Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Mol Gastroenterol Hepatol ; 12(5): 1809-1830, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34237462

RESUMEN

BACKGROUND & AIMS: The gastrointestinal epithelium plays a crucial role in maintaining homeostasis with the gut microbiome. Mucins are essential for intestinal barrier function and serve as a scaffold for antimicrobial factors. Mucin 2 (MUC2) is the major intestinal gel-forming mucin produced predominantly by goblet cells. Goblet cells express anterior gradient 2 (AGR2), a protein disulfide isomerase that is crucial for proper processing of gel-forming mucins. Here, we investigated 2 siblings who presented with severe infantile-onset inflammatory bowel disease. METHODS: We performed whole-genome sequencing to identify candidate variants. We quantified goblet cell numbers using H&E histology and investigated the expression of gel-forming mucins, stress markers, and goblet cell markers using immunohistochemistry. AGR2-MUC2 binding was evaluated using co-immunoprecipitation. Endoplasmic reticulum (ER) stress regulatory function of mutant AGR2 was examined by expression studies in Human Embryonic Kidney 293T (HEK293T) using tunicamycin to induce ER stress. RESULTS: Both affected siblings were homozygous for a missense variant in AGR2. Patient biopsy specimens showed reduced goblet cells; depletion of MUC2, MUC5AC, and MUC6; up-regulation of AGR2; and increased ER stress. The mutant AGR2 showed reduced capacity to bind MUC2 and alleviate tunicamycin-induced ER stress. CONCLUSIONS: Phenotype-genotype segregation, functional experiments, and the striking similarity of the human phenotype to AGR2-/- mouse models suggest that the AGR2 missense variant is pathogenic. The Mendelian deficiency of AGR2, termed "Enteropathy caused by AGR2 deficiency, Goblet cell Loss, and ER Stress" (EAGLES), results in a mucus barrier defect, the inability to mitigate ER stress, and causes infantile-onset inflammatory bowel disease.


Asunto(s)
Susceptibilidad a Enfermedades , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Mucoproteínas/deficiencia , Moco/metabolismo , Proteínas Oncogénicas/deficiencia , Secuencia de Aminoácidos , Animales , Biomarcadores , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Predisposición Genética a la Enfermedad , Células Caliciformes/metabolismo , Células Caliciformes/patología , Humanos , Enfermedades Inflamatorias del Intestino/diagnóstico , Mucosa Intestinal/patología , Masculino , Ratones Noqueados , Mucinas/genética , Mucinas/metabolismo , Mucoproteínas/química , Mucoproteínas/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Fenotipo , Análisis de Secuencia de ADN , Hermanos , Relación Estructura-Actividad , Secuenciación Completa del Genoma
2.
Small GTPases ; 11(4): 293-300, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-29173011

RESUMEN

Macrophages are innate immune cells that constantly patrol an organism to fulfill protective and homeostatic roles. Previous studies have shown that Rho GTPase activity is required for macrophage mobility, yet the roles of upstream regulatory proteins controlling Rho GTPase function in these cells are not well defined. Previously we have shown that the RhoA GEF Net1 is required for human breast cancer cell motility and extracellular matrix invasion. To assess the role of Net1 in macrophage motility, we isolated bone marrow macrophage (BMM) precursors from wild type and Net1 knockout mice. Loss of Net1 did not affect the ability of BMM precursors to differentiate into mature macrophages in vitro, as measured by CD68 and F4/80 staining. However, Net1 deletion significantly reduced RhoA activation, F-actin accumulation, adhesion, and motility in these cells. Nevertheless, similar to RhoA/RhoB double knockout macrophages, Net1 deletion did not impair macrophage recruitment to the peritoneum in a mouse model of sterile inflammation. These data demonstrate that Net1 is an important regulator of RhoA signaling and motility in mouse macrophages in vitro, but that its function may be dispensable for macrophage recruitment to inflammatory sites in vivo.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Macrófagos/metabolismo , Proteínas Oncogénicas/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Animales , Diferenciación Celular , Células Cultivadas , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo
3.
Elife ; 82019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31769754

RESUMEN

Comprehensive knowledge of the host factors required for picornavirus infection would facilitate antiviral development. Here we demonstrate roles for three human genes, TNK2, WASL, and NCK1, in infection by multiple picornaviruses. CRISPR deletion of TNK2, WASL, or NCK1 reduced encephalomyocarditis virus (EMCV), coxsackievirus B3 (CVB3), poliovirus and enterovirus D68 infection, and chemical inhibitors of TNK2 and WASL decreased EMCV infection. Reduced EMCV lethality was observed in mice lacking TNK2. TNK2, WASL, and NCK1 were important in early stages of the viral lifecycle, and genetic epistasis analysis demonstrated that the three genes function in a common pathway. Mechanistically, reduced internalization of EMCV was observed in TNK2 deficient cells demonstrating that TNK2 functions in EMCV entry. Domain analysis of WASL demonstrated that its actin nucleation activity was necessary to facilitate viral infection. Together, these data support a model wherein TNK2, WASL, and NCK1 comprise a pathway important for multiple picornaviruses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interacciones Huésped-Patógeno , Proteínas Oncogénicas/metabolismo , Picornaviridae/crecimiento & desarrollo , Proteínas Tirosina Quinasas/metabolismo , Internalización del Virus , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Infecciones por Cardiovirus/patología , Línea Celular , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/deficiencia , Análisis de Supervivencia , Proteína Neuronal del Síndrome de Wiskott-Aldrich/antagonistas & inhibidores , Proteína Neuronal del Síndrome de Wiskott-Aldrich/deficiencia
4.
J Pathol ; 245(4): 445-455, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29756208

RESUMEN

RNF43 mutations are frequently detected in colorectal cancer cells and lead to a loss of function of the ubiquitin E3 ligase. Here, we investigated the clinical significance of RNF43 mutations in a large Japanese cohort and the role of RNF43 at various stages of colorectal cancer development and progression. Mutation analysis of the RNF43 gene locus with pyrosequencing technology detected RNF43 hotspot mutations in one (0.88%) of 113 colorectal polyp cases and in 30 (6.45%) of 465 colorectal cancer cases. Moreover, patients with colorectal cancer harbouring mutated RNF43 experienced a higher recurrence rate than those harbouring non-mutated RNF43. In addition, the growth of RNF43 wild-type colorectal cancer cell lines was significantly increased by RNF43 silencing. We generated Rnf43 knockout mice in a C57BL/6 N background by using the CRISPR-Cas9 system. Although intestinal organoids from Rnf43 knockout mice did not show continuous growth in the absence of R-spondin, an azoxymethane/dextran sodium sulphate mouse model demonstrated that tumours were markedly larger in Rnf43 knockout mice than in wild-type mice. These findings provide evidence that Wnt signalling activation by RNF43 mutations during the tumourigenic stage enhances tumour growth and promotes a high recurrence rate in colorectal cancer patients. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/genética , Mutación con Pérdida de Función , Proteínas Oncogénicas/genética , Ubiquitina-Proteína Ligasas/genética , Anciano , Animales , Biomarcadores de Tumor/deficiencia , Movimiento Celular , Proliferación Celular , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Proteínas de Unión al ADN/deficiencia , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Células HCT116 , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Japón , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Proteínas Oncogénicas/deficiencia , Fenotipo , Factores de Riesgo , Factores de Tiempo , Carga Tumoral , Ubiquitina-Proteína Ligasas/deficiencia , Vía de Señalización Wnt
5.
Proc Natl Acad Sci U S A ; 115(5): 1015-1020, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339491

RESUMEN

E-type cyclins (cyclins E1 and E2) are components of the core cell cycle machinery and are overexpressed in many human tumor types. E cyclins are thought to drive tumor cell proliferation by activating the cyclin-dependent kinase 2 (CDK2). The cyclin E1 gene represents the site of recurrent integration of the hepatitis B virus in the pathogenesis of hepatocellular carcinoma, and this event is associated with strong up-regulation of cyclin E1 expression. Regardless of the underlying mechanism of tumorigenesis, the majority of liver cancers overexpress E-type cyclins. Here we used conditional cyclin E knockout mice and a liver cancer model to test the requirement for the function of E cyclins in liver tumorigenesis. We show that a ubiquitous, global shutdown of E cyclins did not visibly affect postnatal development or physiology of adult mice. However, an acute ablation of E cyclins halted liver cancer progression. We demonstrated that also human liver cancer cells critically depend on E cyclins for proliferation. In contrast, we found that the function of the cyclin E catalytic partner, CDK2, is dispensable in liver cancer cells. We observed that E cyclins drive proliferation of tumor cells in a CDK2- and kinase-independent mechanism. Our study suggests that compounds which degrade or inhibit cyclin E might represent a highly selective therapeutic strategy for patients with liver cancer, as these compounds would selectively cripple proliferation of tumor cells, while sparing normal tissues.


Asunto(s)
Ciclina E/metabolismo , Neoplasias Hepáticas/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular , Ciclina E/deficiencia , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Ciclinas/deficiencia , Ciclinas/genética , Ciclinas/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo
6.
Exp Hematol ; 59: 40-50.e3, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29288703

RESUMEN

Self-renewing hematopoietic stem cells and multipotent progenitor cells are responsible for maintaining hematopoiesis throughout an individual's lifetime. For overall health and survival, it is critical that the genome stability of these cells is maintained and that the cell population is not exhausted. Previous reports have indicated that the DEK protein, a chromatin structural protein that functions in numerous nuclear processes, is required for DNA damage repair in vitro and long-term engraftment of hematopoietic stem cells in vivo. Therefore, we investigated the role of DEK in normal hematopoiesis and response to DNA damaging agents in vivo. Here, we report that hematopoiesis is largely unperturbed in DEK knockout mice compared with wild-type (WT) controls. However, DEK knockout mice have fewer radioprotective units, but increased capacity to survive repeated sublethal doses of radiation exposure compared with WT mice. Furthermore, this increased survival correlated with a sustained quiescent state in which DEK knockout restricted hematopoietic progenitor cells (HPC-1) were nearly three times more likely to be quiescent following irradiation compared with WT cells and were significantly more radioresistant during the early phases of myeloid reconstitution. Together, our studies indicate that DEK functions in the normal hematopoietic stress response to recurrent radiation exposure.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/deficiencia , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Oncogénicas/deficiencia , Proteínas de Unión a Poli-ADP-Ribosa/deficiencia , Tolerancia a Radiación/fisiología , Animales , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados
7.
Mol Biol Cell ; 28(24): 3500-3516, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28954862

RESUMEN

Although it is known that noncatalytic region of tyrosine kinase (Nck) regulates cell adhesion and migration by bridging tyrosine phosphorylation with cytoskeletal remodeling, the role of Nck in tumorigenesis and metastasis has remained undetermined. Here we report that Nck is required for the growth and vascularization of primary tumors and lung metastases in a breast cancer xenograft model as well as extravasation following injection of carcinoma cells into the tail vein. We provide evidence that Nck directs the polarization of cell-matrix interactions for efficient migration in three-dimensional microenvironments. We show that Nck advances breast carcinoma cell invasion by regulating actin dynamics at invadopodia and enhancing focalized extracellular matrix proteolysis by directing the delivery and accumulation of MMP14 at the cell surface. We find that Nck-dependent cytoskeletal changes are mechanistically linked to enhanced RhoA but restricted spatiotemporal activation of Cdc42. Using a combination of protein silencing and forced expression of wild-type/constitutively active variants, we provide evidence that Nck is an upstream regulator of RhoA-dependent, MMP14-mediated breast carcinoma cell invasion. By identifying Nck as an important driver of breast carcinoma progression and metastasis, these results lay the groundwork for future studies assessing the therapeutic potential of targeting Nck in aggressive cancers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Neoplasias de la Mama/metabolismo , Proteínas Oncogénicas/deficiencia , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Transformación Celular Neoplásica , Femenino , Xenoinjertos , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Fosforilación , Podosomas/metabolismo , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo
8.
Cell Signal ; 36: 79-90, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28455143

RESUMEN

PTP1B, a prototype of the non-receptor subfamily of the protein tyrosine phosphatase superfamily, plays a key role in regulating intracellular signaling from various receptor and non-receptor protein tyrosine kinases. Previously, we reported that silencing Nck1 in human hepatocellular carcinoma HepG2 cells enhances basal and growth factor-induced activation of the PI3K-Akt pathway through attenuating PTP1B expression. However, the underlying mechanism by which Nck1 depletion represses PTP1B expression remains unclear. In this study, we found that silencing Nck1 attenuates PTP1B expression in HepG2 cells through down-regulation of IRE1α. Indeed, we show that silencing Nck1 in HepG2 cells leads to decreased IRE1α expression and signaling. Accordingly, IRE1α depletion using siRNA in HepG2 cells enhances PI3K-dependent basal and growth factor-induced Akt activation, reproducing the effects of silencing Nck1 on activation of this pathway. In addition, depletion of IRE1α also leads to reduced PTP1B expression, which was rescued by ectopic expression of IRE1α in Nck1-depleted cells. Mechanistically, we found that silencing either Nck1 or IRE1α in HepG2 cells decreases PTP1B mRNA levels and stability. However, despite miR-122 levels, a miRNA targeting PTP1B 3' UTR and inducing PTP1B mRNA degradation in HepG2 cells, are increased in both Nck1- and IRE1α-depleted HepG2 cells, a miR-122 antagomir did not rescue PTP1B expression in these cells. Overall, this study highlights an important role for Nck1 in fine-tuning IRE1α expression and signaling that regulate PTP1B expression and subsequent activation of the PI3K-Akt pathway in HepG2 cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endorribonucleasas/metabolismo , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Silenciador del Gen/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Ratones , MicroARNs/metabolismo , Proteínas Oncogénicas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina/farmacología
9.
Stem Cells ; 35(7): 1773-1785, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28436588

RESUMEN

ERG, an ETS family transcription factor frequently overexpressed in human leukemia, has been implicated as a key regulator of hematopoietic stem cells. However, how ERG controls normal hematopoiesis, particularly at the stem and progenitor cell level, and how it contributes to leukemogenesis remain incompletely understood. Using homologous recombination, we generated an Erg knockdown allele (Ergkd ) in which Erg expression can be conditionally restored by Cre recombinase. Ergkd/kd animals die at E10.5-E11.5 due to defects in endothelial and hematopoietic cells, but can be completely rescued by Tie2-Cre-mediated restoration of Erg in these cells. In Ergkd/+ mice, ∼40% reduction in Erg dosage perturbs both fetal liver and bone marrow hematopoiesis by reducing the numbers of Lin- Sca-1+ c-Kit+ (LSK) hematopoietic stem and progenitor cells (HSPCs) and megakaryocytic progenitors. By genetic mosaic analysis, we find that Erg-restored HSPCs outcompete Ergkd/+ HSPCs for contribution to adult hematopoiesis in vivo. This defect is in part due to increased apoptosis of HSPCs with reduced Erg dosage, a phenotype that becomes more drastic during 5-FU-induced stress hematopoiesis. Expression analysis reveals that reduced Erg expression leads to changes in expression of a subset of ERG target genes involved in regulating survival of HSPCs, including increased expression of a pro-apoptotic regulator Bcl2l11 (Bim) and reduced expression of Jun. Collectively, our data demonstrate that ERG controls survival of HSPCs, a property that may be used by leukemic cells. Stem Cells 2017;35:1773-1785.


Asunto(s)
Apoptosis/genética , Dosificación de Gen , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Oncogénicas/genética , Regulador Transcripcional ERG/genética , Animales , Antimetabolitos/farmacología , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Femenino , Fluorouracilo/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Prueba de Complementación Genética , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Integrasas/genética , Integrasas/metabolismo , Masculino , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Regulador Transcripcional ERG/deficiencia
10.
Circulation ; 133(4): 409-21, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26659946

RESUMEN

BACKGROUND: Sprouting angiogenesis is a key process driving blood vessel growth in ischemic tissues and an important drug target in a number of diseases, including wet macular degeneration and wound healing. Endothelial cells forming the sprout must develop front-rear polarity to allow sprout extension. The adaptor proteins Nck1 and 2 are known regulators of cytoskeletal dynamics and polarity, but their function in angiogenesis is poorly understood. Here, we show that the Nck adaptors are required for endothelial cell front-rear polarity and migration downstream of the angiogenic growth factors VEGF-A and Slit2. METHODS AND RESULTS: Mice carrying inducible, endothelial-specific Nck1/2 deletions fail to develop front-rear polarized vessel sprouts and exhibit severe angiogenesis defects in the postnatal retina and during embryonic development. Inactivation of NCK1 and 2 inhibits polarity by preventing Cdc42 and Pak2 activation by VEGF-A and Slit2. Mechanistically, NCK binding to ROBO1 is required for both Slit2- and VEGF-induced front-rear polarity. Selective inhibition of polarized endothelial cell migration by targeting Nck1/2 prevents hypersprouting induced by Notch or Bmp signaling inhibition, and pathological ocular neovascularization and wound healing, as well. CONCLUSIONS: These data reveal a novel signal integration mechanism involving NCK1/2, ROBO1/2, and VEGFR2 that controls endothelial cell front-rear polarity during sprouting angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Polaridad Celular/fisiología , Células Endoteliales/fisiología , Eliminación de Gen , Neovascularización Fisiológica/fisiología , Proteínas Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Secuencia de Aminoácidos , Animales , Marcación de Gen/métodos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Oncogénicas/deficiencia
11.
Proc Natl Acad Sci U S A ; 112(50): 15432-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621720

RESUMEN

Migration of the cells in osteoblastic lineage, including preosteoblasts and osteoblasts, has been postulated to influence bone formation. However, the molecular bases that link preosteoblastic/osteoblastic cell migration and bone formation are incompletely understood. Nck (noncatalytic region of tyrosine kinase; collectively referred to Nck1 and Nck2) is a member of the signaling adaptors that regulate cell migration and cytoskeletal structures, but its function in cells in the osteoblastic lineage is not known. Therefore, we examined the role of Nck in migration of these cells. Nck is expressed in preosteoblasts/osteoblasts, and its knockdown suppresses migration as well as cell spreading and attachment to substrates. In contrast, Nck1 overexpression enhances spreading and increases migration and attachment. As for signaling, Nck double knockdown suppresses migration toward IGF1 (insulin-like growth factor 1). In these cells, Nck1 binds to IRS-1 (insulin receptor substrate 1) based on immunoprecipitation experiments using anti-Nck and anti-IRS-1 antibodies. In vivo, Nck knockdown suppresses enlargement of the pellet of DiI-labeled preosteoblasts/osteoblasts placed in the calvarial defects. Genetic experiments indicate that conditional double deletion of both Nck1 and Nck2 specifically in osteoblasts causes osteopenia. In these mice, Nck double deficiency suppresses the levels of bone-formation parameters such as bone formation rate in vivo. Interestingly, bone-resorption parameters are not affected. Finally, Nck deficiency suppresses repair of bone injury after bone marrow ablation. These results reveal that Nck regulates preosteoblastic/osteoblastic migration and bone mass.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Huesos/citología , Movimiento Celular , Proteínas Oncogénicas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Técnicas de Silenciamiento del Gen , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Tamaño de los Órganos , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Radiografía , Cráneo/efectos de los fármacos , Cráneo/metabolismo , Cicatrización de Heridas/efectos de los fármacos
12.
Neuroscience ; 311: 539-51, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26546471

RESUMEN

Parkinson's disease (PD) is a well-characterized neurological disorder with regard to its neuropathological and symptomatic appearance. At the genetic level, mutations of particular genes, e.g. Parkin and DJ-1, were found in human hereditary PD with early onset. Neurotransmitter receptors constitute decisive elements in neural signal transduction. Furthermore, since they are often altered in neurological and psychiatric diseases, receptors have been successful targets for pharmacological agents. However, the consequences of PD-associated gene mutations on the expression of transmitter receptors are largely unknown. Therefore, we studied the expression of 16 different receptor binding sites of the neurotransmitters glutamate, GABA, acetylcholine, adrenaline, serotonin, dopamine and adenosine by means of quantitative receptor autoradiography in Parkin and DJ-1 knockout mice. These knockout mice exhibit electrophysiological and behavioral deficits, but do not show the typical dopaminergic cell loss. We demonstrated differential changes of binding site densities in eleven brain regions. Most prominently, we found an up-regulation of GABA(B) and kainate receptor densities in numerous cortical areas of Parkin and DJ-1 knockout mice, as well as increased NMDA but decreased AMPA receptor densities in different brain regions of the Parkin knockout mice. The alterations of three different glutamate receptor types may indicate the potential relevance of the glutamatergic system in the pathogenesis of PD. Furthermore, the cholinergic M1, M2 and nicotinic receptors as well as the adrenergic α2 and the adenosine A(2A) receptors showed differentially increased densities in Parkin and DJ-1 knockout mice. Taken together, knockout of the PD-associated genes Parkin or DJ-1 results in differential changes of neurotransmitter receptor densities, highlighting a possible role of altered non-dopaminergic, and in particular of glutamatergic neurotransmission in PD pathogenesis.


Asunto(s)
Encéfalo/metabolismo , Proteínas Oncogénicas/genética , Peroxirredoxinas/genética , Receptores de Neurotransmisores/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Autorradiografía , Encéfalo/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Peroxirredoxinas/deficiencia , Proteína Desglicasa DJ-1 , Ubiquitina-Proteína Ligasas/deficiencia
13.
Life Sci ; 143: 50-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26408915

RESUMEN

AIMS: The purpose of this study is to explore whether antioxidant DJ-1 protein affects the atrophy of skeletal muscle cell induced by undernutrition. MAIN METHODS: To determine cell atrophic responses, L6 cell line and skeletal primary cells from mouse hind limbs were cultivated under condition of FBS-free and low glucose. Changes of protein expression were analyzed using Western blot. Overexpression and knockdown of DJ-1 was performed in cells to assess its influence on cell atrophic responses. KEY FINDINGS: Undernutrition decreased cell size and increased the abundance of oxidized form and total form of DJ-1 protein in L6 myoblasts. The undernourished cells revealed an elevation in the expression of muscle-specific RING finger-1 (MuRF-1) and atrogin-1, and in the phosphorylations of p38 mitogen-activated protein kinase (MAPK) and stress-activated protein kinase/c-Jun N-terminal kinase compared with control groups. Moreover, DJ-1-knockout mice showed a decrease in cell size and an enhancement in the expression of MuRF-1 and atrogin-1, as well as in the phosphorylation of MAPKs in gastrocnemius muscles; these changes were also observed in L6 cells transfected with siRNA of DJ-1. On the other hand, L6 cells overexpressing full-length DJ-1 did not exhibit the alterations in cell size and ubiquitin ligases seen after undernourished states of control cells. Myotubes differentiated from L6 cells also showed elevated expression of MuRF-1 and atrogin-1 in response to undernutrition. SIGNIFICANCE: These results suggest that DJ-1 protein may contribute to undernutrition-induced atrophy via MAPKs/ubiquitin ligase pathway in skeletal muscle cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Desnutrición/metabolismo , Mioblastos/enzimología , Proteínas Oncogénicas/deficiencia , Peroxirredoxinas/deficiencia , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Atrofia/enzimología , Atrofia/prevención & control , Línea Celular , Femenino , Masculino , Desnutrición/prevención & control , Ratones , Ratones Noqueados , Mioblastos/patología , Técnicas de Cultivo de Órganos , Proteína Desglicasa DJ-1 , Ratas
14.
Hypertension ; 66(1): 158-66, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25987661

RESUMEN

Activation of the mineralocorticoid receptor has been shown to be deleterious in cardiovascular diseases (CVDs). We have recently shown that lipocalin 2 (Lcn2), or neutrophil gelatinase-associated lipocalin (NGAL), is a primary target of aldosterone/mineralocorticoid receptor in the cardiovascular system. Lcn2 is a circulating protein, which binds matrix metalloproteinase 9 and modulates its stability. We hypothesized that Lcn2 could be a mediator of aldosterone/mineralocorticoid receptor profibrotic effects in the cardiovascular system. Correlations between aldosterone and profibrotic markers, such as procollagen type I N-terminal peptide, were investigated in healthy subjects and subjects with abdominal obesity. The implication of Lcn2 in the mineralocorticoid pathway was studied using Lcn2 knockout mice subjected to a nephrectomy/aldosterone/salt (NAS) challenge for 4 weeks. In human subjects, NGAL/matrix metalloproteinase 9 was positively correlated with plasma aldosterone and fibrosis biomarkers. In mice, loss of Lcn2 prevented the NAS-induced increase of plasma procollagen type I N-terminal peptide, as well as the increase of collagen fibers deposition and collagen I expression in the coronary vessels and the aorta. The lack of Lcn2 also blunted the NAS-induced increase in systolic blood pressure. Ex vivo, treatment of human fibroblasts with recombinant Lcn2 induced the expression of collagen I and the profibrotic galectin-3 and cardiotrophin-1 molecules. Our results showed that Lcn2 plays a key role in aldosterone/mineralocorticoid receptor-mediated vascular fibrosis. The clinical data indicate that this may translate in human patients. Lcn2 is, therefore, a new biotarget in cardiovascular fibrosis induced by mineralocorticoid activation.


Asunto(s)
Proteínas de Fase Aguda/fisiología , Aldosterona/toxicidad , Lipocalinas/fisiología , Obesidad Abdominal/fisiopatología , Proteínas Oncogénicas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas de Fase Aguda/deficiencia , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/farmacología , Aldosterona/sangre , Animales , Aorta/efectos de los fármacos , Aorta/patología , Cardiomiopatía Hipertrófica/inducido químicamente , Cardiomiopatía Hipertrófica/fisiopatología , Células Cultivadas , Citocinas/biosíntesis , Citocinas/genética , Femenino , Fibroblastos , Fibrosis , Galectina 3/biosíntesis , Galectina 3/sangre , Galectina 3/genética , Humanos , Hipertensión/fisiopatología , Hipertrofia , Riñón/patología , Lipocalina 2 , Lipocalinas/sangre , Lipocalinas/genética , Lipocalinas/farmacología , Masculino , Ratones , Miocardio/citología , Miocardio/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Nefrectomía/efectos adversos , Obesidad Abdominal/sangre , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Fragmentos de Péptidos/sangre , Procolágeno/sangre , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/farmacología , Ratas , Proteínas Recombinantes/farmacología
15.
J Leukoc Biol ; 98(3): 301-11, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25995205

RESUMEN

Understanding the molecular mechanisms regulating T cell reactivity is required for successful reprogramming of immune responses in medical conditions, characterized by dysfunctions of the immune system. Nck proteins are cytoplasmic adaptors mediating diverse cellular functions, including TCR signaling. By enhancing TCR signal strength, Nck proteins influence thymic selection and regulate the size and sensitivity of the peripheral T cell repertoire. Here, we investigated the contribution of Nck proteins to CD4(+) T cell differentiation and effector function using Nck.T(-/-) mice. Impaired GC formation and reduced Tfh were observed in Nck.T(-/-) mice after immunization with T cell-dependent antigens. Th2/Tfh-related cytokines, such as IL-4, IL-10, and IL-21, were decreased in Nck.T(-/-) mice T cells. Moreover, an increased susceptibility to cell death of Tfh cells in Nck.T(-/-) mice was associated with decreased levels of Akt phosphorylation. As a result of this dysregulation in Tfh cells of Nck.T(-/-) mice, we found impaired production and affinity maturation of antibodies against T cell-dependent antigens. Thus, Nck proteins not only participate in thymic selection and generation of the peripheral T cell repertoire but also are involved in the differentiation and effector functions of CD4(+) T cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular , Proteínas Oncogénicas/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Formación de Anticuerpos , Apoptosis , Citocinas/biosíntesis , Eliminación de Gen , Centro Germinal/citología , Humanos , Ratones , Proteínas Oncogénicas/deficiencia , Factores de Transcripción/metabolismo
16.
Cell Res ; 25(6): 691-706, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26021615

RESUMEN

Inappropriate inflammation responses contribute to mortality during sepsis. Through Toll-like receptors (TLRs), reactive oxygen species (ROS) produced by NADPH oxidase could modulate the inflammation responses. Parkinson disease (autosomal recessive, early onset) 7 (Park7) has a cytoprotective role by eliminating ROS. However, whether Park7 could modulate inflammation responses and mortality in sepsis is unclear. Here, we show that, compared with wild-type mice, Park7(-/-) mice had significantly increased mortality and bacterial burdens in sepsis model along with markedly decreased systemic and local inflammation, and drastically impaired macrophage phagocytosis and bacterial killing abilities. Surprisingly, LPS and phorbol-12-myristate-13-acetate stimulation failed to induce ROS and proinflammatory cytokine production in Park7(-/-) macrophages and Park7-deficient RAW264.7 cells. Through its C-terminus, Park7 binds to p47(phox), a subunit of the NADPH oxidase, to promote NADPH oxidase-dependent production of ROS. Restoration of Park7 expression rescues ROS production and improves survival in LPS-induced sepsis. Together, our study shows that Park7 has a protective role against sepsis by controlling macrophage activation, NADPH oxidase activation and inflammation responses.


Asunto(s)
NADPH Oxidasas/metabolismo , Proteínas Oncogénicas/metabolismo , Peroxirredoxinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sepsis/prevención & control , Animales , Lipopolisacáridos/administración & dosificación , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/deficiencia , Peroxirredoxinas/biosíntesis , Peroxirredoxinas/deficiencia , Proteína Desglicasa DJ-1 , Sepsis/inducido químicamente , Sepsis/metabolismo
17.
Liver Int ; 35(4): 1195-202, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25040147

RESUMEN

BACKGROUND & AIMS: Various immune mediators such as interleukin-6 (IL-6) have been implicated in the process of liver regeneration. Lipocalin-2 (LCN2) has been recently characterized as a prototypic immune mediator produced by various cell types being involved mainly in host defence. In addition, numerous studies have demonstrated its clinical value as a biomarker. This study aimed at defining the role of LCN2 in liver regeneration. METHODS: We studied LCN2 expression in wild-type mice in a model of partial hepatectomy (PH). Furthermore, we evaluated liver regeneration after PH in LCN-deficient mice compared to littermate controls. Serum levels of LCN2 were assessed in a small group of patients undergoing hepatic resection. RESULTS: LCN2 is dramatically induced in livers and sera of wild-type mice after PH, whereas liver LCN2-receptor expression was decreased. Sham operations did not affect hepatic and serum LCN2 expression. Although LCN2-deficient mice exhibited increased baseline liver expression indices, LCN2-deficient mice did not differ from wild-type mice with respect to hepatic proliferation suggesting that this molecule is not involved in hepatic repair. Only serum IL-1ß levels were slightly lower in LCN(-/-) mice, whereas IL-6 serum levels did not differ between various tested animal groups. In humans undergoing hepatic resection, LCN2 levels increased significantly within 24 h following surgery. CONCLUSIONS: LCN2, although massively induced in mice after PH, is not relevant in murine hepatic regeneration. Further, human studies have to define whether LCN2 could evolve as biomarker after liver surgery.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Lipocalinas/sangre , Lipocalinas/metabolismo , Regeneración Hepática , Hígado/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/sangre , Proteínas de Fase Aguda/deficiencia , Proteínas de Fase Aguda/genética , Adulto , Anciano , Animales , Biomarcadores/sangre , Femenino , Hepatectomía/métodos , Heterocigoto , Homocigoto , Humanos , Interleucina-6/sangre , Lipocalina 2 , Lipocalinas/genética , Hígado/fisiopatología , Hígado/cirugía , Masculino , Ratones Noqueados , Persona de Mediana Edad , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Fenotipo , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba
18.
J Nutr Biochem ; 26(1): 75-81, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25448609

RESUMEN

DJ-1 constitutes a ubiquitously expressed, oxidative stress-responsive protein with multiple functions. DJ-1 emerged as a candidate from our previous proteome analysis investigating alterations in the hypothalamus in three mouse strains differing in their susceptibility to diet-induced obesity (DIO). Validation studies demonstrated a high-fat diet (HFD)-induced shift in the DJ-1 isoform pattern in the hypothalamus and several other tissues of mice. Others found HFD-induced alterations in DJ-1 protein abundance in adipose tissue and pancreatic islets in wild-type rodents. Here, we investigated the gene-diet interaction by challenging Dj-1(-/-) mice with a HFD. We demonstrate that the development of diet-induced obesity (DIO) Dj-1(-/-) mice is according to wild-type mice with the exception of transient higher gains in fat mass at the expense of lean mass after 14 weeks of feeding.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Obesidad/fisiopatología , Proteínas Oncogénicas/deficiencia , Peroxirredoxinas/deficiencia , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Calorimetría Indirecta , Ingestión de Energía , Femenino , Prueba de Tolerancia a la Glucosa , Hipotálamo/metabolismo , Insulina/sangre , Islotes Pancreáticos/metabolismo , Leptina/administración & dosificación , Leptina/sangre , Masculino , Ratones , Ratones Noqueados , Proteínas Oncogénicas/genética , Estrés Oxidativo , Peroxirredoxinas/genética , Proteína Desglicasa DJ-1 , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Análisis de Secuencia de ARN
19.
PLoS One ; 9(10): e109597, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25313900

RESUMEN

Hedgehog (Hh) signaling plays important roles in various development processes. This signaling is necessary for osteoblast formation during endochondral ossification. In contrast to the established roles of Hh signaling in embryonic bone formation, evidence of its roles in adult bone homeostasis is not complete. Here we report the involvement of Gli1, a transcriptional activator induced by Hh signaling activation, in postnatal bone homeostasis under physiological and pathological conditions. Skeletal analyses of Gli1+/- adult mice revealed that Gli1 haploinsufficiency caused decreased bone mass with reduced bone formation and accelerated bone resorption, suggesting an uncoupling of bone metabolism. Hh-mediated osteoblast differentiation was largely impaired in cultures of Gli1+/- precursors, and the impairment was rescued by Gli1 expression via adenoviral transduction. In addition, Gli1+/- precursors showed premature differentiation into osteocytes and increased ability to support osteoclastogenesis. When we compared fracture healing between wild-type and Gli1+/- adult mice, we found that the Gli1+/- mice exhibited impaired fracture healing with insufficient soft callus formation. These data suggest that Gli1, acting downstream of Hh signaling, contributes to adult bone metabolism, in which this molecule not only promotes osteoblast differentiation but also represses osteoblast maturation toward osteocytes to maintain normal bone homeostasis.


Asunto(s)
Haploinsuficiencia , Proteínas Oncogénicas/genética , Transactivadores/genética , Animales , Densidad Ósea , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Fémur/diagnóstico por imagen , Fémur/patología , Fracturas Óseas/patología , Regulación de la Expresión Génica , Genotipo , Proteínas Hedgehog/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Radiografía , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Transactivadores/deficiencia , Transactivadores/metabolismo , Proteína con Dedos de Zinc GLI1
20.
FEBS Lett ; 588(21): 3808-15, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25218436

RESUMEN

The Nck adapter protein is involved in key cellular functions, such as actin polymerization and reorganization, serving as a molecular bridge between the surface complex essential for foreign antigen recognition, the T-cell antigen receptor (TCR), and the actin machinery. However, the mechanisms regulating Nck expression and functions are unknown. In this study, we revealed Nck negative regulation and demonstrated that Nck is ubiquitylated following cellular activation. We identified the molecular determinants and mediators involved in this process. Our data suggest that Nck ubiquitylation might serve as a mechanism controlling Nck-mediated effector functions during cellular activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación hacia Abajo , Proteínas Oncogénicas/metabolismo , Ubiquitinación , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Adhesión Celular , Silenciador del Gen , Células HEK293 , Humanos , Células Jurkat , Mutación , Proteínas Oncogénicas/química , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-cbl/metabolismo , ARN Interferente Pequeño/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...