Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(43): e2304689120, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37856544

RESUMEN

The importance of classical CD8+ T cells in tumor eradication is well acknowledged. However, the anti-tumor activity of MHC (major histocompatibility complex) Ib-restricted CD8+ T (Ib-CD8+ T) cells remains obscure. Here, we show that CX3CR1-expressing Ib-CD8+ T cells (Ib-restricted CD8+ T cells) highly express cytotoxic factors, austerely resist exhaustion, and effectively eliminate various tumors. These Ib-CD8+ T cells can be primed by MHC Ia (MHC class Ia molecules) expressed on various cell types for optimal activation in a Tbet-dependent manner. Importantly, MHC Ia does not allogeneically activate Ib-CD8+ T cells, rather, sensitizes these cells for T cell receptor activation. Such effects were observed when MHC Ia+ cells were administered to tumor-bearing Kb-/-Db-/-mice. A similar population of tumoricidal CX3CR1+CD8+ T cells was identified in wild-type mice and melanoma patients. Adoptive transfer of Ib-CD8+ T cells to wild-type mice inhibited tumor progression without damaging normal tissues. Taken together, we demonstrate that MHC class Ia can prime Ib-CD8+ T cells for robust tumoricidal activities.


Asunto(s)
Linfocitos T CD8-positivos , Antígenos de Histocompatibilidad Clase I , Humanos , Ratones , Animales , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos H-2 , Antígenos de Histocompatibilidad/metabolismo , Ratones Endogámicos C57BL
2.
Hepatology ; 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38466833

RESUMEN

BACKGROUND AND AIMS: RAD51 recombinase (RAD51) is a highly conserved DNA repair protein and is indispensable for embryonic viability. As a result, the role of RAD51 in liver development and function is unknown. Our aim was to characterize the function of RAD51 in postnatal liver development. APPROACH AND RESULTS: RAD51 is highly expressed during liver development and during regeneration following hepatectomy and hepatic injury, and is also elevated in chronic liver diseases. We generated a hepatocyte-specific Rad51 deletion mouse model using Alb -Cre ( Rad51 -conditional knockout (CKO)) and Adeno-associated virus 8-thyroxine-binding globulin-cyclization recombination enzyme to evaluate the function of RAD51 in liver development and regeneration. The phenotype in Rad51 -CKO mice is dependent on CRE dosage, with Rad51fl/fl ; Alb -Cre +/+ manifesting a more severe phenotype than the Rad51fl/fl ; Alb -Cre +/- mice. RAD51 deletion in postnatal hepatocytes results in aborted mitosis and early onset of pathological polyploidization that is associated with oxidative stress and cellular senescence. Remarkable liver fibrosis occurs spontaneously as early as in 3-month-old Rad51fl/fl ; Alb -Cre +/+ mice. While liver regeneration is compromised in Rad51 -CKO mice, they are more tolerant of carbon tetrachloride-induced hepatic injury and resistant to diethylnitrosamine/carbon tetrachloride-induced HCC. A chronic inflammatory microenvironment created by the senescent hepatocytes appears to activate ductular reaction the transdifferentiation of cholangiocytes to hepatocytes. The newly derived RAD51 functional immature hepatocytes proliferate vigorously, acquire increased malignancy, and eventually give rise to HCC. CONCLUSIONS: Our results demonstrate a novel function of RAD51 in liver development, homeostasis, and tumorigenesis. The Rad51 -CKO mice represent a unique genetic model for premature liver senescence, fibrosis, and hepatocellular carcinogenesis.

3.
Mol Ther ; 32(4): 1144-1157, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38310354

RESUMEN

The potent immunomodulatory function of mesenchymal stem/stromal cells (MSCs) elicited by proinflammatory cytokines IFN-γ and TNF-α (IT) is critical to resolve inflammation and promote tissue repair. However, little is known about how the immunomodulatory capability of MSCs is related to their differentiation competency in the inflammatory microenvironment. In this study, we demonstrate that the adipocyte differentiation and immunomodulatory function of human adipose tissue-derived MSCs (MSC(AD)s) are mutually exclusive. Mitochondrial reactive oxygen species (mtROS), which promote adipocyte differentiation, were decreased in MSC(AD)s due to IT-induced upregulation of superoxide dismutase 2 (SOD2). Furthermore, knockdown of SOD2 led to enhanced adipogenic differentiation but reduced immunosuppression capability of MSC(AD)s. Interestingly, the adipogenic differentiation was associated with increased mitochondrial biogenesis and upregulation of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PPARGC1A/PGC-1α) expression. IT inhibited PGC-1α expression and decreased mitochondrial mass but promoted glycolysis in an SOD2-dependent manner. MSC(AD)s lacking SOD2 were compromised in their therapeutic efficacy in DSS-induced colitis in mice. Taken together, these findings indicate that the adipogenic differentiation and immunomodulation of MSC(AD)s may compete for resources in fulfilling the respective biosynthetic needs. Blocking of adipogenic differentiation by mitochondrial antioxidant may represent a novel strategy to enhance the immunosuppressive activity of MSCs in the inflammatory microenvironment.


Asunto(s)
Células Madre Mesenquimatosas , Superóxido Dismutasa , Ratones , Humanos , Animales , Diferenciación Celular , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Adipocitos , Células Madre Mesenquimatosas/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(5): 2462-2472, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31953260

RESUMEN

Preadipocytes can give rise to either white adipocytes or beige adipocytes. Owing to their distinct abilities in nutrient storage and energy expenditure, strategies that specifically promote "beiging" of adipocytes hold great promise for counterbalancing obesity and metabolic diseases. Yet, factors dictating the differentiation fate of adipocyte progenitors remain to be elucidated. We found that stearoyl-coenzyme A desaturase 1 (Scd1)-deficient mice, which resist metabolic stress, possess augmentation in beige adipocytes under basal conditions. Deletion of Scd1 in mature adipocytes expressing Fabp4 or Ucp1 did not affect thermogenesis in mice. Rather, Scd1 deficiency shifted the differentiation fate of preadipocytes from white adipogenesis to beige adipogenesis. Such effects are dependent on succinate accumulation in adipocyte progenitors, which fuels mitochondrial complex II activity. Suppression of mitochondrial complex II by Atpenin A5 or oxaloacetic acid reverted the differentiation potential of Scd1-deficient preadipocytes to white adipocytes. Furthermore, supplementation of succinate was found to increase beige adipocyte differentiation both in vitro and in vivo. Our data reveal an unappreciated role of Scd1 in determining the cell fate of adipocyte progenitors through succinate-dependent regulation of mitochondrial complex II.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Grasas/metabolismo , Obesidad/enzimología , Estearoil-CoA Desaturasa/genética , Ácido Succínico/metabolismo , Adipocitos Beige/citología , Adipocitos Beige/metabolismo , Adipogénesis , Animales , Metabolismo Energético , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Estearoil-CoA Desaturasa/metabolismo , Termogénesis
5.
Cell Immunol ; 379: 104576, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35797932

RESUMEN

Neutrophils are the most predominant cell population in the innate immune system. The role of neutrophils in the initiation, development and metastasis of tumor has been actively studied in recent years. In cancer, neutrophils exert both pro- and anti-cancer effects, and their phenotype and function are affected by the tumor microenvironment (TME). This review aims to summarize the role of neutrophils in tumorigenesis with emphasis on their interaction with mesenchymal stromal cells (MSCs).


Asunto(s)
Células Madre Mesenquimatosas , Neoplasias , Carcinogénesis , Humanos , Neutrófilos/patología , Microambiente Tumoral
6.
J Pathol ; 254(2): 185-198, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33638154

RESUMEN

Tamoxifen (TAM) resistance is a significant clinical challenge in endocrine therapies for estrogen receptor (ER)-positive breast cancer patients. Cullin 4B (CUL4B), which acts as a scaffold protein in CUL4B-RING ubiquitin ligase complexes (CRL4B), is frequently overexpressed in cancer and represses tumor suppressors through diverse epigenetic mechanisms. However, the role and the underlying mechanisms of CUL4B in regulating drug resistance remain unknown. Here, we showed that CUL4B promotes TAM resistance in breast cancer cells through a miR-32-5p/ER-α36 axis. We found that upregulation of CUL4B correlated with decreased TAM sensitivity of breast cancer cells, and knockdown of CUL4B or expression of a dominant-negative CUL4B mutant restored the response to TAM in TAM-resistant MCF7-TAMR and T47D-TAMR cells. Mechanistically, we demonstrated that CUL4B renders breast cancer cells TAM-resistant by upregulating ER-α36 expression, which was mediated by downregulation of miR-32-5p. We further showed that CRL4B epigenetically represses the transcription of miR-32-5p by catalyzing monoubiquitination at H2AK119 and coordinating with PRC2 and HDAC complexes to promote trimethylation at H3K27 at the promoter of miR-32-5p. Pharmacologic or genetic inhibition of CRL4B/PRC2/HDAC complexes significantly increased TAM sensitivity in breast cancer cells in vitro and in vivo. Taken together, our findings thus establish a critical role for the CUL4B-miR-32-5p-ER-α36 axis in the regulation of TAM resistance and have important therapeutic implications for combined application of TAM and the inhibitors of CRL4B/PRC2/HDAC complex in breast cancer treatment. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/patología , Proteínas Cullin/metabolismo , Receptor alfa de Estrógeno/genética , MicroARNs/genética , Tamoxifeno/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proteínas Cullin/genética , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Ratones , Mutación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer ; 18(1): 177, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31805946

RESUMEN

Cancer immunotherapies that engage immune cells to fight against tumors are proving to be powerful weapons in combating cancer and are becoming increasingly utilized in the clinics. However, for the majority of patients with solid tumors, little or no progress has been seen, presumably due to lack of adequate approaches that can reprogram the local immunosuppressive tumor milieu and thus reinvigorate antitumor immunity. Tumor-associated macrophages (TAMs), which abundantly infiltrate most solid tumors, could contribute to tumor progression by stimulating proliferation, angiogenesis, metastasis, and by providing a barrier against antitumor immunity. Initial TAMs-targeting strategies have shown efficacy across therapeutic modalities and tumor types in both preclinical and clinical studies. TAMs-targeted therapeutic approaches can be roughly divided into those that deplete TAMs and those that modulate TAMs activities. We here reviewed the mechanisms by which macrophages become immunosuppressive and compromise antitumor immunity. TAMs-focused therapeutic strategies are also summarized.


Asunto(s)
Inmunoterapia , Macrófagos/inmunología , Neoplasias/etiología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Humanos , Inmunoterapia/métodos , Macrófagos/metabolismo , Macrófagos/patología , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/patología
8.
Cytometry A ; 95(3): 302-308, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30508271

RESUMEN

Small cell lung cancer (SCLC) needs to be classified from poorly differentiated lung adenocarcinoma (PDLAC) for appropriate treatment of lung cancer patients. Currently, the classification is achieved by experienced clinicians, radiologists and pathologists based on subjective and qualitative analysis of imaging, cytological and immunohistochemical (IHC) features. Label-free classification of lung cancer cell lines is developed here by using two-dimensional (2D) light scattering static cytometric technique. Measurements of scattered light at forward scattering (FSC) and side scattering (SSC) by using conventional cytometry show that SCLC cells are overlapped with PDLAC cells. However, our 2D light scattering static cytometer reveals remarkable differences between the 2D light scattering patterns of SCLC cell lines (H209 and H69) and PDLAC cell line (SK-LU-1). By adopting support vector machine (SVM) classifier with leave-one-out cross-validation (LOO-CV), SCLC and PDLAC cells are automatically classified with an accuracy of 99.87%. Our label-free 2D light scattering static cytometer may serve as a new, accurate, and easy-to-use method for the automatic classification of SCLC and PDLAC cells. © 2018 International Society for Advancement of Cytometry.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Citometría de Flujo/métodos , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Línea Celular Tumoral , Humanos , Láseres de Semiconductores , Aprendizaje Automático , Máquina de Vectores de Soporte
9.
Hum Mol Genet ; 24(3): 853-64, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25274780

RESUMEN

CUL4A and CUL4B are closely related cullin family members and can each assemble a Cullin-RING E3 ligase complex (CRL) and participate in a variety of biological processes. While the CRLs formed by the two cullin members may have common targets, the two appeared to have very different consequences when mutated or disrupted in mammals. We here investigated the roles of cul4a and cul4b during zebrafish embryogenesis by using the morpholino knockdown approach. We found that cul4a is essential for cardiac development as well as for pectoral fin development. Whereas cul4a morphants appeared to be unperturbed in chamber specification, they failed to undergo heart looping. The failures in heart looping and pectoral fin formation in cul4a morphants were accompanied by greatly reduced proliferation of cardiac cells and pectoral fin-forming cells. We demonstrated that tbx5a, a transcription factor essential for heart and limb development, is transcriptionally upregulated by cul4a and mediates the function of cul4a in cardiac and pectoral fin development. In contrast to the critical importance of cul4a, cul4b appeared to be dispensable for zebrafish development and was incapable of compensating for the loss of cul4a. This work provides the first demonstration of an essential role of cul4a, but not cul4b, in cardiac development and in the regulation of tbx5a in zebrafish. These findings justify exploring the functional role of CUL4A in human cardiac development.


Asunto(s)
Proteínas Cullin/metabolismo , Miembro Anterior/crecimiento & desarrollo , Corazón/crecimiento & desarrollo , Proteínas de Dominio T Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Cardiopatías Congénitas/metabolismo , Miocitos Cardíacos/metabolismo , Pez Cebra/metabolismo
10.
Hum Mol Genet ; 24(16): 4686-97, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26025376

RESUMEN

Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Glial fibrillary acidic protein (GFAP) is regarded as a hallmark of mature astrocytes, though some GFPA-positive cells may act as neural stem cells. Missense heterozygous mutations in GFAP cause Alexander disease that manifests leukodystrophy and intellectual disability. Here, we show that CUL4B, a scaffold protein that assembles E3 ubiquitin ligase, represses the expression of GFAP in neural progenitor cells (NPCs) during brain development. Lack of Cul4b in NPCs in cultures led to increased generation of astrocytes, marked by GFAP and S100ß. The GFAP+ cells were also found to be more abundant in the brains of nervous system-specific Cul4b knockout mice in vivo. Moreover, we demonstrated that the increased generation of GFAP+ cells from Cul4b-null NPCs was mediated by an upregulation of prostaglandin D2 synthase PTGDS. We showed that the increased GFAP expression can be attenuated by pharmacological inhibition of the PTGDS enzymatic activity or by shRNA-mediated knockdown of Ptgds. Importantly, exogenously added PTGDS could promote the generation of GFAP+ cells from wild-type NPCs. We further observed that Ptgds is targeted and repressed by the CUL4B/PRC2 complex. Together, our results demonstrate CUL4B as a negative regulator of GFAP expression during neural development.


Asunto(s)
Encéfalo/metabolismo , Proteínas Cullin/metabolismo , Oxidorreductasas Intramoleculares/biosíntesis , Lipocalinas/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Células-Madre Neurales/metabolismo , Animales , Encéfalo/citología , Proteínas Cullin/genética , Técnicas de Silenciamiento del Gen , Proteína Ácida Fibrilar de la Glía , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología
12.
J Cell Sci ; 127(Pt 21): 4679-91, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25189618

RESUMEN

CUL4B, a scaffold protein that assembles the CRL4B ubiquitin ligase complex, participates in the regulation of a broad spectrum of biological processes. Here, we demonstrate a crucial role of CUL4B in driving cell cycle progression. We show that loss of CUL4B results in a significant reduction in cell proliferation and causes G1 cell cycle arrest, accompanied by the upregulation of the cyclin-dependent kinase (CDK) inhibitors (CKIs) p21 and p57 (encoded by CDKN1A and CDKN1C, respectively). Strikingly, CUL4B was found to negatively regulate the function of p21 through transcriptional repression, but not through proteolysis. Furthermore, we demonstrate that CRL4B and SIN3A-HDAC complexes interact with each other and co-occupy the CDKN1A and CDKN1C promoters. Lack of CUL4B led to a decreased retention of SIN3A-HDAC components and increased levels of acetylated H3 and H4. Interestingly, the ubiquitylation function of CRL4B is not required for the stable retention of SIN3A-HDAC on the promoters of target genes. Thus, in addition to directly contributing to epigenetic silencing by catalyzing H2AK119 monoubiquitylation, CRL4B also facilitates the deacetylation function of SIN3A-HDAC. Our findings reveal a coordinated action between CRL4B and SIN3A-HDAC complexes in transcriptional repression.


Asunto(s)
Proteínas Cullin/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Represoras/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Proteínas Cullin/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Células HeLa , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Inmunoprecipitación , Masculino , Ratones , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Complejo Correpresor Histona Desacetilasa y Sin3
13.
Opt Express ; 24(19): 21700-7, 2016 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-27661908

RESUMEN

Two-dimensional (2D) light scattering patterns of single microspheres, normal granulocytes and leukemic cells are obtained by label-free static cytometry. Statistical results of experimental 2D light scattering patterns obtained from standard microspheres with a mean diameter of 4.19 µm agree well with theoretical simulations. High accuracy rates (greater than 92%) for label-free differentiation of normal granulocytes and leukemic cells, both the acute and chronic leukemic cells, are achieved by analyzing the 2D light scattering patterns. Our label-free static cytometry is promising for leukemia screening in clinics.

14.
Tumour Biol ; 37(7): 8721-9, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26738868

RESUMEN

Ovarian cancer is the most lethal gynecological malignancy. Patients usually have poor prognosis because of late diagnosis, relapse, and chemoresistance. It is pressing to seek novel agent for the treatment of ovarian cancer. Neferine is a bisbenzylisoquinoline alkaloid isolated from the embryos of lotus (Nelumbo nucifera). In this study, we investigated the antitumor effect of neferine on ovarian cancer cells. We found that neferine exhibited growth-inhibitory effect on human ovarian cancer cells, whereas showing less cytotoxic to non-malignant fallopian tube epithelial cells. Furthermore, we demonstrated that neferine induced autophagy and inactivated the mTOR pathway. Finally, we found that both p38 MAPK and JNK signaling pathways were activated by neferine treatment and contributed to the induction of autophagy in ovarian cancer cells. In conclusion, our findings showed that neferine induced autophagy of human ovarian cancer cells via p38 MAPK/JNK activation. Neferine may be explored as a promising antitumoral agent in ovarian cancer.


Asunto(s)
Autofagia/efectos de los fármacos , Bencilisoquinolinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Humanos , Nelumbo/química , Neoplasias Ováricas/metabolismo , Extractos Vegetales/farmacología , Serina-Treonina Quinasas TOR/metabolismo
15.
J Pathol ; 235(5): 784-95, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25430888

RESUMEN

Activation of Wnt/ß-catenin signalling is frequently observed in many types of cancer including hepatocellular carcinoma (HCC). We recently reported that cullin 4B (CUL4B), a scaffold protein that assembles CRL4B ubiquitin ligase complexes, is overexpressed in many types of solid tumours and contributes to epigenetic silencing of tumour suppressors. In this study, we characterized the function of CUL4B in HCC and investigated whether CUL4B is involved in the regulation of Wnt/ß-catenin signalling. CUL4B and ß-catenin were frequently up-regulated and positively correlated in HCC tissues. CUL4B activated Wnt/ß-catenin signalling by protecting ß-catenin from GSK3-mediated degradation, achieved through CUL4B-mediated epigenetic silencing of Wnt pathway antagonists. Knockdown of CUL4B resulted in the up-regulation of Wnt signal antagonists such as DKK1 and PPP2R2B. Simultaneous knockdown of PPP2R2B partially reversed the down-regulation of ß-catenin signalling caused by CUL4B depletion. Furthermore, CRL4B promoted the recruitment and/or retention of PRC2 at the promoters of Wnt antagonists and CUL4B knockdown decreased the retention of PRC2 components as well as H3K27me3. Knockdown of CUL4B reduced the proliferation, colony formation, and invasiveness of HCC cells in vitro and inhibited tumour growth in vivo, and these effects were attenuated by introduction of exogenous ß-catenin or simultaneous knockdown of PPP2R2B. Conversely, ectopic expression of CUL4B enhanced the proliferation and invasiveness of HCC cells. We conclude that CUL4B can up-regulate Wnt/ß-catenin signalling in human HCC through transcriptionally repressing Wnt antagonists and thus contributes to the malignancy of HCC.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Proteínas Cullin/metabolismo , Neoplasias Hepáticas/enzimología , Vía de Señalización Wnt , beta Catenina/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Movimiento Celular , Proliferación Celular , Proteínas Cullin/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteolisis , Factores de Tiempo , Transcripción Genética , Transfección , Carga Tumoral , beta Catenina/genética
16.
Hum Mutat ; 36(2): 240-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25402622

RESUMEN

Using whole-exome sequencing, we surveyed all the potential pathogenic variants in an SPG42 family and found five SNPs and four indels that are shared by two patients and lie in the mapped region. Two variants, SLC33A1 p.Ser113Arg and VEPH1 p.Gln433His, cosegregated with the disease. However, VEPH1 p.Gln433His was predicted to be tolerated, thus leaving SLC33A1 p.Ser113Arg as the most plausible causal variant in this family. We found that the phosphorylated SMAD1/5/8 (P-SMAD1/5/8) and BMP receptor type 1A (BMPR1A) were substantially upregulated in fibroblasts derived from an SPG42 individual. Slc33a1 knockdown zebrafish, which exhibited defects in morphology and axon outgrowth, also showed a significant elevation in the level of P-smad1/5/8. While the phenotypes in slc33a1 knockdown zebrafish could be rescued by human wild-type SLC33A1 mRNA, this rescuing effect was diminished by coinjected mutant mRNA encoding p.Ser113Arg, indicating that p.Ser113Arg variant acts in a dominant-negative manner. Importantly, pharmacological blockade of BMPR1 activity by dorsomorphin could efficiently rescue the phenotypic defects in slc33a1 knockdown zebrafish. These results indicate that SLC33A1 can negatively regulate BMP signaling.


Asunto(s)
Proteínas de Transporte de Membrana/genética , Paraplejía Espástica Hereditaria/genética , Sustitución de Aminoácidos , Animales , Proteínas Morfogenéticas Óseas/fisiología , Estudios de Asociación Genética , Humanos , Mutación INDEL , Polimorfismo de Nucleótido Simple , Transducción de Señal , Pez Cebra
17.
Hum Mutat ; 36(1): 98-105, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25331638

RESUMEN

Autosomal-recessive nonsyndromic hearing loss (ARNSHL) features a high degree of genetic heterogeneity. Many genes responsible for ARNSHL have been identified or mapped. We previously mapped an ARNSHL locus at 17q12, herein designated DFNB99, in a consanguineous Chinese family. In this study, whole-exome sequencing revealed a homozygous missense mutation (c.1259G>A, p.Arg420Gln) in the gene-encoding transmembrane protein 132E (TMEM132E) as the causative variant. Immunofluorescence staining of the Organ of Corti showed Tmem132e highly expressed in murine inner hair cells. Furthermore, knockdown of the tmem132e ortholog in zebrafish affected the mechanotransduction of hair cells. Finally, wild-type human TMEM132E mRNA, but not the mRNA carrying the c.1259G>A mutation rescued the Tmem132e knockdown phenotype. We conclude that the variant in TMEM132E is the most likely cause of DFNB99.


Asunto(s)
Células Ciliadas Auditivas Internas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Análisis de Secuencia de ADN/métodos , Animales , China , Cromosomas Humanos Par 15/genética , Sordera/genética , Exoma , Técnicas de Silenciamiento del Gen , Genes Recesivos , Humanos , Masculino , Ratones , Mutación Missense , Linaje , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
18.
Stem Cells ; 32(2): 327-37, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24123709

RESUMEN

An imbalance between normal adipogenesis and osteogenesis by mesenchymal stem cells (MSCs) has been shown to be related to various human metabolic diseases, such as obesity and osteoporosis; however, the underlying mechanisms remain elusive. We found that the interaction between osteopontin (OPN), an arginine-glycine-aspartate-containing glycoprotein, and integrin αv/ß1 plays a critical role in the lineage determination of MSCs. Although OPN is a well-established marker during osteogenesis, its role in MSC differentiation is still unknown. Our study reveals that blockade of OPN function promoted robust adipogenic differentiation, while inhibiting osteogenic differentiation. Re-expression of OPN restored a normal balance between adipogenesis and osteogenesis in OPN(-/-) MSCs. Retarded bone formation by OPN(-/-) MSCs was also verified by in vivo implantation with hydroxyapatite-tricalcium phosphate, a bone-forming matrix. The role of extracellular OPN in MSC differentiation was further demonstrated by supplementation and neutralization of OPN. Blocking well-known OPN receptors integrin αv/ß1 but not CD44 also affected MSC differentiation. Further studies revealed that OPN inhibits the C/EBPs signaling pathway through integrin αv/ß1. Consistent with these in vitro results, OPN(-/-) mice had a higher fat to total body weight ratio than did wild-type mice. Therefore, our study demonstrates a novel role for OPN-integrin αv/ß1 in regulating MSC differentiation.


Asunto(s)
Adipogénesis/genética , Osteogénesis/genética , Osteopontina/metabolismo , Receptores de Vitronectina/metabolismo , Adipocitos/citología , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Humanos , Células Madre Mesenquimatosas , Ratones , Osteoblastos/metabolismo , Osteopontina/genética , Mapas de Interacción de Proteínas/genética , Receptores de Vitronectina/genética
19.
Biochim Biophys Acta ; 1832(5): 595-605, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23357576

RESUMEN

Cullin 4B (CUL4B) is a scaffold protein involved in the assembly of cullin-RING ubiquitin ligase (E3) complexes. Contemporary reports have identified multiple mutations of CUL4B gene as being causally associated with X-linked intellectual disability (XLID). Identifying the specific protein substrates will help to better understand the physiological functions of CUL4B. The current study identified Jun activation domain-binding protein (Jab1/CSN5) in the COP9 signalosome (CSN) complex as a novel proteolytic target for the CUL4B ubiquitin ligase complex. The impaired degradation of Jab1 was observed in cells after RNAi-mediated CUL4B depletion. Integrity of DDB1-CUL4B-ROC1 was further demonstrated to be indispensable for the degradation of Jab1. In addition, the degradation of Jab1 is independent of CUL4A, a cullin family member closely related to CUL4B. In vitro and in vivo ubiquitination assays revealed that CUL4B promoted the polyubiquitination of Jab1. Interestingly, CUL4B-silenced cells were shown to exhibit abnormal upregulation of bone morphogenetic protein (BMP) signaling. Furthermore, in vivo studies of embryonic fibroblasts in Cul4b-deficient mice demonstrated Jab1 accumulation and increased activation of the BMP signaling pathway. Together, the current findings demonstrate the CUL4B E3 ubiquitin ligase plays a key role in targeting Jab1 for degradation, potentially revealing a previously undocumented mechanism for regulation of the BMP signaling pathway involved with the CUL4B-based E3 complex. This observation may provide novel insights into the molecular mechanisms underlying CUL4B-associated XLID pathogenesis.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Cullin/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptido Hidrolasas/metabolismo , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/genética , Complejo del Señalosoma COP9 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteínas Cullin/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Genes Ligados a X/genética , Células HEK293 , Humanos , Immunoblotting , Discapacidad Intelectual/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Microscopía Fluorescente , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Péptido Hidrolasas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Interferencia de ARN , Ubiquitinación
20.
J Immunol ; 189(3): 1182-92, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22753940

RESUMEN

Suppression of immune response by mesenchymal stem/stromal cells (MSCs) is well documented. However, their regulatory effects on immune cells, especially regulatory dendritic cells, are not fully understood. We have identified a novel Sca-1(+)Lin(-)CD117(-) MSC population isolated from mouse embryonic fibroblasts (MEF) that suppressed lymphocyte proliferation in vitro. Moreover, the Sca-1(+)Lin(-)CD117(-) MEF-MSCs induced hematopoietic stem/progenitor cells to differentiate into novel regulatory dendritic cells (DCs) (Sca-1(+)Lin(-)CD117(-) MEF-MSC-induced DCs) when cocultured in the absence of exogenous cytokines. Small interfering RNA silencing showed that Sca-1(+)Lin(-)CD117(-) MEF-MSCs induced the generation of Sca-1(+)Lin(-)CD117(-) MEF-MSC-induced DCs via IL-10-activated SOCS3, whose expression was regulated by the JAK-STAT pathway. We observed a high degree of H3K4me3 modification mediated by MLL1 and a relatively low degree of H3K27me3 modification regulated by SUZ12 on the promoter of SOCS3 during SOCS3 activation. Importantly, infusion of Sca-1(+)CD117(-)Lin(-) MEF-MSCs suppressed the inflammatory response by increasing DCs with a regulatory phenotype. Thus, our results shed new light on the role of MSCs in modulating regulatory DC production and support the clinical application of MSCs to reduce the inflammatory response in numerous disease states.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Interleucina-10/fisiología , Células Madre Mesenquimatosas/inmunología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Células del Estroma/inmunología , Células del Estroma/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA