Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Environ Toxicol ; 38(8): 2022-2030, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37163415

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Proteína X Asociada a bcl-2 , Neoplasias de la Mama/metabolismo , Apoptosis , Proliferación Celular , Autofagia , Línea Celular Tumoral
3.
Sci Adv ; 8(46): eadc9222, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36383652

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Humanos , Células Madre Mesenquimatosas/metabolismo , Epigénesis Genética , Histonas/metabolismo , Diferenciación Celular/genética , Osteoblastos , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo
5.
Sci Rep ; 12(1): 9039, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35641537

RESUMEN

Miniature pigs are an ideal animal model for translational research to evaluate stem cell therapies and regenerative applications. While the derivation of induced pluripotent stem cells (iPSCs) from miniature pigs has been demonstrated, there is still a lack of a reliable method to generate and maintain miniature pig iPSCs. In this study, we derived iPSCs from fibroblasts of Wisconsin miniature swine (WMS), Yucatan miniature swine (YMS), and Göttingen minipigs (GM) using our culture medium. By comparing cells of the different pig breeds, we found that YMS fibroblasts were more efficiently reprogrammed into iPSCs, forming colonies with well-defined borders, than WMS and GM fibroblasts. We also demonstrated that YMS iPSC lines with a normal pig karyotype gave rise to cells of the three germ layers in vitro and in vivo. Mesenchymal stromal cells expressing phenotypic characteristics were derived from established iPSC lines as an example of potential applications. In addition, we found that the expression level of the switch/sucrose nonfermentable component BAF60A regulated by STAT3 signaling determined the efficiency of pig iPSC generation. The findings of this study provide insight into the underlying mechanism controlling the reprogramming efficiency of miniature pig cells to develop a viable strategy to enhance the generation of iPSCs for biomedical research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Reprogramación Celular/genética , Epigénesis Genética , Fibroblastos/metabolismo , Porcinos , Porcinos Enanos
6.
Cartilage ; 13(2_suppl): 530S-543S, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34467773

RESUMEN

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.


Asunto(s)
Cartílago Articular , Células Madre Mesenquimatosas , Animales , Condrogénesis/genética , Proteínas Hedgehog , Proteínas de Homeodominio , Humanos , Líquido Sinovial
7.
Sci Adv ; 7(21)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34138734

RESUMEN

Generating phenotypic chondrocytes from pluripotent stem cells is of great interest in the field of cartilage regeneration. In this study, we differentiated human induced pluripotent stem cells into the mesodermal and ectomesodermal lineages to prepare isogenic mesodermal cell-derived chondrocytes (MC-Chs) and neural crest cell-derived chondrocytes (NCC-Chs), respectively, for comparative evaluation. Our results showed that both MC-Chs and NCC-Chs expressed hyaline cartilage-associated markers and were capable of generating hyaline cartilage-like tissue ectopically and at joint defects. Moreover, NCC-Chs revealed closer morphological and transcriptional similarities to native articular chondrocytes than MC-Chs. NCC-Ch implants induced by our growth factor mixture demonstrated increased matrix production and stiffness compared to MC-Ch implants. Our findings address how chondrocytes derived from pluripotent stem cells through mesodermal and ectomesodermal differentiation are different in activities and functions, providing the crucial information that helps make appropriate cell choices for effective regeneration of articular cartilage.


Asunto(s)
Cartílago Articular , Células Madre Pluripotentes Inducidas , Diferenciación Celular , Condrocitos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Regeneración
9.
J Biomech ; 116: 110215, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33482593

RESUMEN

Non-invasive estimation of cartilage material properties is useful for understanding cartilage health and creating subject-specific computational models. Bi-component T2 mapping measured using Multi-Component Driven Equilibrium Single Shot Observation of T1 and T2 (mcDESPOT) is sensitive for detecting cartilage degeneration within the human knee joint, but has not been correlated with cartilage composition and mechanical properties. Therefore, the purpose of this study was to investigate the relationship between bi-component T2 parameters measured using mcDESPOT at 3.0 T and cartilage composition and mechanical properties. Ex-vivo patellar cartilage specimens harvested from five human cadaveric knees were imaged using mcDESPOT at 3.0 T. Cartilage samples were removed from the patellae, mechanically tested to determine linear modulus and dissipated energy, and chemically tested to determine proteoglycan and collagen content. Parameter maps of single-component T2 relaxation time (T2), the T2 relaxation times of the fast relaxing macromolecular bound water component (T2F) and slow relaxing bulk water component (T2S), and the fraction of the fast relaxing macromolecular bound water component (FF) were compared to mechanical and chemical measures using linear regression. FF was significantly (p < 0.05) correlated with energy dissipation and linear modulus. T2 was significantly (p ≤ 0.05) correlated with elastic modulus at 1 Hz and energy dissipated at all frequencies. There were no other significant (p = 0.13-0.97) correlations between mcDESPOT parameters and mechanical properties. FF was significantly (p = 0.04) correlated with proteoglycan content. There were no other significant (p = 0.19-0.92) correlations between mcDESPOT parameters and proteoglycan or collagen content. This study suggests that FF measured using mcDESPOT at 3.0 T could be used to non-invasively estimate cartilage proteoglycan content, elastic modulus, and energy dissipation.


Asunto(s)
Cartílago Articular , Humanos , Rodilla , Articulación de la Rodilla , Imagen por Resonancia Magnética , Rótula
10.
Stem Cells ; 39(1): 62-77, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33252174

RESUMEN

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.


Asunto(s)
Senescencia Celular , Factor de Transcripción GATA6/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal , Adulto , Animales , Femenino , Factor de Transcripción GATA6/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad
11.
Int J Biol Macromol ; 170: 248-260, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359806

RESUMEN

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.


Asunto(s)
Sulfatos de Condroitina/farmacología , Colágeno/farmacología , Ingeniería de Tejidos/métodos , Adulto , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sulfatos de Condroitina/metabolismo , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Matriz Extracelular , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Tendones/citología , Tendones/fisiología , Andamios del Tejido/química
12.
Biochem Biophys Res Commun ; 529(2): 180-185, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32703408

RESUMEN

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.


Asunto(s)
Condrogénesis , Endotelina-1/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/metabolismo
13.
Am J Sports Med ; 47(11): 2729-2736, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31339739

RESUMEN

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.


Asunto(s)
Trasplante de Médula Ósea , Huesos/cirugía , Trasplante de Células Madre Mesenquimatosas , Tendones/trasplante , Cicatrización de Heridas/fisiología , Animales , Huesos/fisiología , Masculino , Modelos Animales , Distribución Aleatoria , Ratas Sprague-Dawley , Tendones/fisiología , Trasplante Autólogo
14.
J Orthop Translat ; 17: 73-81, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31194067

RESUMEN

The discovery of induced pluripotent stem cells (iPSCs) has revolutionized biomedicine. Although the potential of iPSCs for tissue regeneration, disease modeling and drug screening has been largely recognized, findings of iPSC research to date are mostly focused on neurology, cardiology and haematology. For orthopaedics, growing interest in the unique cell type has prompted more researchers to get involved in iPSC research. In this article, we introduce the brief history of cellular reprogramming and different reprogramming methods that have been developed, discuss the biology of iPSCs and review previously reported findings of iPSC studies in orthopaedics. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: Stem cell therapies hold great promise for treating orthopaedic diseases, manifested in recent study findings and results of clinical trials. iPSCs are a unique stem cell type derived from a patient's own cells while still possessing the embryonic stem cell-featured pluripotency for generation of all tissues in the body. The distinctive properties make iPSCs much desirable to fulfill the promise of regenerative medicine for clinical orthopaedics.

15.
Sci Adv ; 5(3): eaau7375, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30891496

RESUMEN

Brain pericytes play important roles in the formation and maintenance of the neurovascular unit (NVU), and their dysfunction has been implicated in central nervous system disorders. While human pluripotent stem cells (hPSCs) have been used to model other NVU cell types, including brain microvascular endothelial cells (BMECs), astrocytes, and neurons, hPSC-derived brain pericyte-like cells have not been integrated into these models. In this study, we generated neural crest stem cells (NCSCs), the embryonic precursor to forebrain pericytes, from hPSCs and subsequently differentiated NCSCs to brain pericyte-like cells. These cells closely resembled primary human brain pericytes and self-assembled with endothelial cells. The brain pericyte-like cells induced blood-brain barrier properties in BMECs, including barrier enhancement and reduced transcytosis. Last, brain pericyte-like cells were incorporated with iPSC-derived BMECs, astrocytes, and neurons to form an isogenic human model that should prove useful for the study of the NVU.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Cresta Neural/metabolismo , Pericitos/metabolismo , Transcitosis/genética , Animales , Antígenos/genética , Antígenos/metabolismo , Astrocitos/citología , Astrocitos/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Células Endoteliales/citología , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/citología , Neuronas/citología , Neuronas/metabolismo , Pericitos/citología , Cultivo Primario de Células , Prosencéfalo/citología , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
16.
Stem Cells Transl Med ; 8(6): 522-534, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30784225

RESUMEN

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.


Asunto(s)
Enfermedades Óseas Metabólicas/etiología , Proteína Morfogenética Ósea 6/metabolismo , Diabetes Mellitus Tipo 1/patología , Animales , Densidad Ósea , Proteína Morfogenética Ósea 6/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Huesos/diagnóstico por imagen , Huesos/patología , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/complicaciones , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Microtomografía por Rayos X
17.
FASEB J ; 33(1): 996-1007, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30096039

RESUMEN

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.


Asunto(s)
Tejido Adiposo/citología , Células de la Médula Ósea/citología , Endotelina-1/metabolismo , Células Madre/citología , Adulto , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Osteogénesis , Receptores de Endotelina/metabolismo
18.
J Phys Condens Matter ; 31(3): 035501, 2019 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-30523842

RESUMEN

Topological semimetals have a variety of phases, whose Fermi surfaces can be nodal points, nodal lines and nodal loops. Here we construct four classes of 3D minimal models via vertically stacking a 2D nonsymmorphic lattice with and without breaking crystalline symmetries. As a result, four distinct topological phases can be generated in our minimal model, such as Dirac nodal line semimetals, Weyl nodal line semimetals, unconventional Weyl semimetals with topological charge [Formula: see text], and weak topological insulators. Unexpectedly, Weyl nodal loops are generated without mirror symmetry protection, where nontrivial 'drumhead' surface states emerge within the loops.

19.
J Biomed Mater Res A ; 106(6): 1595-1603, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29427535

RESUMEN

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.


Asunto(s)
Materiales Biocompatibles/química , Diferenciación Celular , Células Endoteliales/citología , Células Madre Mesenquimatosas/citología , Andamios del Tejido/química , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Dureza , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Nanofibras/química , Nanofibras/ultraestructura , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Cancer Res ; 77(22): 6179-6189, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28972074

RESUMEN

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.


Asunto(s)
Proliferación Celular , Neoplasias Mamarias Experimentales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Saposinas/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Matriz Extracelular/metabolismo , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...