Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Environ Toxicol ; 38(8): 2022-2030, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37163415

RESUMO

Breast cancer is a leading cause of cancer-related death worldwide, and chemoresistance often leads to poor patient outcomes. In this study, we investigated the anticancer activity of synthetic diphenyl disulfide (DPDS) in breast cancer cell lines. DPDS inhibited cellular proliferation and viability in a dose-dependent manner and reduced colony formation, an index of clonogenicity. Annexin-V and 7-AAD double staining showed that DPDS could induce the apoptosis of breast cancer cells. Western blotting of the expression of Bax p21 and its cleaved form p18 suggested the activation of p18 Bax-induced apoptosis. Furthermore, the increased expression of the autophagy marker LC3B-II indicated autophagic lysosome accumulation induced by DPDS. Our findings suggest that DPDS has potential as a candidate for treating breast cancer, and further modifications and optimizations are warranted.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Proteína X Associada a bcl-2 , Neoplasias da Mama/metabolismo , Apoptose , Proliferação de Células , Autofagia , Linhagem Celular Tumoral
2.
Sci Adv ; 8(46): eadc9222, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36383652

RESUMO

Cell fate determination of human mesenchymal stem/stromal cells (hMSCs) is precisely regulated by lineage-specific transcription factors and epigenetic enzymes. We found that CTR9, a key scaffold subunit of polymerase-associated factor complex (PAFc), selectively regulates hMSC differentiation to osteoblasts and chondrocytes, but not to adipocytes. An in vivo ectopic osteogenesis assay confirmed the essentiality of CTR9 in hMSC-derived bone formation. CTR9 counteracts the activity of Enhancer Of Zeste 2 (EZH2), the epigenetic enzyme that deposits H3K27me3, in hMSCs. Accordingly, CTR9 knockdown (KD) hMSCs gain H3K27me3 mark, and the osteogenic differentiation defects of CTR9 KD hMSCs can be partially rescued by treatment with EZH2 inhibitors. Transcriptome analyses identified bone morphology protein-2 (BMP-2) as a downstream effector of CTR9. BMP-2 secretion, membrane anchorage, and the BMP-SMAD pathway were impaired in CTR9 KD MSCs, and the effects were rescued by BMP-2 supplementation. This study uncovers an epigenetic mechanism engaging the CTR9-H3K27me3-BMP-2 axis to regulate the osteochondral lineage differentiation of hMSCs.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Células-Tronco Mesenquimais/metabolismo , Epigênese Genética , Histonas/metabolismo , Diferenciação Celular/genética , Osteoblastos , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo
3.
Cartilage ; 13(2_suppl): 530S-543S, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34467773

RESUMO

OBJECTIVES: Functions of mesenchymal stem/stromal cells (MSCs) are affected by patient-dependent factors such as age and health condition. To tackle this problem, we used the cellular reprogramming technique to epigenetically alter human MSCs derived from the synovial fluid of joints with osteoarthritis (OA) to explore the potential of reprogrammed MSCs for repairing articular cartilage. MATERIALS AND METHODS: MSCs isolated from the synovial fluid of three patients' OA knees (Pa-MSCs) were reprogrammed through overexpression of pluripotency factors and then induced for differentiation to establish reprogrammed MSC (Re-MSC) lines. We compared the in vitro growth characteristics, chondrogenesis for articular cartilage chondrocytes, and immunomodulatory capacity. We also evaluated the capability of Re-MSCs to repair articular cartilage damage in an animal model with spontaneous OA. RESULTS: Our results showed that Re-MSCs increased the in vitro proliferative capacity and improved chondrogenic differentiation toward articular cartilage-like chondrocyte phenotypes with increased THBS4 and SIX1 and decreased ALPL and COL10A1, compared to Pa-MSCs. In addition, Re-MSC-derived chondrocytes expressing elevated COL2A and COL2B were more mature than parental cell-derived ones. The enhancement in chondrogenesis of Re-MSC involves the upregulation of sonic hedgehog signaling. Moreover, Re-MSCs improved the repair of articular cartilage in an animal model of spontaneous OA. CONCLUSIONS: Epigenetic reprogramming promotes MSCs harvested from OA patients to increase phenotypic characteristics and gain robust functions. In addition, Re-MSCs acquire an enhanced potential for articular cartilage repair. Our study here demonstrates that the reprogramming strategy provides a potential solution to the challenge of variation in MSC quality.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Animais , Condrogênese/genética , Proteínas Hedgehog , Proteínas de Homeodomínio , Humanos , Líquido Sinovial
4.
Stem Cells ; 39(1): 62-77, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33252174

RESUMO

Cellular reprogramming forcing the expression of pluripotency markers can reverse aging of cells, but how molecular mechanisms through which reprogrammed cells alter aging-related cellular activities still remains largely unclear. In this study, we reprogrammed human synovial fluid-derived mesenchymal stem cells (MSCs) into induced pluripotent stem cells (iPSCs) using six reprogramming factors and reverted the iPSCs back to MSCs, as an approach to cell rejuvenation. Using the parental and reprogrammed MSCs as control nonrejuvenated and rejuvenated cells, respectively, for comparative analysis, we found that aging-related activities were greatly reduced in reprogrammed MSCs compared with those in their parental lines, indicating reversal of cell aging. Global transcriptome analysis revealed differences in activities of regulatory networks associated with inflammation and proliferation. Mechanistically, we demonstrated that, compared with control cells, the expression of GATA binding protein 6 (GATA6) in reprogrammed cells was attenuated, resulting in an increase in the activity of sonic hedgehog signaling and the expression level of downstream forkhead box P1 (FOXP1), in turn ameliorating cellular hallmarks of aging. Lower levels of GATA6 expression were also found in cells harvested from younger mice or lower passage cultures. Our findings suggest that GATA6 is a critical regulator increased in aged MSCs that controls the downstream sonic hedgehog signaling and FOXP1 pathway to modulate cellular senescence and aging-related activities.


Assuntos
Senescência Celular , Fator de Transcrição GATA6/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Adulto , Animais , Feminino , Fator de Transcrição GATA6/genética , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade
5.
Int J Biol Macromol ; 170: 248-260, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33359806

RESUMO

Functional tendon tissue engineering depends on harnessing the biochemical and biophysical cues of the native tendon extracellular matrix. In this study, we fabricated highly-aligned poly(L-lactic acid) (PLLA) fibers with surfaces decorated by two of the crucial tendon ECM components, type 1 collagen (COL1) and chondroitin sulfate (CS), through a coaxial stable jet electrospinning approach. Effects of the biomimetic COL1-CS (shell)/PLLA (core) fibers on the tenogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro were investigated. Higher rates of cell spreading and proliferation are observed on the aligned COL1-CS/PLLA fibers compared to that on the plain PLLA fibers. Expression of the tendon-associated genes scleraxis (SCX) and COL1 as well as protein tenomodulin (TNMD) are significantly increased. Introduction of mechanical stimulation gives rise to synergistic effect on tenogenic differentiation of hMSCs. Higher expression of TGF-ß2, TGFßR-II, and Smad3 by the cells on the COL1-CS/PLLA fiber substrates are observed, which indicates that COL1-CS/PLLA ultrafine fibers dictate the hMSC tenogenic differentiation through activating the TGF-ß signaling pathway. Animal study in rat Achilles tendon repair model corroborated the promoting role of COL1-CS/PLLA in regenerating a tendon-like tissue. Thus, our highly aligned biomimicking fibers may serve as an efficient scaffolding system for functional tendon regeneration.


Assuntos
Sulfatos de Condroitina/farmacologia , Colágeno/farmacologia , Engenharia Tecidual/métodos , Adulto , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sulfatos de Condroitina/metabolismo , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Matriz Extracelular , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Tendões/citologia , Tendões/fisiologia , Alicerces Teciduais/química
6.
Biochem Biophys Res Commun ; 529(2): 180-185, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32703408

RESUMO

Human mesenchymal stem/stromal cells (hMSCs) reside in a vascularized microenvironment and experience a host of blood vessel secretions, including endothelin-1 (ET1). Previously, our group has demonstrated improved induction of osteogenesis and chondrogenesis in hMSCs through an ET1-induced increase in production of anabolic factors. The current study explores effects of ET1 on catabolic factors secreted by hMSCs during chondrogenesis and osteogenesis. Cell proliferation and extracellular matrix (ECM) deposition were also explored. Our results demonstrated that ET1 reduced mRNA transcript levels of MMP2, MMP13, ADAMTS4, and ADAMTS5 in chondrogenic hMSCs, and MMP13 and ADAMTS5 in osteogenic hMSCs. Furthermore, ET1-treated chondrogenic and osteogenic hMSCs showed more intense stains for Alcian blue and Alizarin red S, respectively, than control cells. Immunocytochemical results demonstrated that the ET1-mediated reduction of MMP13 could be reversed through blocking ET1 induction. Overall, our findings indicate that hMSCs treated with ET1 during chondrogenic or osteogenic induction attenuate catabolic activities of the cell to reduce ECM degradation, suggesting that it may be beneficial to use ET1 to enhance hMSC differentiation and protect newly synthesized ECM from degradation.


Assuntos
Condrogênese , Endotelina-1/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo
7.
Am J Sports Med ; 47(11): 2729-2736, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31339739

RESUMO

BACKGROUND: Despite widespread acceptance of fresh autologous bone marrow (BM) for use in clinical practice, limited information exists to analyze if tendon-to-bone healing could be accelerated with local use of fresh autologous BM. PURPOSE: To investigate the effect of fresh autologous BM on tendon-to-bone healing with a novel rat model. STUDY DESIGN: Controlled laboratory study. METHODS: An extra-articular bone tunnel was created and filled with an autologous tendon graft in skeletally mature Sprague-Dawley rats (N = 60). They were then randomly divided into 3 groups: BM group (injection of fresh autologous BM into the tendon-bone interface, n = 20), BM-derived mesenchymal stem cell (BMSC) group (injection of allogenic cultured BMSCs, n = 20), and the control group (tendon-bone interface without injection of BM or BMSCs, n = 20). Biomechanical, histological, and immunohistochemical analyses were performed at 2 and 6 weeks after surgery. RESULTS: The BM group showed a relatively well-organized and dense connective tissue interface with better orientation of collagen fibers as compared with the BMSC group. At 2 weeks, the tendon-bone interface tissue thickness of the BMSC group was 140 ± 25 µm (mean ± SEM), which was significantly greater than the BM group (58 ± 15 µm). The BM group showed fewer M1 macrophages at the tendon-bone interface at 2 and 6 weeks (P < .001). In contrast, there were more M2 macrophages at the interface in the BM group 2 and 6 weeks postoperatively when compared with controls and the BMSC group (P < .001). Biomechanical tests revealed significantly higher stiffness in the BM group versus the control and BMSC groups at 2 and 6 weeks after surgery (P < .05). Load to failure showed similar trends to stiffness. CONCLUSION: These findings indicate that local delivery of fresh autologous BM enhances tendon-to-bone healing better than the alternative treatments in this study. This effect may be partially due to the observed modulation of inflammatory processes, especially in M2 macrophage polarization. CLINICAL RELEVANCE: Fresh autologous BM could be a treatment option for this disorder.


Assuntos
Transplante de Medula Óssea , Osso e Ossos/cirurgia , Transplante de Células-Tronco Mesenquimais , Tendões/transplante , Cicatrização/fisiologia , Animais , Osso e Ossos/fisiologia , Masculino , Modelos Animais , Distribuição Aleatória , Ratos Sprague-Dawley , Tendões/fisiologia , Transplante Autólogo
8.
Stem Cells Transl Med ; 8(6): 522-534, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30784225

RESUMO

Patients with type 1 diabetes mellitus (T1DM) often suffer from osteopenia or osteoporosis. Although most agree that T1DM-induced hyperglycemia is a risk factor for progressive bone loss, the mechanisms for the link between T1DM and bone loss still remain elusive. In this study, we found that bone marrow-derived mesenchymal stem cells (BMSCs) isolated from T1DM donors were less inducible for osteogenesis than those from non-T1DM donors and further identified a mechanism involving bone morphogenetic protein-6 (BMP6) that was produced significantly less in BMSCs derived from T1DM donors than that in control cells. With addition of exogenous BMP6 in culture, osteogenesis of BMSCs from T1DM donors was restored whereas the treatment of BMP6 seemed not to affect non-T1DM control cells. We also demonstrated that bone mineral density (BMD) was reduced in streptozotocin-induced diabetic mice compared with that in control animals, and intraperitoneal injection of BMP6 mitigated bone loss and increased BMD in diabetic mice. Our results suggest that bone formation in T1DM patients is impaired by reduction of endogenous BMP6, and supplementation of BMP6 enhances osteogenesis of BMSCs to restore BMD in a mouse model of T1DM, which provides insight into the development of clinical treatments for T1DM-assocaited bone loss. Stem Cells Translational Medicine 2019;8:522-534.


Assuntos
Doenças Ósseas Metabólicas/etiologia , Proteína Morfogenética Óssea 6/metabolismo , Diabetes Mellitus Tipo 1/patologia , Animais , Densidade Óssea , Proteína Morfogenética Óssea 6/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/complicações , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese/efeitos dos fármacos , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Microtomografia por Raio-X
9.
FASEB J ; 33(1): 996-1007, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30096039

RESUMO

Blood vessels composed of endothelial cells (ECs) contact with mesenchymal stem cells (MSCs) in different tissues, suggesting possible interaction between these 2 types of cells. We hypothesized that endothelin-1 (ET1), a secreted paracrine factor of ECs, can differentially direct the lineages of adipose-derived stem cells (ASCs) and bone marrow-derived MSCs (BMSCs). Predifferentiated ASCs and BMSCs were treated with ET1 for 2 cell passages and then induced for multilineage differentiation. Our results showed that adipogenesis of ET1-pretreated ASCs and osteogenesis of ET1-pretreated BMSCs were increased compared to those of control cells. The effect of ET1 on enhancing adipogenesis of ASCs and osteogenesis of BMSCs was attenuated by blocking endothelin receptor type A (ETAR) and/or endothelin receptor type B (ETBR). Western blot analysis indicated that regulation by ET1 was mediated through activation of the protein kinase B and ERK1/2 signaling pathways. We analyzed subpopulations of ASCs and BMSCs with or without ETAR and/or ETBR, and we found that ETAR+/ETBR- and ETAR-/ETBR+ subpopulations of ASCs and those of BMSCs pretreated with ET1 were prone to turning into adipocytes and osteoblasts, respectively, after differentiation induction. Our findings provide insight into the differential regulation of MSC specification by ET1, which may help develop viable approaches for tissue regeneration.-Lee, M.-S., Wang, J., Yuan, H., Jiao, H., Tsai, T.-L., Squire, M. W., Li, W.-J. Endothelin-1 differentially directs lineage specification of adipose- and bone marrow-derived mesenchymal stem cells.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Endotelina-1/metabolismo , Células-Tronco/citologia , Adulto , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Masculino , Osteogênese , Receptores de Endotelina/metabolismo
10.
J Biomed Mater Res A ; 106(6): 1595-1603, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29427535

RESUMO

During the process of tissue regeneration facilitated by stem cells, physical properties of a scaffold affect behavior and activities of the cell. To enhance differentiation of human mesenchymal stem cells (MSCs) into endothelial-like cells (ELCs), we used electrospun fibrous substrates with different stiffness to enhance the differentiation. A simple method of annealing with different lengths of treatment time was employed to modulate stiffness of electrospun fibrous substrates without changing their chemistry. We seeded MSCs on substrates with different stiffness to study how stiffness of a culture substrate affects differentiation of MSCs into ELCs. Results of RT-PCR and western blotting revealed that stiffer substrates with the average surface modulus of 7.82 MPa induced differentiated MSCs to express more VEGF, CD31, and vWF mRNA transcripts and proteins than softer ones with that of 3.8 or 1.44 MPa. We also found that the production of macrophage migration inhibitory factor (MIF) in ELCs was increased with substrate stiffness. After silencing MIF mRNA, MSCs during differentiation showed lower expression levels of VEGF, CD31, and vWF than control cells whereas VEGF-silenced and control cells expressed comparable levels of MIF, indicating that MIF is an upstream molecule regulating VEGF in the mechanism. Our findings provide new insight into how stiffness of a culture substrate regulates differentiation of MSCs into ELCs. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1595-1603, 2018.


Assuntos
Materiais Biocompatíveis/química , Diferenciação Celular , Células Endoteliais/citologia , Células-Tronco Mesenquimais/citologia , Alicerces Teciduais/química , Proliferação de Células , Células Cultivadas , Células Endoteliais/metabolismo , Dureza , Humanos , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Nanofibras/química , Nanofibras/ultraestrutura , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Ann N Y Acad Sci ; 1409(1): 3-17, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28984359

RESUMO

Mesenchymal stem cells (MSCs) have been extensively used for cell therapies and tissue engineering. The current MSC strategy requires a large quantity of cells for such applications, which can be achieved through cell expansion in culture. In the body, stem cell fate is largely determined by their microenvironment, known as the niche. The complex and dynamic stem cell niche provides physical, mechanical, and chemical cues to collaboratively regulate cell activities. It remains a great challenge to maintain the properties of MSCs in culture. Constructing a microenvironment as an engineered stem cell niche in culture to maintain MSC phenotypes, properties, and functions is a viable strategy to address the issue. Here, we review the current understanding of MSC behavior in the bone marrow niche, describe different strategies to engineer an in vitro microenvironment for maintaining MSC properties and functions, and discuss previous findings on environmental factors critical to the modulation of MSC activities in engineered microenvironments.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Proliferação de Células , Células-Tronco Mesenquimais/citologia , Células Cultivadas , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Transplante de Células-Tronco Mesenquimais/tendências , Nicho de Células-Tronco , Engenharia Tecidual/métodos , Engenharia Tecidual/tendências
12.
Cancer Res ; 77(22): 6179-6189, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28972074

RESUMO

In response to chemical stimuli from cancer cells, mesenchymal stem cells (MSC) can differentiate into cancer-associated fibroblasts (CAF) and promote tumor progression. How mechanical stimuli such as stiffness of the extracellular matrix (ECM) contribute to MSC phenotype in cancer remains poorly understood. Here, we show that ECM stiffness leads to mechano-signal transduction in MSC, which promotes mammary tumor growth in part through secretion of the signaling protein prosaposin. On a stiff matrix, MSC cultured with conditioned media from mammary cancer cells expressed increased levels of α-smooth muscle actin, a marker of CAF, compared with MSC cultured on a soft matrix. By contrast, MSC cultured on a stiff matrix secreted prosaposin that promoted proliferation and survival of mammary carcinoma cells but inhibited metastasis. Our findings suggest that in addition to chemical stimuli, increased stiffness of the ECM in the tumor microenvironment induces differentiation of MSC to CAF, triggering enhanced proliferation and survival of mammary cancer cells. Cancer Res; 77(22); 6179-89. ©2017 AACR.


Assuntos
Proliferação de Células , Neoplasias Mamárias Experimentais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Saposinas/metabolismo , Animais , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Matriz Extracelular/metabolismo , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Mecanotransdução Celular/efeitos dos fármacos , Mecanotransdução Celular/genética , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
13.
Stem Cell Reports ; 8(2): 387-400, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28162996

RESUMO

Maintaining properties of human bone marrow-derived mesenchymal stem cells (BMSCs) in culture for regenerative applications remains a great challenge. An emerging approach of constructing a culture environment mimicking the bone marrow niche to regulate BMSC activities has been developed. In this study, we have demonstrated a systematic approach to identify soluble factors of interest extracted from human bone marrow and used them in BMSC culture for tissue regeneration. We have found that lipocalin-2 and prolactin are key factors in bone marrow, involved in regulating BMSC activities. Treating the cell with lipocalin-2 and prolactin delays cellular senescence of BMSCs and primes the cell for osteogenesis and chondrogenesis. We have also demonstrated that BMSCs pretreated with lipocalin-2 and prolactin can enhance the repair of calvarial defects in mice. Together, our study provides research evidence of using a viable approach to prime BMSC properties in vitro for improving cell-based tissue regeneration in vivo.


Assuntos
Medula Óssea/metabolismo , Regeneração Óssea , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Regeneração Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem da Célula/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Microambiente Celular , Senescência Celular/genética , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Meios de Cultivo Condicionados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fenótipo , Engenharia Tecidual
14.
J Cell Biochem ; 118(1): 172-181, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27292615

RESUMO

For decades stem cells have proven to be invaluable to the study of tissue development. More recently, mesenchymal stem cells (MSCs) derived from embryonic stem cells (ESCs) (ESC-MSCs) have emerged as a cell source with great potential for the future of biomedical research due to their enhanced proliferative capability compared to adult tissue-derived MSCs and effectiveness of musculoskeletal lineage-specific cell differentiation compared to ESCs. We have previously compared the properties and differentiation potential of ESC-MSCs to bone marrow-derived MSCs. In this study, we evaluated the potential of TGFß1 and BMP7 to induce chondrogenic differentiation of ESC-MSCs compared to that of TGFß1 alone and further investigated the cellular phenotype and intracellular signaling in response to these induction conditions. Our results showed that the expression of cartilage-associated markers in ESC-MSCs induced by the TGFß1 and BMP7 combination was increased compared to induction with TGFß1 alone. The TGFß1 and BMP7 combination upregulated the expression of TGFß receptor and the production of endogenous TGFßs compared to TGFß1 induction. The growth factor combination also increasingly activated both of the TGF and BMP signaling pathways, and inhibition of the signaling pathways led to reduced chondrogenesis of ESC-MSCs. Our findings suggest that by adding BMP7 to TGFß1-supplemented induction medium, ESC-MSC chondrogenesis is upregulated through increased production of endogenous TGFß and activities of TGFß and BMP signaling. J. Cell. Biochem. 118: 172-181, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Condrogênese/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Fator de Crescimento Transformador beta1 , Linhagem Celular , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
15.
Acta Biomater ; 42: 247-257, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27370906

RESUMO

UNLABELLED: Stiffness of biomaterial substrates plays a critical role in regulation of cell behavior. Although the effect of substrate stiffness on cell behavior has been extensively studied, molecular mechanisms of regulation rather than those involving cytoskeletal activities still remain elusive. In this study, we fabricated aligned ultrafine fibers and treated the fiber with different annealing temperatures to produce fibrous substrates with different stiffness. Human mesenchymal stem cells (hMSCs) were then cultured on these fibrous substrates. Our results showed that annealing treatment did not change the diameter of electrospun fibers but increased their polymer crystallinity and mechanical properties. The mRNA expression of RUNX2 was upregulated while the mRNA expression of scleraxis was downregulated in response to an increase in substrate stiffness, suggesting that increased stiffness favorably drives hMSCs into the osteogenic lineage. With subsequent induction of osteogenic differentiation, osteogenesis of hMSCs on stiffer substrates was increased compared to that of the cells on control substrates. Cells on stiffer substrates increasingly activated AKT and YAP and upregulated transcript expression of YAP target genes compared to those on control substrates, and inhibition of AKT led to decreased expression of YAP and RUNX2. Furthermore, macrophage migration inhibitory factor (MIF) was increasingly produced by the cell on stiffer substrates, and knocking down MIF by siRNA resulted in decreased AKT phosphorylation. Taken together, we hereby demonstrate that simply using the annealing approach can manipulate stiffness of an aligned fibrous substrate without altering the material chemistry, and substrate stiffness dictates hMSC differentiation through the MIF-mediated AKT/YAP/RUNX2 pathway. STATEMENT OF SIGNIFICANCE: Stiffness of biomaterial substrates plays a critical role in regulation of cell behavior. Although the effect of substrate stiffness on cell behavior has been extensively studied, molecular mechanisms of regulation rather than those involving cytoskeletal activities still remain elusive. In this manuscript, we report our new findings that simply using the annealing approach can manipulate stiffness of an aligned fibrous substrate without altering the material chemistry, and substrate stiffness dictates human mesenchymal stem cell (hMSC) differentiation through the macrophage migration inhibitory factor-mediated AKT/YAP/RUNX2 pathway. The findings are novel and interesting because we have identified a new mechanism rather than those involving cytoskeleton activity, by which substrate stiffness regulates hMSC behavior.


Assuntos
Células-Tronco Mesenquimais/citologia , Poliésteres/química , Poliésteres/farmacologia , Biomarcadores/metabolismo , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Humanos , Fatores Inibidores da Migração de Macrófagos , Osteogênese/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Stem Cell Rev Rep ; 12(5): 560-572, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27342267

RESUMO

Stem cell fate is largely determined by the microenvironment called niche. The extracellular matrix (ECM), as a key component in the niche, is responsible for maintaining structural stability and regulating cell proliferation, differentiation, migration and other cellular activities. Each tissue has a unique ECM composition for its needs. Here we investigated the effect of a bioengineered human dermal fibroblast-derived ECM (hECM) on the regulation of human mesenchymal stem cell (hMSC) proliferation and multilineage differentiation. Human MSCs were maintained on hECM for two passages followed by the analysis of mRNA expression levels of potency- and lineage-specific markers to determine the capacity of MSC stemness and differentiation, respectively. Mesenchymal stem cells pre-cultured with or without hECM were then induced and analyzed for osteogenesis, adipogenesis and chondrogenesis. Our results showed that compared to MSCs maintained on control culture plates without hECM coating, cells on hECM-coated plates proliferated more rapidly with a higher percentage of cells in S phase of the cell cycle, resulting in an increase in the CD90+/CD105+/CD73+/CD45- subpopulation. In addition, hECM downregulated osteogenesis and adipogenesis of hMSCs but significantly upregulated chondrogenesis with increased production of collagen type 2. In sum, our findings suggest that hECM may be used to culture hMSCs for the application of cartilage tissue engineering.


Assuntos
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Adipogenia/fisiologia , Cartilagem/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Condrogênese/fisiologia , Humanos , Pessoa de Meia-Idade , Osteogênese/fisiologia , Engenharia Tecidual/métodos , Regulação para Cima/fisiologia
17.
Stem Cell Res Ther ; 6: 88, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25998005

RESUMO

INTRODUCTION: Human mesenchymal stem cells (hMSCs) reside in a perivascular niche of the body, suggesting that they interact closely with vascular endothelial cells (ECs) through cell-cell interaction or paracrine signaling to maintain cell functions. Endothelin-1 (ET1) is a paracrine factor mainly secreted by ECs. We thus hypothesize that ECs can regulate cellular activities of hMSCs and direct their stem cell fate. METHODS: We investigated whether co-cultured human aortic endothelial cells (HAECs) were able to regulate expression of potency- and lineage-related markers in bone marrow-derived hMSCs. We further explored the regulatory effects of ET1 on cell proliferation, expression of surface antigens and pluripotency-related markers, and multilineage differentiation in hMSCs. Activation of the AKT signaling pathway in hMSCs was also analyzed to identify its mechanistic role in the ET1-induced regulation. RESULTS: Co-cultured HAECs enhanced expression of mesenchymal lineage-related markers in hMSCs. Treatment of ET receptor antagonist downregulated the increased expression of CBFA1 in hMSCs cultured with HAEC-conditioned medium. hMSCs treated with ET1 showed cell proliferation and expression of surface antigens, CD73, CD90, and CD105, comparable with those without ET1 treatment. ET1-treated hMSCs also expressed upregulated mRNA transcript levels of OCT3/4, NANOG, CBFA1 and SOX9. When induced for lineage-specific differentiation, hMSCs pre-treated with ET1 showed enhanced osteogenesis and chondrogenesis. However, adipogenic differentiation of hMSCs was not affected by ET1 pretreatment. We further showed that the ET1-induced regulation was mediated by activation of AKT signaling. CONCLUSION: Our results demonstrate that ET1 secreted by HAECs can direct bone marrow-derived hMSCs for osteo- and chondro-lineage differentiation through activation of the AKT signaling pathway, suggesting that ET1 plays a crucial role in regulation of hMSC activity. Our findings may help understand how hMSCs interact with ECs in a perivascular niche.


Assuntos
Células Endoteliais/citologia , Endotelina-1/metabolismo , Células-Tronco Mesenquimais/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antígenos CD/metabolismo , Aorta/citologia , Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Condrogênese/efeitos dos fármacos , Técnicas de Cocultura , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Meios de Cultivo Condicionados/farmacologia , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotelina-1/análise , Endotelina-1/farmacologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
Ultrasound Med Biol ; 41(7): 1842-52, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25922132

RESUMO

This study was performed to investigate osteoblastogenesis of human mesenchymal stem cells (hMSCs) cultured in 3-D scaffolds stimulated with low-intensity pulsed ultrasound and to identify the underlying mechanism mediated by soluble receptor activator of nuclear factor kappa B ligand (sRANKL) secreted by hMSCs. The results indicate that the mRNA levels of core-binding factor subunit alpha subunit 1 (CBFA1), osterix (OSX), alkaline phosphatase (ALP), osteocalcin and osteoprotegerin (OPG) and sRANKL production of hMSCs stimulated by ultrasound were significantly increased compared with the levels without ultrasound stimulation. Attenuating the sRANKL activity of ultrasound-treated hMSCs significantly reduced the mRNA expression of CBFA1, OSX, ALP and OPG. Adding sRANKL in hMSC culture significantly increased the mRNA expression of CBFA1, OSX and OPG. Together, the results suggest that osteoblastogenesis of hMSCs enhanced by ultrasound stimulation is mediated by endogenous sRANKL.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Ligante RANK/metabolismo , Ondas Ultrassônicas , Diferenciação Celular/fisiologia , Diferenciação Celular/efeitos da radiação , Células Cultivadas , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos da radiação , Pessoa de Meia-Idade , Osteoblastos/efeitos da radiação , Osteogênese/fisiologia , Osteogênese/efeitos da radiação , Doses de Radiação
19.
Organogenesis ; 11(1): 1-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25915734

RESUMO

Tissue development is orchestrated by the coordinated activities of both chemical and physical regulators. While much attention has been given to the role that chemical regulators play in driving development, researchers have recently begun to elucidate the important role that the mechanical properties of the extracellular environment play. For instance, the stiffness of the extracellular environment has a role in orienting cell division, maintaining tissue boundaries, directing cell migration, and driving differentiation. In addition, extracellular matrix stiffness is important for maintaining normal tissue homeostasis, and when matrix mechanics become imbalanced, disease progression may ensue. In this article, we will review the important role that matrix stiffness plays in dictating cell behavior during development, tissue homeostasis, and disease progression.


Assuntos
Progressão da Doença , Matriz Extracelular/metabolismo , Homeostase , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Citoesqueleto/metabolismo , Elasticidade , Fibrose/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Neoplasias/patologia , Transdução de Sinais , Estresse Mecânico
20.
Stem Cells Dev ; 24(8): 995-1007, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25519748

RESUMO

Human bone marrow stromal/stem cells (hBMSCs) have an inherent tendency to undergo hypertrophy when induced into the chondrogenic lineage using transforming growth factor-beta 1 (TGFß) in vitro, reminiscent of what occurs during endochondral ossification. Surprisingly, Indian Hedgehog (IHH) has received little attention for its role during hBMSC chondrogenesis despite being considered a master regulator of endochondral ossification. In this study, we investigated the role that endogenously produced IHH plays during hBMSC chondrogenesis. We began by analyzing the expression of IHH throughout differentiation using quantitative polymerase chain reaction and found that IHH expression was upregulated dramatically upon chondrogenic induction and peaked from days 9 to 12 of differentiation, which coincided with a concomitant increase in the expression of chondrogenesis- and hypertrophy-related markers, suggesting a potential role for endogenously produced IHH in driving hBMSC chondrogenesis. More importantly, pharmacological inhibition of Hedgehog signaling with cyclopamine or knockdown of IHH almost completely blocked TGFß1-induced chondrogenesis in hBMSCs, demonstrating that endogenously produced IHH is necessary for hBMSC chondrogenesis. Furthermore, overexpression of IHH was sufficient to drive chondrogenic differentiation, even when TGFß signaling was inhibited. Finally, stimulation with TGFß1 induced a significant and sustained upregulation of IHH expression within 3 h that preceded an upregulation in all cartilage-related genes analyzed, and knockdown of IHH blocked the effects of TGFß1 entirely, suggesting that the effects of TGFß1 are being mediated through endogenously produced IHH. Together, our findings demonstrate that endogenously produced IHH is playing a critical role in regulating hBMSC chondrogenesis.


Assuntos
Condrogênese , Proteínas Hedgehog/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Cultivadas , Proteínas Hedgehog/genética , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA