obtained during parturition, sectioned and transported in six different media: MEM, low glucose DMEM, M199, RPMI 1640, PBS and saline. For each transportation medium, four culture media were used and the tissue was explanted in 24-well plates and cultured in MEM, low glucose DMEM, M199 and RPMI 1640, all with 10% FBS. Every experiment was conducted with low-passage (P2), investigating MTT viability during four days and adipogenic, chondrogenic and osteogenesis differentiation was induced in vitro. The most effective transport medium (p<0.1) was low glucose DMEM. There was no bacterial or fungal contamination from collection. Cells from Wharton's jelly of ovine umbilical cords collected at natural birth possess fibroblastic morphology and the capacity for in vitro differentiation into adipogenic, chondrogenic and osteogenic cell lines. MTT tests and in vitro differentiation experiments revealed that cell culture medium modulates the behavior of cells and is an important factor for proliferation and maintenance of multipotency. Low glucose DMEM was the most suitable medium for the isolation of cells from Wharton's jelly of ovine umbilical cord.(AU)


A geleia de Wharton do cordão umbilical (GWCU) de mamíferos é uma fonte promissora de células multipotentes, sendo vantajosa por aspectos éticos, facilidade de coleta e não causar teratomas em ensaios pré-clínicos. Em ovinos, células multipotentes já foram isoladas de vários tecidos, no entanto, não existem relatos do isolamento a partir do cordão umbilical nesta espécie. O objetivo do presente estudo foi investigar o melhor meio para o transporte do cordão umbilical, isolar e manter as células da GWCU ovino em diferentes meios e comparar a proliferação e o potencial de diferenciação mesodermal in vitro. Oito cordões umbilicais foram obtidos, por ocasião do parto natural, seccionados e transportados em seis diferentes meios que consistiram em MEM, DMEM baixa glicose, M199, RPMI 1640, PBS e soro fisiológico. Para cada meio de transporte foram utilizados quatro meios de cultivo, sendo o tecido explantado em placas de 24 poços e cultivados em MEM, DMEM baixa glicose, M199 e RPMI 1640, todos com 10% SFB. Todo o experimento foi realizado em baixa passagem (P2) investigando viabilidade pelo MTT por quatro dias além da indução à diferenciação adipogênica, condrogênica e osteogênica in vitro. O meio de transporte mais efetivo (P<0,10) foi o DMEM baixa glicose. Não houve contaminações bacterianas ou fúngicas decorrentes da coleta. Células oriundas da geleia de Wharton do cordão umbilical ovino colhido por ocasião do parto natural possuem morfologia fibroblastóide e capacidade de diferenciação in vitro nas linhagens adipogênica, condrogênica e osteogênica. Os ensaios de MTT e diferenciação in vitro, revelaram que o meio de cultura celular modula o comportamento destas células, sendo um fator importante tanto para a proliferação como para a manutenção da multipotência, destacando o DMEM baixa glicose como o meio mais adequado para o transporte e isolamento de células da geleia de Wharton do cordão umbilical ovino.(AU)


Subject(s)
Animals , Multipotent Stem Cells , Sheep , Umbilical Cord , Wharton Jelly , Adipogenesis , Chondrogenesis , Osteogenesis
13.
Rev. Ateneo Argent. Odontol ; 55(1): 69-70, 2016. ilus
Article in Spanish | LILACS | ID: lil-794296

ABSTRACT

La utilización de células indiferenciadas embrionarias y de células diferenciadas inducidas para que se comporten como las anteriores permite dar origen adiferentes tejidos que pueden ser usados en medicina reconstructiva en reemplazo de los deteriorados...


Subject(s)
Humans , Multipotent Stem Cells/physiology , Pluripotent Stem Cells/physiology , Totipotent Stem Cells/physiology , Stem Cells/physiology , Plastic Surgery Procedures/methods , Mesenchymal Stem Cells/physiology , Fetal Stem Cells/physiology , Tissue Engineering/methods
14.
International Journal of Stem Cells ; : 115-123, 2016.
Article in English | WPRIM | ID: wpr-171023

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) from different sources have different characteristics. Moreover, MSCs are not isolated and characterized in Guinea pig for animal model of cell therapy. AIM OF THE WORK: was the isolating of bone marrow MSCs (BM-MSCs) and adipose tissue MSCs (AT-MSCs) from Guinea pig and assessing their characteristics. MATERIAL AND METHODS: In this study, bone marrow and adipose tissue were collected from three Guinea pigs and cultured and expanded through eight passages. BM-MSCs and AT-MSCs at passages 2, 5 and 8 were seeded in 24-well plates in triplicate. Cells were counted from each well 1~7 days after seeding to determine population doubling time (PDT) and cell growth curves. Cells of passage 3 were cultured in osteogenic and adipogenic differentiation media. RESULTS: BM-MSCs and AT-MSCs attached to the culture flask and displayed spindle-shaped morphology. Proliferation rate of AT-MSCs in the analyzed passages was more than BM-MSCs. The increase in the PDT of MSCs occurs with the increase in the number of passages. Moreover, after culture of BM-MSCs and AT-MSCs in differentiation media, the cells differentiated toward osteoblasts and adipocytes as verified by Alizarin Red staining and Oil Red O staining, respectively. CONCLUSION: BM-MSCs and AT-MSCs of Guinea pig could be valuable source of multipotent stem cells for use in experimental and preclinical studies in animal models.


Subject(s)
Animals , Adipocytes , Adipose Tissue , Bone Marrow , Cell- and Tissue-Based Therapy , Guinea Pigs , Guinea , Mesenchymal Stem Cells , Models, Animal , Multipotent Stem Cells , Osteoblasts
15.
Article in English | IMSEAR | ID: sea-162103

ABSTRACT

Introduction: Contrary to its name, synovial sarcoma does not arise from the synovial membrane but from multi-potent stem cells and can present in any part of the body. Very few cases of vulval synovial sarcoma have been reported in the literature; we report on such a presentation. Th ese tumors can present as painless lumps, which must be completely excised to give the best prognosis. Th erefore the diagnosis of synovial sarcoma should always be kept in mind in the management of vulval masses, especially in young patients.


Subject(s)
Adult , Asian People , Female , Humans , Multipotent Stem Cells/diagnosis , Multipotent Stem Cells/therapy , Prognosis , Sarcoma, Synovial/diagnosis , Sarcoma, Synovial/epidemiology , Sarcoma, Synovial/radiotherapy , Vulva/pathology , Vulva/therapy
16.
International Journal of Oral Science ; (4): 205-212, 2015.
Article in English | WPRIM | ID: wpr-290152

ABSTRACT

Mesenchymal stem cells (MSCs) are a promising tool in regenerative medicine due to their capacity to differentiate into multiple lineages. In addition to MSCs isolated from bone marrow (BMSCs), adult MSCs are isolated from craniofacial tissues including dental pulp tissues (DPs) using various stem cell surface markers. However, there has been a lack of consensus on a set of surface makers that are reproducibly effective at isolating putative multipotent dental mesenchymal stem cells (DMSCs). In this study, we used different combinations of surface markers (CD51/CD140α, CD271, and STRO-1/CD146) to isolate homogeneous populations of DMSCs from heterogeneous dental pulp cells (DPCs) obtained from DP and compared their capacity to undergo multilineage differentiation. Fluorescence-activated cell sorting revealed that 27.3% of DPCs were CD51(+)/CD140α(+), 10.6% were CD271(+), and 0.3% were STRO-1(+)/CD146(+). Under odontogenic conditions, all three subsets of isolated DMSCs exhibited differentiation capacity into odontogenic lineages. Among these isolated subsets of DMSCs, CD271(+) DMSCs demonstrated the greatest odontogenic potential. While all three combinations of surface markers in this study successfully isolated DMSCs from DPCs, the single CD271 marker presents the most effective stem cell surface marker for identification of DMSCs with high odontogenic potential. Isolated CD271(+) DMSCs could potentially be utilized for future clinical applications in dentistry and regenerative medicine.


Subject(s)
Adult , Humans , Adult Stem Cells , Cell Biology , Antigens, CD , Antigens, Surface , Biomarkers , CD146 Antigen , Cell Culture Techniques , Cell Differentiation , Physiology , Cell Lineage , Cell Separation , Methods , Cells, Cultured , Chondrogenesis , Physiology , Dental Pulp , Cell Biology , Flow Cytometry , Methods , Integrin alphaV , Mesenchymal Stem Cells , Cell Biology , Multipotent Stem Cells , Cell Biology , Nerve Tissue Proteins , Odontogenesis , Physiology , Receptor, Platelet-Derived Growth Factor alpha , Receptors, Nerve Growth Factor
17.
International Journal of Oral Science ; (4): 213-219, 2015.
Article in English | WPRIM | ID: wpr-290151

ABSTRACT

Mesenchymal stem cell (MSC)-mediated therapy has been shown to be clinically effective in regenerating tissue defects. For improved regenerative therapy, it is critical to isolate homogenous populations of MSCs with high capacity to differentiate into appropriate tissues. The utilization of stem cell surface antigens provides a means to identify MSCs from various tissues. However, few surface markers that consistently isolate highly regenerative MSCs have been validated, making it challenging for routine clinical applications and making it all the more imperative to identify reliable surface markers. In this study, we used three surface marker combinations: CD51/CD140α, CD271, and STRO-1/CD146 for the isolation of homogenous populations of dental mesenchymal stem cells (DMSCs) from heterogeneous periodontal ligament cells (PDLCs). Fluorescence-activated cell sorting analysis revealed that 24% of PDLCs were CD51(+)/CD140α(+), 0.8% were CD271(+), and 2.4% were STRO-1(+)/CD146(+). Sorted cell populations were further assessed for their multipotent properties by inducing osteogenic and chondrogenic differentiation. All three subsets of isolated DMSCs exhibited differentiation capacity into osteogenic and chondrogenic lineages but with varying degrees. CD271(+) DMSCs demonstrated the greatest osteogenic potential with strong induction of osteogenic markers such as DLX5, RUNX2, and BGLAP. Our study provides evidence that surface marker combinations used in this study are sufficient markers for the isolation of DMSCs from PDLCs. These results provide important insight into using specific surface markers for identifying homogenous populations of DMSCs for their improved utilization in regenerative medicine.


Subject(s)
Adult , Humans , Adaptor Proteins, Signal Transducing , Aggrecans , Antigens, CD , Antigens, Surface , CD146 Antigen , Cell Differentiation , Physiology , Cell Lineage , Cell Separation , Methods , Cells, Cultured , Chondrogenesis , Physiology , Collagen Type II , Core Binding Factor Alpha 1 Subunit , Flow Cytometry , Methods , Homeodomain Proteins , Integrin alphaV , Mesenchymal Stem Cells , Cell Biology , Physiology , Multipotent Stem Cells , Cell Biology , Physiology , Nerve Tissue Proteins , Osteogenesis , Physiology , Periodontal Ligament , Cell Biology , Receptor, Platelet-Derived Growth Factor alpha , Receptors, Nerve Growth Factor , SOX9 Transcription Factor , Time Factors , Transcription Factors
18.
Journal of Korean Medical Science ; : 1764-1776, 2015.
Article in English | WPRIM | ID: wpr-164159

ABSTRACT

Adipose tissue stem cells (ADSCs) would be an attractive autologous cell source. However, ADSCs require invasive procedures, and has potential complications. Recently, urine stem cells (USCs) have been proposed as an alternative stem cell source. In this study, we compared USCs and ADSCs collected from the same patients on stem cell characteristics and capacity to differentiate into various cell lineages to provide a useful guideline for selecting the appropriate type of cell source for use in clinical application. The urine samples were collected via urethral catheterization, and adipose tissue was obtained from subcutaneous fat tissue during elective laparoscopic kidney surgery from the same patient (n = 10). Both cells were plated for primary culture. Cell proliferation, colony formation, cell surface markers, immune modulation, chromosome stability and multi-lineage differentiation were analyzed for each USCs and ADSCs at cell passage 3, 5, and 7. USCs showed high cell proliferation rate, enhanced colony forming ability, strong positive for stem cell markers expression, high efficiency for inhibition of immune cell activation compared to ADSCs at cell passage 3, 5, and 7. In chromosome stability analysis, both cells showed normal karyotype through all passages. In analysis of multi-lineage capability, USCs showed higher myogenic, neurogenic, and endogenic differentiation rate, and lower osteogenic, adipogenic, and chondrogenic differentiation rate compared to ADSCs. Therefore, we expect that USC can be an alternative autologous stem cell source for muscle, neuron and endothelial tissue reconstruction instead of ADSCs.


Subject(s)
Humans , Adult Stem Cells/cytology , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Separation , Chromosomal Instability , Colony-Forming Units Assay , Karyotyping , Multipotent Stem Cells/cytology , Subcutaneous Fat, Abdominal/cytology , Transplantation, Autologous , Urine/cytology
19.
Rev. méd. Chile ; 142(8): 1034-1046, ago. 2014. ilus, graf, tab
Article in Spanish | LILACS | ID: lil-728352

ABSTRACT

Available medical therapy is unable to completely prevent or revert the pathological cardiac remodeling secondary to ischemia or other injuries, which is responsible for the development of heart failure. Regenerative medicine through stem cells had an explosive development in the cardiovascular area during the past decade. Stem cells possess the capacity to regenerate, repair or substitute damaged tissue, allowing the reestablishment of its function. Stem cells can also modulate apoptosis, angiogenesis, fibrosis and inflammation, favoring the endogenous regenerative process initiated by the damaged tissue. These capacities have been corroborated in several animal models of cardiovascular diseases with positive results. In humans, therapies with bone marrow mononuclear stem cells, mesenchymal stem cells and cardiac stem cells are safe. Most randomized clinical trials in patients with myocardial infarction or cardiomyopathies of different etiologies have reported benefits on ventricular function, quality of life and even over mortality of treated patients. This article reviews the state of art of stem cell therapy in cardiovascular diseases, focusing on the most common cellular types used in patients with acute myocardial infarction and chronic cardiomyopathies of different etiologies.


Subject(s)
Humans , Cardiovascular Diseases/surgery , Stem Cell Transplantation/methods , Cell Transdifferentiation , Chronic Disease , Heart Diseases/surgery , Multipotent Stem Cells/physiology , Multipotent Stem Cells/transplantation , Myocardial Infarction/surgery
20.
Pesqui. vet. bras ; 33(9): 1151-1154, set. 2013. ilus, graf, tab
Article in English | LILACS | ID: lil-694066

ABSTRACT

The objective of the study was to isolate, cultivate and characterize equine peripheral blood-derived multipotent mesenchymal stromal cells (PbMSCs). Peripheral blood was collected, followed by the isolation of mononuclear cells using density gradient reagents, and the cultivation of adherent cells. Monoclonal mouse anti-horse CD13, mouse anti-horse CD44, and mouse anti-rat CD90 antibodies were used for the immunophenotypic characterization of the surface of the PbMSCs. These cells were also cultured in specific media for adipogenic and chondrogenic differentiation. There was no expression of the CD13 marker, but CD44 and CD90 were expressed in all of the passages tested. After 14 days of cell differentiation into adipocytes, lipid droplets were observed upon Oil Red O (ORO) staining. Twenty-one days after chondrogenic differentiation, the cells were stained with Alcian Blue. Although the technique for the isolation of these cells requires improvement, the present study demonstrates the partial characterization of PbMSCs, classifying them as a promising type of progenitor cells for use in equine cell therapy.


O objetivo deste estudo foi isolar, cultivar e caracterizar as células mesenquimais multipotentes estromais derivadas do sangue periférico (SpCTMs) equino. O sangue periférico foi coletado, seguido do isolamento das células mononucleadas utilizando o reagente de gradiente de densidade e o cultivo das células aderentes. Os anticorpos monoclonais mouse anti-horse CD13, mouse anti-horse CD44 e mouse anti-rat CD90 foram utilizados para a caracterização imunofenotípica da superfície das SpCTMs. Estas células também foram cultivadas utilizando meio de cultura específico para a diferenciação adipogênica e condrogênica. Não houve expressão do marcador CD13, mas os marcadores CD44 e CD90 foram expressos em todas as passagens testadas. Após 14 dias da diferenciação das células em adipócitos, gotículas de lipídeos foram observados através da coloração com Oil Red O. Vinte e um dias após a diferenciação condrogênica, as células foram coradas com o Alcian Blue. Embora a técnica de isolamento destas células necessite ser otimizada, o presente estudo demonstra a caracterização parcial das SpCTMs, classificando-as como um tipo de células progenitoras promissoras para o uso na terapia celular em equinos.


Subject(s)
Animals , Adult , Horses/blood , Mesenchymal Stem Cells/cytology , Blood Cells/classification , Multipotent Stem Cells/physiology , Immunophenotyping/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL