Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Genesis ; 62(2): e23592, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38587195

RESUMO

Mesenchymal stem cells (MSCs) derived from fetal membranes (FMs) have the potential to exhibit immunosuppression, improve blood flow, and increase capillary density during transplantation. In the field of medicine, opening up new avenues for disease treatment. Chicken embryo chorioallantoic membrane (CAM), as an important component of avian species FM structure, has become a stable tissue engineering material in vivo angiogenesis, drug delivery, and toxicology studies. Although it has been confirmed that chorionic mesenchymal stem cells (Ch-MSCs) can be isolated from the outer chorionic layer of FM, little is known about the biological characteristics of MSCs derived from chorionic mesodermal matrix of chicken embryos. Therefore, we evaluated the characteristics of MSCs isolated from chorionic tissues of chicken embryos, including cell proliferation ability, stem cell surface antigen, genetic stability, and in vitro differentiation potential. Ch-MSCs exhibited a broad spindle shaped appearance and could stably maintain diploid karyotype proliferation to passage 15 in vitro. Spindle cells were positive for multifunctional markers of MSCs (CD29, CD44, CD73, CD90, CD105, CD166, OCT4, and NANOG), while hematopoietic cell surface marker CD34, panleukocyte marker CD45, and epithelial cell marker CK19 were negative. In addition, chicken Ch-MSC was induced to differentiate into four types of mesodermal cells in vitro, including osteoblasts, chondrocytes, adipocytes, and myoblasts. Therefore, the differentiation potential of chicken Ch-MSC in vitro may have great potential in tissue engineering. In conclusion, chicken Ch-MSCs may be an excellent model cell for stem cell regenerative medicine and chorionic tissue engineering.


Assuntos
Galinhas , Células-Tronco Mesenquimais , Animais , Embrião de Galinha , Membrana Corioalantoide , Diferenciação Celular/fisiologia , Células Cultivadas
2.
Bull Exp Biol Med ; 174(4): 538-543, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36899207

RESUMO

The differentiation potential of individual clones of fibroblast CFU (CFU-F) was studied and the relative expression level of genes was analyzed in the culture of CFU-F from the bone marrow in patients with non-severe and severe forms of aplastic anemia at the onset of the disease. The differentiation potential of CFU-F clones was determined by the relative expression of marker genes using quantitative PCR. In aplastic anemia, the ratio of CFU-F clones with different differentiation potential changes, but the molecular mechanisms of this phenomenon are different in non-severe and severe aplastic anemia. In the culture of CFU-F in non-severe and severe aplastic anemia, the relative expression level of genes associated with the maintenance of the hematopoietic stem cell in the bone marrow niche changes, but the decrease in the expression of immunoregulatory genes occurs in severe form only, which may reflect differences in the pathogenesis of non-severe and severe aplastic anemia.


Assuntos
Anemia Aplástica , Humanos , Anemia Aplástica/genética , Anemia Aplástica/patologia , Medula Óssea/patologia , Células Cultivadas , Células-Tronco Hematopoéticas/patologia , Diferenciação Celular/genética , Gravidade do Paciente , Fibroblastos/patologia , Expressão Gênica/genética , Ensaio de Unidades Formadoras de Colônias
3.
FASEB J ; 34(4): 5740-5753, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32112486

RESUMO

Pluripotent stem cells (PSCs) are important models for analyzing cellular metabolism and individual development. As a hypoxia-inducible factor subunit, HIF-1α plays an important role in maintaining the pluripotency of PSCs under hypoxic conditions. However, the mechanisms underlying the self-renewal and pluripotency maintenance of human induced pluripotent stem cells (hiPSCs) via regulating HIF-1α largely remain elusive. In this study, we found that disrupting the expression of HIF-1α reduced self-renewal and pluripotency of hiPSCs. Additionally, HIF-1α-knockdown led to lower mitochondrial membrane potential (ΔΨm ) and higher reactive oxygen species production in hiPSCs. However, HIF-1α-overexpression increased ATP content in hiPSCs, while the role of HIF-1α-knockdown was opposite. The embryoid body (EB) and teratoma formation assays showed that HIF-1α-knockdown promoted endoderm differentiation and development in vitro and in vivo. In terms of the underlying molecular mechanisms, HIF-1α-knockdown inhibited the expression of Actl6a and histone H3K9ac acetylation (H3K9ac). Actl6a knockdown reduced the expression of H3K9ac and the pluripotency of hiPSCs, and also affected endoderm differentiation. These data suggest that hindering HIF-1α expression causes the changes in mitochondrial properties and metabolic disorders in hiPSCs. Furthermore, HIF-1α affects hiPSC pluripotency, and germ layer differentiation via Actl6a and histone acetylation.


Assuntos
Actinas/metabolismo , Diferenciação Celular , Linhagem da Célula , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Acetilação , Actinas/genética , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Feminino , Histonas/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Processamento de Proteína Pós-Traducional
4.
Cell Mol Life Sci ; 77(2): 253-265, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31468060

RESUMO

Dysregulation of angiogenesis is a phenomenon observed in several disorders such as diabetic foot, critical limb ischemia and myocardial infarction. Mesenchymal stromal cells (MSCs) possess angiogenic potential and have recently emerged as a powerful tool for cell therapy to promote angiogenesis. Although bone marrow-derived MSCs are the primary cell of choice, obtaining them has become a challenge. The placenta has become a popular alternative as it is a highly vascular organ, easily available and ethically more favorable with a rich supply of MSCs. Comparatively, placenta-derived MSCs (PMSCs) are clinically promising due to their proliferative, migratory, clonogenic and immunomodulatory properties. PMSCs release a plethora of cytokines and chemokines key to angiogenic signaling and facilitate the possibility of delivering PMSC-derived exosomes as a targeted therapy to promote angiogenesis. However, there still remains the challenge of heterogeneity in the isolated populations, questions on the maternal or fetal origin of these cells and the diversity in previously reported isolation and culture conditions. Nonetheless, the growing rate of clinical trials using PMSCs clearly indicates a shift in favor of PMSCs. The overall aim of the review is to highlight the importance of this rather poorly understood cell type and emphasize the need for further investigations into their angiogenic potential as an alternative source for therapeutic angiogenesis.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica/fisiologia , Placenta/fisiologia , Animais , Exossomos/fisiologia , Feminino , Humanos , Gravidez
5.
Biochem Biophys Res Commun ; 521(2): 375-382, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31668921

RESUMO

Assessment of differentiation potential is a basic requirement to obtain qualified human pluripotent stem cells (hPSCs). Here, we report a simple differentiation method using fetal bovine serum (FBS) to estimate differentiation potential and propensity of hPSCs. PluriTest using RNA-sequencing showed that cells differentiated after treatment with 5% FBS. Expression patterns of three germ layer markers revealed that cells cultured in Knockout Serum Replacement-containing medium (KSR) with mouse feeder cells had higher differentiation potential than cells cultured in a chemically defined medium (E8) with recombinant matrix proteins, especially into the mesoderm and endoderm lineages. Analysis of differentially expressed genes between KSR and E8 identified DUSP6 as a marker for where cells had been cultured. Expression of DUSP6 correlated with FGF-ERK signaling activity. Fine-tuning of FGF-ERK signaling activity to a range that can shut down DUSP6 transcription but sustain NANOG transcription partially increased the differentiation potential. Our data suggest that differentiation with 5% FBS is good to estimate differentiation potential and propensity at the early stage, and that DUSP6 is an excellent marker to monitor ERK signaling activity.


Assuntos
Diferenciação Celular , Fosfatase 6 de Especificidade Dupla/análise , Sistema de Sinalização das MAP Quinases , Células-Tronco Pluripotentes/metabolismo , Soro , Animais , Biomarcadores/análise , Técnicas de Cultura de Células/métodos , Meios de Cultura/farmacologia , Células Alimentadoras , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia
6.
Mol Biol Rep ; 47(5): 3963-3972, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32394306

RESUMO

Heterogeneity of Mesenchymal stem cells (MSCs) imposes limitations for their in vitro expansion and accounts for the lack of reproducibility in some clinical studies. So, this study was designed to isolate and enrich clones of multipotent and self-renewing MSCs from cord blood (CB). Enriched clones with higher proliferation and differentiation potential provide regenerative cells suitable for various clinical demands. MSCA and MSCB original (progenitor) cells were isolated from CB samples, and single cells were cloned by limiting dilution method, in mouse embryonic fibroblast conditioned media. Original MSCs and their single-cell derived clones were characterized by identifying their proliferation rate, immunophenotyping of surface antigens, expression of pluripotency and proliferation genes (Oct4, Sox2, Nanog, KLF4, c-Myc, and PDGFRA), and differentiation potential into multiple lineages (osteogenic, adipogenic, and chondrogenic). Some single-cell clones of MSCA showed a higher proliferation rate and greater differentiation potential than their original cells. However, original MSCB cells were of greater proliferation and differentiation potential than their derived single-cell clones, except for one clone which had comparable results. Cloning of MSCs was attainable when cultured in mouse embryonic fibroblast conditioned media. Single clones with higher proliferation and differentiation potential than their original progenitor cells were obtained by cloning of poorly functioning MSCs progenitor cells, enabling the selection of more therapeutically efficacious MSCs with better performance in clinical applications. Moreover, this study draws attention to the importance of CD105 as a possible MSCs biomarker associated with the multilineage commitment of MSCs.


Assuntos
Clonagem Molecular/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Adipogenia/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Condrogênese/fisiologia , Sangue Fetal/citologia , Humanos , Imunofenotipagem , Fator 4 Semelhante a Kruppel , Reprodutibilidade dos Testes , Cordão Umbilical/citologia
7.
J Cell Biochem ; 120(8): 13881-13892, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30983000

RESUMO

Bone marrow stromal cells (BMSCs) are a unique population of multipotent cells that exhibit pluripotent properties to a certain extent and are significantly heterogeneous in terms of the cell population. We isolate a small cell subpopulation from bovine BMSCs, bovine small stem cells (bSSCs), and herein characterize their properties. The bSSCs are smaller in size and express nuclear Oct-4 and other pluripotency markers. In addition, when cultured in suspension conditions, bSSCs form three-dimensional spheres and display a strong capability for self-renewal and differentiation into cells from three germ layers. Notably, bSSCs display neural features with Sox1 and Pax6 expression. Using bSSCs as donor nuclear cells for somatic cell nuclear transfer, we further demonstrate that the developmental potential of cloned embryos in vitro is significantly increased. Our study identifies a new bovine bone marrow stromal cell-derived stem cell subtype that could have broad importance for developmental biology as well as great potential for regenerative medicine.


Assuntos
Tamanho Celular , Células-Tronco Mesenquimais/citologia , Animais , Apoptose , Biomarcadores/metabolismo , Blastocisto/citologia , Bovinos , Contagem de Células , Diferenciação Celular , Proliferação de Células , Autorrenovação Celular , Separação Celular , Clonagem de Organismos , Embrião de Mamíferos/citologia , Células-Tronco Mesenquimais/metabolismo , Placa Neural/citologia , Fator 3 de Transcrição de Octâmero/metabolismo
8.
Biochem Biophys Res Commun ; 516(1): 307-312, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31256938

RESUMO

As a novel type of mesenchymal stem cell, induced pluripotent stem cell-derived mesenchymal stem cells (iPMSCs) have huge potential for cell therapy. iPMSCs exhibited the typical characteristics of MSCs, whereas the tri-lineage differentiation potential is limited, especially the adipogenic propensity. Here, to reveal the molecular mechanism we carried out the epigenetic comparisons between the iPMSCs and the bone marrow-derived mesenchymal stem cells (BMSCs) and embryonic stem cell-derived mesenchymal stem cells (EMSCs). We found that the iPMSCs was significantly higher than the BMSCs in terms of genome-wide DNA methylation. Meanwhile, the adipogenic gene PPARγ promoter region existed hypermethylation. In addition, compared with EMSCs and BMSCs, iPMSCs had significant differences in the histones epigenetic modification of methylation and acetylation, especially high levels of histone 27 lysine trimethylation (H3K27me3). Furthermore, the epigenetic modifiers Decitabine and EPZ6438 effectively upregulated the gene expression of PPARγ and promoted the adipogenic differentiation of iPMSCs via chromatin remodeling. Taken together, our findings set new metrics to the applications for improving the efficiency and the therapeutic potential of iPMSCs.


Assuntos
Adipogenia/efeitos dos fármacos , Benzamidas/farmacologia , Decitabina/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Piridonas/farmacologia , Animais , Compostos de Bifenilo , Células Cultivadas , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Morfolinas
9.
Cytotherapy ; 21(1): 32-40, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30447901

RESUMO

BACKGROUND AIMS: Multipotent mesenchymal stromal cell (MSC)-based medicines are extensively investigated for use in regenerative medicine and immunotherapy applications. The International Society for Cell and Gene Therapy (ISCT) proposed a panel of cell surface molecules for MSC identification that includes human leukocyte antigen (HLA)-DR as a negative marker. However, its expression is largely unpredictable despite production under tightly controlled conditions and compliance with current Good Manufacturing Practices. Herein, we report the frequency of HLA-DR expression in 81 batches of clinical grade bone marrow (BM)-derived MSCs and investigated its impact on cell attributes and culture environment. METHODS: The levels of 15 cytokines (interleukin [IL]-1ß, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21, IL-22, IL-23, IL-25, IL-31, IL-33, interferon-γ, soluble CD40 ligand and tumor necrosis factor-α) were determined in sera supplements and supernatants of BM-MSC cultures. Identity, multipotentiality and immunopotency assays were performed on high (>20% of cells) and low (≤20% of cells) HLA-DR+ cultures. RESULTS: A correlation was found between HLA-DR expression and levels of IL-17F and IL-33. Expression of HLA-DR did neither affect MSC identity, in vitro tri-lineage differentiation potential (into osteogenic, chondrogenic and adipogenic lineages), nor their ability to inhibit the proliferation of stimulated lymphocytes. DISCUSSION: Out of 81 batches of BM-MSCs for autologous use analyzed, only three batches would have passed the ISCT criteria (<2%), whereas 60.5% of batches were compliant with low HLA-DR values (≤20%). Although a cause-effect relationship cannot be drawn, we have provided a better understanding of signaling events and cellular responses in expansion culture conditions relating with HLA-DR expression.


Assuntos
Antígenos HLA-DR/imunologia , Interleucina-17/sangue , Interleucina-33/sangue , Células-Tronco Mesenquimais/imunologia , Cultura Primária de Células/métodos , Adipogenia , Biomarcadores/metabolismo , Medula Óssea/imunologia , Diferenciação Celular/fisiologia , Células Cultivadas , Condrogênese , Humanos , Ativação Linfocitária , Transplante de Células-Tronco Mesenquimais , Osteogênese
10.
Ultrastruct Pathol ; 43(6): 261-272, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31589079

RESUMO

Mesenchymal stem cells (MSCs) share many properties with other tissue stromal cells, including cell morphology, immunophenotype, differentiation and immunologic properties. In this study, we compared the immunophenotype and the differentiation potential of human vascular wall mesenchymal stem cells (hVW-MSCs) with those of human dermal fibroblasts and myofibroblasts. Cell morphology and surface markers were evaluated by immunofluorescence and flow cytometry; functional assays for immunomodulation, angiogenesis, adipogenesis and osteogenesis were performed, together with the mRNA analysis of the critical differentiation genes. hVW-MSCs, dermal fibroblasts and myofibroblasts were all negative to CD34, whereas the expression of CD44 stemness marker was more intense in hVW-MSCs. As expected, hVW-MSC plasticity was wide and the angiogenic, adipogenic, osteogenic features were confirmed. Fibroblasts were the less effective in terms of immunomodulation, angiogenesis and adipogenic differentiation; differently from fibroblasts, the myofibroblasts showed a poor angiogenic commitment. The mineralization assay was positive in all the three cell types, but ultrastructure interestingly evidenced differential osteogenic patterns among them. Our study supports the higher anti-inflammatory and wound healing repair features of hVW-MSCs, in comparison to the other stromal cells investigated. Moreover, we underline the importance of ultrastructure for investigating the specific osteogenic pattern for each cell type.


Assuntos
Diferenciação Celular/fisiologia , Fibroblastos/citologia , Células-Tronco Mesenquimais/citologia , Miofibroblastos/citologia , Artérias/citologia , Células Cultivadas , Humanos , Osteogênese/fisiologia , Pele/citologia
11.
Int J Mol Sci ; 20(19)2019 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-31623314

RESUMO

We aimed to immortalize primarily isolated human deciduous tooth-derived dental pulp cells (HDDPCs) by transfection with piggyBac (PB)-based transposon vectors carrying E7 from human papilloma virus 16 or complementary DNA (cDNA) encoding human telomerase reverse transcriptase (hTERT). HDDPCs were co-transfected with pTrans (conferring PB transposase expression) + pT-pac (conferring puromycin acetyltransferase expression) + pT-tdTomato (conferring tdTomato cDNA expression) and pT-E7 (conferring E7 expression) or pTrans + pT-pac + pT-EGFP (conferring enhanced green fluorescent protein cDNA expression) + pT-hTERT (conferring hTERT expression). After six days, these cells were selected in medium containing 5 µg/mL puromycin for one day, and then cultured in normal medium allowing cell survival. All resultant colonies were harvested and propagated as a pool. Stemness and tumorigenic properties of the established cell lines ("MT_E7" for E7 and "MT_hTERT" for hTERT) with untransfected parental cells (MT) were examined. Both lines exhibited proliferation similar to that of MT, with alkaline phosphatase activity and stemness-specific factor expression. They displayed differentiation potential into multi-lineage cells with no tumorigenic property. Overall, we successfully obtained HDDPC-derived immortalized cell lines using a PB-based transfection system. The resultant and parental cells were indistinguishable. Thus, E7 and hTERT could immortalize HDDPCs without causing cancer-associated changes or altering phenotypic properties.


Assuntos
Diferenciação Celular , Elementos de DNA Transponíveis , Polpa Dentária/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Diferenciação Celular/genética , Linhagem Celular Transformada , Transformação Celular Neoplásica , Feminino , Vetores Genéticos/genética , Humanos , Proteínas Oncogênicas Virais/genética , Células-Tronco/patologia , Telomerase/genética , Telomerase/metabolismo , Dente Decíduo , Transfecção
12.
Cryobiology ; 75: 7-14, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28284665

RESUMO

Lung mesenchymal stem cells (L-MSCs) characterized by plasticity, reduced relative immune privilege and high anti-fibrosis characteristics play the crucial role in lung tissue regenerative processes. However, up to date, the multi-differentiation potentials and application values of L-MSCs are still uncertain. In the current study, the Small Tailed Han Sheep embryo L-MSCs line from 12 samples, stocking 124 cryogenically-preserved vials, was successfully established by using primary culture and cell cryopreservation techniques. Isolated L-MSCs were morphologically consistent with fibroblasts, could be passaged for at least 18 passages and more than 91.8% of cells were diploid (2n = 54) analyze by G-banding. The majority of cells were in the G0/G1 phase (70.5-91.2%), and the growth curves were all typically sigmoidal. Moreover, L-MSCs were found to express pluripotent genes Oct4, Nanog and MSCs-associated genes ß-integrin, CD29, CD44, CD71, CD73 and CD90, while the expressions of hematopoietic cell markers CD34 and CD45 were negative. In addtion, the L-MSCs could be differentiated into cells of three layers with induction medium in vitro, which confirmed their multilineage differentiation potential. The secretion of urea and ALB showed the differentiated hepatocytes still possessed the detoxification function. These results indicated that the isolated L-MSCs displayed typical characteristics of mesenchymal stem cells and that the culture conditions were suitable for their maintenance of stemness and their proliferation in vitro.


Assuntos
Criopreservação/métodos , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Criopreservação/veterinária , Feminino , Feto , Pulmão , Masculino , Ovinos
13.
Cell Tissue Bank ; 18(4): 513-525, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28255772

RESUMO

Skeletal muscle has a huge regenerative potential for postnatal muscle growth and repair, which mainly depends on a kind of muscle progenitor cell population, called satellite cell. Nowadays, the majority of satellite cells were obtained from human, mouse, rat and other animals but rarely from pig. In this article, the porcine skeletal muscle satellite cells were isolated and cultured in vitro. The expression of surface markers of satellite cells was detected by immunofluorescence and RT-PCR assays. The differentiation capacity was assessed by inducing satellite cells into adipocytes, myoblasts and osteoblasts. The results showed that satellite cells isolated from porcine tibialis anterior were subcultured up to 12 passages and were positive for Pax7, Myod, c-Met, desmin, PCNA and NANOG but were negative for Myogenin. Satellite cells were also induced to differentiate into adipocytes, osteoblasts and myoblasts, respectively. These findings indicated that porcine satellite cells possess similar biological characteristics of stem cells, which may provide theoretical basis and experimental evidence for potential therapeutic application in the treatment of dystrophic muscle and other muscle injuries.


Assuntos
Diferenciação Celular/fisiologia , Separação Celular , Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/citologia , Animais , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Células Cultivadas , Regeneração/fisiologia , Células-Tronco/citologia , Suínos
14.
Cell Tissue Res ; 364(1): 137-47, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26547859

RESUMO

Although adipose-derived stem cells (ADSCs) have many advantageous traits compared with other postnatal stem cells, the consensus is that their differentiation potential must be improved to allow their practical utilization. During the in vitro expansion of human ADSCs (hADSCs), pre-treatment of fibroblast growth factor 2 (FGF2) not only induced an increase of approximately 44-fold in cell number at passage 7 but also augmented the differentiation potential of hADSCs. The effect of FGF2-induced cell preconditioning was evaluated by in vitro and in vivo osteogenesis after pre-treatment with various concentrations of FGF2 (0, 5, 25 ng/ml). FGF2-pre-treated hADSCs showed enhanced in vitro osteogenesis. An evaluation of in vivo osteogenic potential with an ectopic bone model showed that FGF2-preconditioned hADSCs produced an abundant osteoid/bone matrix and the effect was dependent on the concentration of FGF2 pre-treatment; bone matrix formation by control hADSCs was virtually non-existent. FGF2-pre-treated hADSCs also showed enhanced in vitro chondrogenesis, whereas no significant difference was observed in adipogenic potential. Pre-treatment of hADSCs with FGF2 induced an increase in the expression of osteogenic markers such as Cbfa1/Runx2 and alkaline phosphatase and in the expression of ß-catenin. These results suggest that FGF2 plays a highly beneficial role in the preconditioning of ADSCs for musculoskeletal tissue engineering.


Assuntos
Tecido Adiposo/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Osteogênese/efeitos dos fármacos , Células-Tronco/metabolismo , Tecido Adiposo/citologia , Antígenos de Diferenciação/biossíntese , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco/citologia
15.
Stem Cells ; 33(3): 661-73, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25335925

RESUMO

Leukemia inhibitory factor/Stat3 signaling is critical for maintaining the self-renewal and differentiation potential of mouse embryonic stem cells (mESCs). However, the upstream effectors of this pathway have not been clearly defined. Here, we show that periodic tryptophan protein 1 (Pwp1), a WD-40 repeat-containing protein associated with histone H4 modification, is required for the exit of mESCs from the pluripotent state into all lineages. Knockdown (KD) of Pwp1 does not affect mESC proliferation, self-renewal, or apoptosis. However, KD of Pwp1 impairs the differentiation potential of mESCs both in vitro and in vivo. PWP1 chromatin immunoprecipitation-seq results revealed that the PWP1-occupied regions were marked with significant levels of H4K20me3. Moreover, Pwp1 binds to sites in the upstream region of Stat3. KD of Pwp1 decreases the level of H4K20me3 in the upstream region of Stat3 gene and upregulates the expression of Stat3. Furthermore, Pwp1 KD mESCs recover their differentiation potential through suppressing the expression of Stat3 or inhibiting the tyrosine phosphorylation of STAT3. Together, our results suggest that Pwp1 plays important roles in the differentiation potential of mESCs.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HEK293 , Humanos , Camundongos , Proteínas Nucleares/genética , Transdução de Sinais
16.
Cell Biol Int ; 40(10): 1041-9, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27425208

RESUMO

Pulmonary mesenchymal stem cells (PMSCs) have great potential in lung tissue engineering and represent attractive candidates for disease treatment in human and veterinary research. However, a reliable method for isolation and localization of porcine PMSCs in situ is still uncertain. In this study, we successfully isolated PMSCs from Wuzhishan miniature pig embryos in vitro and also attempted to unravel its fundamental differentiation potential and biological characteristics. The isolated PMSCs, which could be cultured and passaged for at least 15 passages, exhibited a typical fibroblast-like morphology and high proliferative potential. Moreover, the PMSCs could express pluripotent marker genes (Oct4 and Nanog) and MSCs-related surface antigens (ß-integrin, CD44, CD71, CD73, CD90, and CD105), while the expressions of CD34 and CD45 were negative. Cell cycle examination showed that the rate of G0/G1 was about 72.1-90.2%. Additionally, the PMSCs not only could be induced to transdifferentiate into mesoblastic cells such as osteoblasts, chondrocytes, and adipocytes in vitro, but also the neural ectoderm and endoderm. Together, these data demonstrate the multiple differentiations potential of PMSCs in vitro, which confers potential use in serving as desirable cell types for lung injury regeneration.


Assuntos
Pulmão/citologia , Células-Tronco Mesenquimais/citologia , Adipócitos/citologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Separação Celular/métodos , Condrócitos/citologia , Fibroblastos/citologia , Imunofluorescência , Pulmão/embriologia , Pulmão/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Suínos , Porco Miniatura , Engenharia Tecidual/métodos
17.
Int J Mol Sci ; 17(7)2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27447618

RESUMO

Regenerative medicine is extensively interested in developing cell therapies using mesenchymal stem cells (MSCs), with applications to several aging-associated diseases. For successful therapies, a substantial number of cells are needed, requiring extensive ex vivo cell expansion. However, MSC proliferation is limited and it is quite likely that long-term culture evokes continuous changes in MSCs. Therefore, a substantial proportion of cells may undergo senescence. In the present review, we will first present the phenotypic characterization of senescent human MSCs (hMSCs) and their possible consequent functional alterations. The accumulation of oxidative stress and dysregulation of key differentiation regulatory factors determine decreased differentiation potential of senescent hMSCs. Senescent hMSCs also show a marked impairment in their migratory and homing ability. Finally, many factors present in the secretome of senescent hMSCs are able to exacerbate the inflammatory response at a systemic level, decreasing the immune modulation activity of hMSCs and promoting either proliferation or migration of cancer cells. Considering the deleterious effects that these changes could evoke, it would appear of primary importance to monitor the occurrence of senescent phenotype in clinically expanded hMSCs and to evaluate possible ways to prevent in vitro MSC senescence. An updated critical presentation of the possible strategies for in vitro senescence monitoring and prevention constitutes the second part of this review. Understanding the mechanisms that drive toward hMSC growth arrest and evaluating how to counteract these for preserving a functional stem cell pool is of fundamental importance for the development of efficient cell-based therapeutic approaches.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Senescência Celular/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Humanos
18.
Exp Dermatol ; 23(6): 395-400, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24689514

RESUMO

Human adipose-derived stroma cells (ADSCs) have successfully been employed in explorative therapeutic studies. Current evidence suggests that ADSCs are unevenly distributed in subcutaneous adipose tissue; therefore, the anatomical origin of ADSCs may influence clinical outcomes. This study was designed to investigate proliferation and differentiation capacities of ADSCs from the gluteal and abdominal depot of 8 females. All had normal BMI (22.01 ± 0.39 kg/m(2) ) and waist circumference (81.13 ± 2.33 cm). Examination by physicians and analysis of 31 laboratory parameters did not reveal possibly confounding medical disorders. Gluteal and abdominal adipose tissue was sampled by en bloc resection on day 7 (±1) after the last menses. Histological examination did not reveal significant depot-specific differences. As assessed by BrdU assay, proliferation of cells from both depots was similar after 24 h and analysis of 15 cell surface markers by flow cytometry identified the isolated cells as ADSCs, again without depot-specific differences. ADSCs from both depots differentiated poorly to chondroblasts. Gluteal ADSCs displayed significantly higher adipogenic differentiation potential than abdominal cells. Osteogenic differentiation was most pronounced in gluteal cells, whereas differentiation of abdominal ADSCs was severely impaired. Our data demonstrate a depot-specific difference in ADSC differentiation potential with abdominal cells failing to meet the criteria of multipotent ADSCs. This finding should be taken into account in future explorations of ADSC-derived therapeutic strategies.


Assuntos
Abdome , Adipogenia/fisiologia , Nádegas , Osteogênese/fisiologia , Células Estromais/citologia , Células Estromais/fisiologia , Gordura Subcutânea/citologia , Adipócitos/citologia , Adipócitos/fisiologia , Adulto , Biópsia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Humanos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/fisiologia , Osteoblastos/citologia , Osteoblastos/fisiologia
19.
J Nutr ; 144(12): 1935-42, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25320190

RESUMO

BACKGROUND: Optimizing calcium nutrition to maximize bone accretion during growth to prevent fragility fractures later in life has spurred greater interest in calcium nutrition in neonates. OBJECTIVE: The aim of this study was to determine the effect of dietary calcium, from deficiency through excess, on bone growth, and the in vivo and in vitro behavior of mesenchymal stem cells (MSCs) in neonatal pigs. METHODS: Twenty-four male and female piglets (24 ± 6 h old) were fed either a calcium-deficient [Ca-D; 0.6% Ca on a dry matter (DM) basis], a calcium-adequate diet (Ca-A; 0.9% Ca on a DM basis), or a calcium-excessive diet (Ca-E; 1.3% Ca on a DM basis) for 14 d to assess the impact of dietary calcium on calcium homeostasis and on the behavior of MSCs. RESULTS: Growth rate was not affected by the Ca-E diet, although bone ash content was 16% higher (P < 0.05) and urinary calcium excretion was 5-fold higher, when normalized to creatinine, compared with the Ca-A group at trial completion. Serum parathyroid hormone (PTH) concentrations were elevated (P < 0.05) in Ca-D piglets in comparison with other groups at both 7 and 14 d. In vivo proliferation of MSCs was 30% higher (P < 0.05) in Ca-E piglets than the other groups. MSCs from both Ca-D- and Ca-E-fed piglets had greater adipogenic potential based on increased gene expression (P < 0.05) of peroxisome proliferator-activated receptor γ (Pparg) and adipocyte fatty acid-binding protein (Ap2) than MSCs from Ca-A piglets. Interestingly, only MSCs from Ca-E-fed piglets had greater (P < 0.05) gene expression of lipoprotein lipase (Lpl) during adipocytic differentiation than those from Ca-A piglets. To assess alterations in lineage allocation and priming, the most and least osteogenic (O+ and O-, respectively) and adipogenic (A+ and A-, respectively) colonies from each MSC isolation were selected on the basis of functional staining. The O+ colonies from Ca-D piglets expressed lower (P < 0.05) levels of osteocalcin (OC) mRNA than did those from other groups, whereas the O- colonies from Ca-E piglets expressed higher (P < 0.05) levels of mRNA of Pparg, Ap2, and Lpl than did those from other groups. CONCLUSIONS: Neonatal calcium deficiency appears to reduce the osteogenic priming of MSCs while enlarging a subpopulation of potentially adipogenic cells, and excess dietary calcium appears to allow greater multipotency of MSCs. These programming alterations of MSCs could have long-term consequences for bone health.


Assuntos
Desenvolvimento Ósseo/efeitos dos fármacos , Cálcio da Dieta/sangue , Cálcio/deficiência , Linhagem da Célula/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Adipócitos/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Cálcio da Dieta/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Creatinina/urina , Dieta , Relação Dose-Resposta a Droga , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/efeitos dos fármacos , PPAR gama/genética , PPAR gama/metabolismo , Hormônio Paratireóideo/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos
20.
Stem Cells Transl Med ; 13(3): 293-308, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38173411

RESUMO

Human adipose-derived stem cells (ASCs) have shown immense potential for regenerative medicine. Our previous work demonstrated that chitosan nano-deposited surfaces induce spheroid formation and differentiation of ASCs for treating sciatic nerve injuries. However, the underlying cell fate and differentiation mechanisms of ASC-derived spheroids remain unknown. Here, we investigate the epigenetic regulation and signaling coordination of these therapeutic spheroids. During spheroid formation, we observed significant increases in histone 3 trimethylation at lysine 4 (H3K4me3), lysine 9 (H3K9me3), and lysine 27 (H3K27me3), accompanied by increased histone deacetylase (HDAC) activities and decreased histone acetyltransferase activities. Additionally, HDAC5 translocated from the cytoplasm to the nucleus, along with increased nuclear HDAC5 activities. Utilizing single-cell RNA sequencing (scRNA-seq), we analyzed the chitosan-induced ASC spheroids and discovered distinct cluster subpopulations, cell fate trajectories, differentiation traits, and signaling networks using the 10x Genomics platform, R studio/language, and the Ingenuity Pathway Analysis (IPA) tool. Specific subpopulations were identified within the spheroids that corresponded to a transient reprogramming state (Cluster 6) and the endpoint cell state (Cluster 3). H3K4me3 and H3K9me3 were discovered as key epigenetic regulators by IPA to initiate stem cell differentiation in Cluster 6 cells, and confirmed by qPCR and their respective histone methyltransferase inhibitors: SNDX-5613 (a KMT2A inhibitor for H3K4me3) and SUVi (an SUV39H1 inhibitor for H3K9me3). Moreover, H3K9me3 and HDAC5 were involved in regulating downstream signaling and neuronal markers during differentiation in Cluster 3 cells. These findings emphasize the critical role of epigenetic regulation, particularly H3K4me3, H3K9me3, and HDAC5, in shaping stem cell fate and directing lineage-specific differentiation.


Assuntos
Quitosana , Histonas , Humanos , Histonas/metabolismo , Epigênese Genética , Lisina/metabolismo , Diferenciação Celular , Células-Tronco , Histona Desacetilases
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa