Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 14(11): 730, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37949874

RESUMEN

Ubiquitin-specific protease 4 (USP4) represents a potential oncogene involved in various human cancers. Nevertheless, the biological roles and precise mechanism of USP4 in esophageal squamous cell carcinoma (ESCC) progression are not understood. Here, USP4 expression was found to be markedly upregulated in ESCC tumor tissues and cells. Loss- and gain-of-function assays suggested that USP4 silencing inhibited ESCC cell proliferation, migration, and invasion, while USP4 overexpression promoted these behaviors. Consistently, USP4 silencing repressed tumor growth and metastasis in an ESCC nude mouse model in vivo. As a target molecule of USP4, transforming growth factor-ß-activated kinase 1 (TAK1) also showed high expression in ESCC. Moreover, we observed that USP4 specifically interacted with TAK1 and stabilized TAK1 protein levels via deubiquitination in ESCC cells. Importantly, USP4 promotes ESCC proliferation, migration, and invasion via the MEK/ERK signaling pathway and can be inhibited by U0126. Neutral red (NR), an inhibitor of USP4 can suppress ESCC progression in vitro and in vivo. Overall, this study revealed that USP4/TAK1 plays crucial roles in ESCC progression by modulating proliferation, migration, and invasion, and USP4 might be a potential therapeutic target in ESCC.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Animales , Humanos , Ratones , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Invasividad Neoplásica , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo
2.
Front Genet ; 13: 894990, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832188

RESUMEN

Esophageal squamous cell carcinoma (ESCC), the most prevalent subtype of esophageal cancer, ranks sixth in cancer-related mortality, making it one of the deadliest cancers worldwide. The identification of potential risk factors for ESCC might help in implementing precision therapies. Autophagy-related lncRNAs are a group of non-coding RNAs that perform critical functions in the tumor immune microenvironment and therapeutic response. Therefore, we aimed to establish a risk model composed of autophagy-related lncRNAs that can serve as a potential biomarker for ESCC risk stratification. Using the RNA expression profile from 179 patients in the GSE53622 and GSE53624 datasets, we found 11 lncRNAs (AC004690.2, AC092159.3, AC093627.4, AL078604.2, BDNF-AS, HAND2-AS1, LINC00410, LINC00588, PSMD6-AS2, ZEB1-AS1, and LINC02586) that were co-expressed with autophagy genes and were independent prognostic factors in multivariate Cox regression analysis. The risk model was constructed using these autophagy-related lncRNAs, and the area under the receiver operating characteristic curve (AUC) of the risk model was 0.728. To confirm that the model is reliable, the data of 174 patients from The Cancer Genome Atlas (TCGA) esophageal cancer dataset were analyzed as the testing set. A nomogram for ESCC prognosis was developed using the risk model and clinic-pathological characteristics. Immune function annotation and tumor mutational burden of the two risk groups were analyzed and the high-risk group displayed higher sensitivity in chemotherapy and immunotherapy. Expression of differentially expressed lncRNAs were further validated in human normal esophageal cells and esophageal cancer cells. The constructed lncRNA risk model provides a useful tool for stratifying risk and predicting the prognosis of patients with ESCC, and might provide novel targets for ESCC treatment.

3.
Cancer Sci ; 112(10): 4064-4074, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34251713

RESUMEN

Programmed cell death ligand 1 (PD-L1) is a major immunosuppressive checkpoint protein expressed by tumor cells to subvert anticancer immunity. Recent studies have shown that ionizing radiation (IR) upregulates the expression of PD-L1 in tumor cells. However, whether an IR-induced DNA damage response (DDR) directly regulates PD-L1 expression and the functional significance of its upregulation are not fully understood. Here, we show that IR-induced upregulation of PD-L1 expression proceeds through both transcriptional and post-translational mechanisms. Upregulated PD-L1 was predominantly present on the cell membrane, resulting in T-cell apoptosis in a co-culture system. Using mass spectrometry, we identified PD-L1 interacting proteins and found that BCLAF1 (Bcl2 associated transcription factor 1) is an important regulator of PD-L1 in response to IR. BCLAF1 depletion decreased PD-L1 expression by promoting the ubiquitination of PD-L1. In addition, we show that CMTM6 is upregulated in response to IR and participates in BCLAF1-dependent PD-L1 upregulation. Finally, we demonstrated that the ATM/BCLAF1/PD-L1 axis regulated PD-L1 stabilization in response to IR. Together, our findings reveal a novel regulatory mechanism of PD-L1 expression in the DDR.


Asunto(s)
Antígeno B7-H1/metabolismo , Radiación Ionizante , Proteínas Represoras/fisiología , Proteínas Supresoras de Tumor/fisiología , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Antígeno B7-H1/efectos de la radiación , Línea Celular Tumoral , Membrana Celular/metabolismo , Técnicas de Cocultivo , Daño del ADN , Humanos , Células Jurkat , Proteínas con Dominio MARVEL/metabolismo , Proteínas con Dominio MARVEL/efectos de la radiación , Espectrometría de Masas , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/efectos de la radiación , Proteínas de Neoplasias/metabolismo , Modificación Traduccional de las Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Represoras/deficiencia , Linfocitos T/citología , Linfocitos T/efectos de la radiación , Proteínas Supresoras de Tumor/deficiencia , Ubiquitinación , Regulación hacia Arriba/efectos de la radiación
5.
Mol Oncol ; 14(11): 2701-2712, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32941674

RESUMEN

Expression of programmed cell death ligand (PD-L1) is associated with poor prognosis in breast cancer. Understanding the regulation of PD-L1 expression in breast cancer could provide a new strategy for breast cancer treatment. Here, we demonstrate that moesin (MSN) phosphorylation by Rho-associated protein kinase (ROCK) stabilizes PD-L1 protein levels. Our results indicate that phosphorylated MSN may compete with the E3 ubiquitin ligase SPOP for binding PD-L1. ROCK inhibition via the Y-27632 inhibitor or MSN silencing decreased PD-L1 expression, resulting in T-cell activation both in vitro and in vivo. Administration of Y-27632 into immunocompetent Balb/c mice bearing breast tumors suppressed tumor progression and enhanced CD4+ and CD8+ T-cell infiltration. RNA-seq analysis of Y-27632-treated mouse tumors revealed that ROCK inhibition upregulated several immune response genes. However, the combination of Y-27632 and an anti-PD-1 antibody did not show additive or synergistic effects due to reduced PD-L1 in the presence of Y-27632. Our study unravels a previously unappreciated mechanism of PD-L1 regulation through the ROCK-MSN pathway. Moreover, we found that ROCK inhibitors could be combined with breast cancer immunotherapy.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Microfilamentos/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones Endogámicos BALB C , Fosforilación , Estabilidad Proteica
6.
Elife ; 92020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32538779

RESUMEN

The DNA damage response (DDR) is a highly orchestrated process but how double-strand DNA breaks (DSBs) are initially recognized is unclear. Here, we show that polymerized SIRT6 deacetylase recognizes DSBs and potentiates the DDR in human and mouse cells. First, SIRT1 deacetylates SIRT6 at residue K33, which is important for SIRT6 polymerization and mobilization toward DSBs. Then, K33-deacetylated SIRT6 anchors to γH2AX, allowing its retention on and subsequent remodeling of local chromatin. We show that a K33R mutation that mimics hypoacetylated SIRT6 can rescue defective DNA repair as a result of SIRT1 deficiency in cultured cells. These data highlight the synergistic action between SIRTs in the spatiotemporal regulation of the DDR and DNA repair in humans and mice.


Asunto(s)
Roturas del ADN , Daño del ADN , Reparación del ADN , Sirtuina 1/fisiología , Sirtuinas/fisiología , Acetilación , Animales , Roturas del ADN de Doble Cadena , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Mutagénesis Sitio-Dirigida , Sirtuina 1/metabolismo , Sirtuinas/metabolismo
7.
Nat Metab ; 1(11): 1141-1156, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-32694864

RESUMEN

The central pacemaker in the hypothalamic suprachiasmatic nucleus (SCN) synchronizes peripheral oscillators to coordinate physiological and behavioural activities throughout the body. How circadian phase coherence between the SCN and the periphery is controlled is not well understood. Here, we identify hepatic SIRT7 as an early responsive element to light that ensures circadian phase coherence in the mouse liver. The SCN-driven body temperature (BT) oscillation induces rhythmic expression of HSP70, which promotes SIRT7 ubiquitination and proteasomal degradation. Acute temperature challenge dampens the BT oscillation and causes an advanced liver circadian phase. Further, hepatic SIRT7 deacetylates CRY1, promotes its FBXL3-mediated degradation and regulates the hepatic clock and glucose homeostasis. Loss of Sirt7 in mice leads to an advanced liver circadian phase and rapid entrainment of the hepatic clock upon daytime-restricted feeding. These data identify a BT-HSP70-SIRT7-CRY1 axis that couples the mouse hepatic clock to the central pacemaker and ensures circadian phase coherence and glucose homeostasis.


Asunto(s)
Temperatura Corporal , Ritmo Circadiano , Gluconeogénesis , Luz , Hígado/metabolismo , Sirtuinas/metabolismo , Animales , Homeostasis , Ratones
8.
Oncogene ; 37(49): 6299-6315, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30038266

RESUMEN

Cancer stem cell (CSC)-dictated intratumor heterogeneity accounts for the majority of drug-resistance and distant metastases of breast cancers. Here, we identify a SIRT1-PRRX1-KLF4-ALDH1 circuitry, which couples CSCs, chemo-resistance, metastasis and aging. Pro-longevity protein SIRT1 deacetylates and stabilizes the epithelial-to-mesenchymal-transition (EMT) inducer PRRX1, which inhibits the transcription of core stemness factor KLF4. Loss of SIRT1 destabilizes PRRX1, disinhibits KLF4, and activates the transcription of ALDH1, which induces and functionally marks CSCs, resulting in chemo-resistance and metastatic relapse. Clinically, the level of PRRX1 is positively linked to SIRT1, whereas KLF4 is reversely correlated. Importantly, KLF4 inhibitor Kenpaullone sensitizes breast cancer cells and xenograft tumors to Paclitaxel and improves therapeutic effects. Our findings delineate a SIRT1-centered circuitry that regulates CSC origination, and targeting this pathway might be a promising therapeutic strategy.


Asunto(s)
Neoplasias de la Mama/patología , Células Madre Neoplásicas/patología , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Familia de Aldehído Deshidrogenasa 1 , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Humanos , Isoenzimas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Células Madre Neoplásicas/metabolismo , Retinal-Deshidrogenasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Elife ; 72018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29717979

RESUMEN

DNA damage accumulates with age (Lombard et al., 2005). However, whether and how robust DNA repair machinery promotes longevity is elusive. Here, we demonstrate that ATM-centered DNA damage response (DDR) progressively declines with senescence and age, while low dose of chloroquine (CQ) activates ATM, promotes DNA damage clearance, rescues age-related metabolic shift, and prolongs replicative lifespan. Molecularly, ATM phosphorylates SIRT6 deacetylase and thus prevents MDM2-mediated ubiquitination and proteasomal degradation. Extra copies of Sirt6 extend lifespan in Atm-/- mice, with restored metabolic homeostasis. Moreover, the treatment with CQ remarkably extends lifespan of Caenorhabditis elegans, but not the ATM-1 mutants. In a progeria mouse model with low DNA repair capacity, long-term administration of CQ ameliorates premature aging features and extends lifespan. Thus, our data highlights a pro-longevity role of ATM, for the first time establishing direct causal links between robust DNA repair machinery and longevity, and providing therapeutic strategy for progeria and age-related metabolic diseases.


Asunto(s)
Cloroquina/administración & dosificación , Progeria/tratamiento farmacológico , Sirtuinas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Caenorhabditis elegans , Reparación del ADN , Longevidad , Ratones , Ratones Noqueados , Actividad Motora , Fosforilación , Procesamiento Proteico-Postraduccional , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/metabolismo
10.
Nat Commun ; 8(1): 318, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28827661

RESUMEN

Distant metastasis is the main cause of breast cancer-related death; however, effective therapeutic strategies targeting metastasis are still scarce. This is largely attributable to the spatiotemporal intratumor heterogeneity during metastasis. Here we show that protein deacetylase SIRT7 is significantly downregulated in breast cancer lung metastases in human and mice, and predicts metastasis-free survival. SIRT7 deficiency promotes breast cancer cell metastasis, while temporal expression of Sirt7 inhibits metastasis in polyomavirus middle T antigen breast cancer model. Mechanistically, SIRT7 deacetylates and promotes SMAD4 degradation mediated by ß-TrCP1, and SIRT7 deficiency activates transforming growth factor-ß signaling and enhances epithelial-to-mesenchymal transition. Significantly, resveratrol activates SIRT7 deacetylase activity, inhibits breast cancer lung metastases, and increases survival. Our data highlight SIRT7 as a modulator of transforming growth factor-ß signaling and suppressor of breast cancer metastasis, meanwhile providing an effective anti-metastatic therapeutic strategy.Metastatic disease is the major reason for breast cancer-related deaths; therefore, a better understanding of this process and its players is needed. Here the authors report the role of SIRT7 in inhibiting SMAD4-mediated breast cancer metastasis providing a possible therapeutic avenue.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/metabolismo , Sirtuinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Interferencia de ARN , Transducción de Señal , Sirtuinas/genética , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Trasplante Heterólogo
11.
Cell Tissue Res ; 366(1): 155-62, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27184949

RESUMEN

Small nuclear ribonucleoprotein-associated polypeptide N mutation in mice is associated with short limbs and lower bone mineral density, yet the role of Sm51 in MSC differentiation to osteoblasts is not known. In the present study, we investigate the role of Sm51 in regulating osteoblastic differentiation of bone marrow-derived mesenchymal stem cells (BM-MSCs). Stable overexpression of Sm51 in rat and human BM-MSCs (Sm51-MSCs) significantly enhanced their osteogenic differentiation potential compared to untransfected cells. Under osteogenic induction, Sm51-MSCs had higher alkaline phosphatase (ALP) activity and mineralization ability; the expression of osteogenic genes such as runt-related transcription factor 2 (Runx2), osteocalcin, osteopontin, ALP and type I collagen was significantly upregulated compared to the control BM-MSCs. Furthermore, we show that Sm51 overexpression upregulated Runx2 expression at both the RNA and protein level; Sm51 could bind to Runx2 RNA and regulate its expression. Finally, knocking down Runx2 abolished the promoting effects of Sm51 on osteogenesis in BM-MSCs. These results demonstrate that Sm51 plays an important role in regulating osteogenic differentiation of MSCs through increasing Runx2 expression and that Sm51 may be a potential new therapeutic target for promoting bone formation.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas Nucleares snRNP/metabolismo , Animales , Biomarcadores/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Unión Proteica , ARN/metabolismo , Ratas
12.
Calcif Tissue Int ; 98(3): 263-74, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26603303

RESUMEN

The inhibition of sclerostin by the systemic administration of a monoclonal antibody (Scl-Ab) significantly increased bone mass and strength in fractured bones in animal models and non-fractured bones in ovariectomised (OVX) rats. In this study, the effects of Scl-Ab on healing were examined in a closed fracture model in OVX rats. Sixty Sprague-Dawley rats underwent an ovariectomy or a sham operation at 4 months of age, and a closed fracture of the right femur was performed 3 months later. Subcutaneous injections with Scl-Ab (25 mg/kg) or saline were then administered on day 1 after the fracture and twice a week for 8 weeks (n = 20 per group), at which time the fractured femurs were harvested for micro-computed tomography analysis, four-point bending mechanical testing and histomorphometric analysis to examine bone mass, bone strength and dynamic bone formation at the fracture site. The angiogenesis at the fracture site was also examined. Bone marrow stem cells were also isolated from the fractured bone to perform a colony-forming unit (CFU) assay and an alkaline phosphatase-positive (ALP(+)) CFU assay. OVX rats treated with Scl-Ab for 8 weeks had significantly increased bone mineral density and relative bone volume compared with OVX rats treated with saline. Similarly, maximum loading, energy to maximum load and stiffness in Scl-Ab-treated OVX rats were significantly higher than those in saline controls. The mineral apposition rate (MAR), mineralising surface (MS/BS) and bone formation rate (BFR/BS) were also significantly increased in Scl-Ab-treated group compared with the saline-treated group in OVX rats. Furthermore, the Scl-Ab-treated group had more CFUs and ALP(+) CFUs than the saline-treated group in OVX rats. No significant difference in angiogenesis at the fracture site was found between the groups. Our study demonstrated that Scl-Ab helped to increase bone mass, bone strength and bone formation at the fracture site in a closed femoral fracture model in OVX rats. Bone marrow stem cells in OVX rats injected with Scl-Ab also had increased CFUs and ALP(+) CFUs.


Asunto(s)
Anticuerpos/química , Proteínas Morfogenéticas Óseas/química , Fracturas del Fémur/inmunología , Fosfatasa Alcalina/metabolismo , Animales , Peso Corporal , Células de la Médula Ósea/citología , Femenino , Fracturas del Fémur/diagnóstico por imagen , Curación de Fractura , Marcadores Genéticos , Inyecciones Subcutáneas , Neovascularización Patológica , Osteogénesis , Ovariectomía , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Estrés Mecánico , Microtomografía por Rayos X
13.
Sci Rep ; 5: 11056, 2015 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-26053250

RESUMEN

Mesenchymal stem cells (MSCs) are highly plastic cells that are able to transdifferentiate or dedifferentiate under appropriate conditions. In the present study, we reported here that after in vitro induction of osteogenic differentiation, MSCs could be reverted to a primitive stem cell population (dedifferentiated osteogenic MSCs, De-Os-MSCs) with improved cell survival, colony formation, osteogenic potential, migratory capacity and increased expression of Nanog, Oct4 and Sox2. Most importantly, our results showed great superiority of the De-Os-MSCs over untreated MSCs in ectopic bone formation in vivo. Furthermore, Nanog-knockdown in MSCs could reverse these enhanced properties in De-Os-MSCs in vitro, indicating a central role of Nanog in the transcriptional network. In addition, epigenetic regulations including DNA methylation and histone modifications may play important roles in regulating the de-osteogenic differentiation process. And we found decreased methylation and promoter accrual of activating histone marks, such as H3K4me3 and H4ac on both Nanog and Oct4 gene promoters. Taken together, our study demonstrated that epigenetic memory in De-Os-MSCs gained by priming with osteogenic induction medium favored their differentiation along osteoblastic lineage with improved cell survival and migratory abilities, which may have application potential in enhancing their regenerative capacity in mammals.


Asunto(s)
Desdiferenciación Celular/fisiología , Epigénesis Genética/genética , Células Madre Mesenquimatosas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción/genética , Adipogénesis/fisiología , Animales , Regeneración Ósea/genética , Regeneración Ósea/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/genética , Células Cultivadas , Condrogénesis/fisiología , Metilación de ADN/genética , Ratones , Ratones Desnudos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Osteogénesis/fisiología , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción/biosíntesis
14.
FASEB J ; 29(4): 1143-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25466891

RESUMEN

Mesenchymal stem cells (MSCs) are a promising cell resource for tissue engineering. Sry-related high-mobility group box 11 (Sox11) plays critical roles in neural development and organogenesis. In the present study, we investigated the role of Sox11 in regulating trilineage differentiation (osteogenesis, adipogenesis, and chondrogenesis) and migration of MSCs, and explored the effect of systemically administrated Sox11-modified MSCs on bone fracture healing using the rat model of open femur fracture. Our results demonstrated that Sox11 overexpression increased the trilineage differentiation and migration of MSCs, as well as cell viability under oxidative stress. The effect of Sox11 on osteogenesis was confirmed by ectopic bone formation assay conducted in nude mice. In addition, we found that Sox11 could activate the bone morphogenetic protein (BMP)/Smad signaling pathway in MSCs. By dual-luciferase reporter assay, we also demonstrated that Sox11 could transcriptionally activate runt-related transcription factor 2 (Runx2) and CXC chemokine receptor-4 (CXCR4) expression. The activation of the BMP/Smad signaling pathway and Runx2, CXCR4 expression may have a synergic effect, which largely contributed to the effect of Sox11 on MSC fate determination and migration. Finally, using an open femur fracture model in rats, we found that a larger number of MSCs stably expressing Sox11 migrated to the fracture site and improved bone fracture healing. Taken together, our study shows that Sox11 is an important regulator of MSC differentiation and migration, and Sox11-modified MSCs may have clinical implication for accelerating bone fracture healing, which can reduce the delayed unions or nonunions.


Asunto(s)
Curación de Fractura/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Factores de Transcripción SOXC/fisiología , Adipogénesis/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Movimiento Celular , Supervivencia Celular , Condrogénesis/fisiología , Fracturas del Fémur/patología , Fracturas del Fémur/terapia , Curación de Fractura/genética , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Desnudos , Osteogénesis/fisiología , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Transcripción SOXC/genética , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo
15.
Stem Cells Dev ; 23(1): 66-75, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23962074

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) have the potential to migrate to the site of injury and regulate the repair process. Aquaporin 1 (Aqp1) is a water channel molecule and a regulator of endothelial cell migration. To study the role of Apq1 in MSC migration, we manipulated the expression of the Aqp1 gene in MSCs and explored its effects on MSC migration both in vitro and in vivo. Overexpression of Aqp1 promoted MSC migration, while depletion of Aqp1 impaired MSC migration in vitro. When the green fluorescent protein (GFP) labeled Aqp1 overexpressing MSCs were systemically injected into rats with a femoral fracture, there were significantly more GFP-MSCs found at the fracture gap in the Aqp1-GFP-MSC-treated group compared to the GFP-MSC group. To elucidate the underlying mechanism, we screened several migration-related regulators. The results showed that ß-catenin and focal adhesion kinase (FAK) were upregulated in the Aqp1-MSCs and downregulated in the Aqp1-depleted MSCs, while C-X-C chemokine receptor type 4 had no change. Furthermore, ß-catenin and FAK were co-immunoprecipitated with Aqp1, and depletion of FAK abolished the Aqp1 effects on MSC migration. This study demonstrates that Aqp1 enhances MSC migration ability mainly through the FAK pathway and partially through the ß-catenin pathway. Our finding suggests a novel function of Aqp1 in governing MSC migration, and this may aid MSC therapeutic applications.


Asunto(s)
Acuaporina 1/metabolismo , Regeneración Ósea/fisiología , Quinasa 1 de Adhesión Focal/metabolismo , Células Madre Mesenquimatosas/metabolismo , beta Catenina/metabolismo , Animales , Acuaporina 1/biosíntesis , Acuaporina 1/genética , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Regeneración Ósea/genética , Movimiento Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos , Regulación hacia Abajo , Quinasa 1 de Adhesión Focal/biosíntesis , Fracturas Óseas/genética , Fracturas Óseas/terapia , Proteínas Fluorescentes Verdes , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/biosíntesis , Tibia/citología , Tibia/lesiones , beta Catenina/biosíntesis
16.
Mol Biol Rep ; 40(3): 2533-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23187741

RESUMEN

N-cadherin, a calcium-dependent cellular adhesive protein, plays important roles during embryonic development and bone formation. The potential of mesenchymal stem cells (MSCs) in osteoblast differentiation and homing to the sites of injury make it a promising cell resource for tissue engineering. However, the role of N-cadherin in MSCs osteoblast differentiation and migration remains still obscure. In the present study, our results showed that prolonged N-cadherin overexpression inhibited osteogenic differentiation of MSCs through negatively regulating ß-catenin and ERK1/2 signaling pathways. The mRNA expression levels of osteogenesis-related genes (Osteopontin, Osteocalcin, runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and bone morphogenetic protein 2) were significantly inhibited by N-cadherin, as well as the ALP activity and calcium deposit as stained by Alizarin Red S. While, silencing N-cadherin using shRNA reversed this effect. Furthermore, ectopic bone formation conducted in nude mice verified that N-cadherin significantly inhibited ectopic bone formation of MSCs in vivo. In addition, we also found that the N-cadherin overexpression could promote the migration potential of MSCs. These findings reveal that N-cadherin inhibits osteogenesis but promotes migration of MSCs. The underlying mechanism of N-cadherin inhibiting osteogenesis may through suppressing ß-catenin and ERK1/2 signaling pathways.


Asunto(s)
Cadherinas/genética , Movimiento Celular/genética , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Animales , Diferenciación Celular , Expresión Génica , Silenciador del Gen , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas/citología , Ratones , Interferencia de ARN , beta Catenina/metabolismo
17.
Cell Biol Int ; 36(4): 415-8, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22233450

RESUMEN

UCB (umbilical cord blood) as a resource of MSCs (mesenchymal stem cells) is widely accepted, but the quantity and characteristics of UCB-MSCs from different gestational ages have not been well studied. We have quantified the number of MSCs in UCB at different gestational ages using a multi-colour flowcytometer and compared the cell proliferation rates of these UCB-MSCs. Defining MSCs as CD44+/CD105+/CD34-/CD45 population, their numbers declined in the UCB at the gestational age. Proliferation rates were significantly higher in UCB before term than at full term. Non-full term UCB samples may be a better source of MSCs.


Asunto(s)
Sangre Fetal/citología , Células Madre Mesenquimatosas/citología , Embarazo/sangre , Adulto , Antígenos CD/análisis , Recuento de Células , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Cesárea , Femenino , Sangre Fetal/fisiología , Feto , Citometría de Flujo , Edad Gestacional , Humanos , Células Madre Mesenquimatosas/fisiología
18.
Int J Biochem Cell Biol ; 44(4): 612-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22230368

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent adult stem cells that can differentiate into osteoblasts, chondrocytes and adipocytes, providing a potential source for musculoskeletal tissue engineering. Retinoid signaling plays very important roles in skeletal development. CRBP1 (cellular retinol binding protein 1), a key component of retinoid signaling pathway, is known to take part in vitamin A metabolism and intracellular transporting of retinoids. However, the role of CRBP1 in MSCs remains still obscure. In this study, we investigated the cellular effects of CRBP1 on osteogenic and adipogenic differentiation of bone marrow derived MSCs in vitro and in vivo. Our results showed that CRBP1 overexpression promoted osteogenic differentiation of bone marrow derived MSCs, while inhibited their adipogenic differentiation. We also demonstrated that the possible underlying mechanism for CRBP1 promoting osteogenic differentiation of MSCs was by inhibiting RXRα-induced ß-catenin degradation, maintaining ß-catenin and pERK1/2 at higher levels. These findings reveal a potential role of CRBP1 in the regulation of ß-catenin turnover which can greatly affect the process of osteogenesis and adipogenesis of MSCs.


Asunto(s)
Adipogénesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Receptor alfa X Retinoide/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , beta Catenina/metabolismo , Animales , Células de la Médula Ósea/citología , Silenciador del Gen , Células HEK293 , Humanos , Lentivirus/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Estabilidad Proteica , Proteolisis , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Proteínas Celulares de Unión al Retinol/deficiencia , Proteínas Celulares de Unión al Retinol/genética , Transducción de Señal
19.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 31(6): 702-6, 2009 Dec.
Artículo en Chino | MEDLINE | ID: mdl-20078938

RESUMEN

OBJECTIVE: To analyze the change of the neuronal restricted silencing factor (NRSF) gene as well as the NRSF regulation genes in beta-mercaptoethanol induction of the marrow mesenchymal stem cells (MSCs) to neurons, and to discuss the function of NRSF in neural induction of the MSCs and the mechanism of the differentiation from MSCs to neurons. METHOD: We used beta-mercaptoethanol, serum-free DMEM, and dimethyl sulfoxide to induce rat MSCs to differentiate to neurons, and then analyzed the changes of the expressions of NRSF gene and NRSF-regulated genes through real-time PCR. RESULTS: The rat MSCs were successfully induced to differentiate into neuron-like cells. The induced neuron marker, neuron-specific enolase, was positive. Real-time PCR showed that the expression of NRSF gene remarkably declined. The expressions of neurotrophic tyrosine kinase receptor, type 3, synaptosomal-associated protein 25, L1 cell adhesion molecular,neuronal pentraxin receptor in the NRSF-regulated genes also increased at varied extents. CONCLUSIONS: The differentiation from MSCs to neurons is relevant with the decline of NRSF expression and the increase of the expressions of NRSF-regulated genes. The NRSF may be the key gene during the differentiation from MSCs to neurons.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , Neuronas/citología , Proteínas Represoras/fisiología , Animales , Células de la Médula Ósea/metabolismo , Células Cultivadas , Células Madre Mesenquimatosas/metabolismo , Neuronas/metabolismo , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Wistar , Proteínas Represoras/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA