Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Immunol ; 45(8): 580-596, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39060140

RESUMEN

The guanine nucleotide exchange factor (GEF) VAV1, a previously 'undruggable' protein integral to T/B lymphocyte antigen-receptor signaling, promotes actin polymerization, immunological synapse formation, T cell activation and differentiation, and cytokine production. With the development of novel modalities for targeting proteins, we hypothesize that interventions targeting VAV1 will have therapeutic potential in T and T/B cell-mediated autoimmune and chronic inflammatory diseases. This opinion is supported by recent CRISPR-Cas9 studies showing VAV1 as a key positive regulator of T cell receptor (TCR) activation and cytokine production in primary human CD4+ and CD8+ T cells; data demonstrating that loss/suppression of VAV1 regulates autoimmunity and inflammation; and promising preclinical data from T and T/B cell-mediated disease models of arthritis and colitis showing the effectiveness of selective VAV1 targeting via protein degradation.


Asunto(s)
Enfermedades Autoinmunes , Inflamación , Proteínas Proto-Oncogénicas c-vav , Proteínas Proto-Oncogénicas c-vav/metabolismo , Humanos , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Animales , Inflamación/inmunología , Enfermedad Crónica , Terapia Molecular Dirigida , Transducción de Señal , Autoinmunidad , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Activación de Linfocitos/inmunología
2.
Eur J Immunol ; 53(3): e2250147, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36541400

RESUMEN

VAV1-MYO1F is a recently identified gain-of-function fusion protein of the proto-oncogene Vav guanine nucleotide exchange factor 1 (VAV1) that is recurrently detected in T-cell non-Hodgkin's lymphoma (T-NHL) patients. However, the pathophysiological functions of VAV1-MYO1F in lymphomagenesis are insufficiently defined. Therefore, we generated transgenic mouse models to conditionally express VAV1-MYO1F in T-cells in vivo. We demonstrate that VAV1-MYO1F triggers cell autonomous activation of T-cell signaling with an activation of the ERK, JNK, and AKT pathways. VAV1-MYO1F expression induces a T-cell activation phenotype with high surface expression of CD25, ICOS, CD44, PD-1, and decreased CD62L as well as aberrant T-cell differentiation, proliferation, and neoplastic transformation. Consequently, the VAV1-MYO1F expressing T-cells induce a malignant T lymphoproliferative disease with 100% penetrance in vivo that mimics key aspects of human peripheral T-cell lymphoma. These results demonstrate that the human T-cell oncogene VAV1-MYO1F is sufficient to trigger oncogenic T-cell signaling and neoplastic transformation, and moreover, it provides a new clinically relevant mouse model to explore the pathogenesis of and treatment concepts for human T-cell lymphoma.


Asunto(s)
Linfoma de Células T Periférico , Proteínas Proto-Oncogénicas c-vav , Ratones , Humanos , Animales , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Linfoma de Células T Periférico/genética , Transducción de Señal , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Ratones Transgénicos , Oncogenes , Miosina Tipo I/genética , Miosina Tipo I/metabolismo
3.
Scand J Immunol ; 98(2): e13283, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38441379

RESUMEN

Multiple sclerosis (MS) is an inflammatory autoimmune disorder of the central nervous system and the leading cause of progressive neurological disability in young adults. It decreases the patient's lifespan by about 10 years and affects women more than men. No medication entirely restricts or reverses neurological degradation. However, early diagnosis and treatment increase the possibility of a better outcome. To identify new MS biomarkers, we tested the expression of six potential markers (P2X4, P2X7, CXCR4, RGS1, RGS16 and VAV1) using qPCR in peripheral blood mononuclear cells (PBMC) of MS patients treated with interferon ß (IFNß), with glatiramer acetate (GA) or untreated. We showed that P2X7 and VAV1 are significantly induced in MS patients. In contrast, the expression of P2X4, CXCR4, RGS1 and RGS16 was not significantly modified by MS in PBMC. P2X7 and VAV1 are essentially induced in female patients, suggesting these markers are connected to sex-specific mechanisms. Strikingly, VAV1 expression is higher in healthy women than healthy men and IFNß treatment of MS reduced VAV1 expression in female MS patients while it up-regulated VAV1 in male MS patients. Our data point to the differential, sex-dependent value of MS markers and treatment effects. Although rgs16 expression in PBMC was not a valid MS marker in patients, the strong upregulation of P2X4 and P2X7 induced in the spinal cord of WT mice by EAE was abrogated in rgs16KO mice suggesting that rgs16 is required for P2X4 and P2X7 induction by neurological diseases.


Asunto(s)
Enfermedades Autoinmunes , Esclerosis Múltiple , Animales , Femenino , Humanos , Masculino , Ratones , Adulto Joven , Sistema Nervioso Central , Interferón beta/uso terapéutico , Leucocitos Mononucleares , Esclerosis Múltiple/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-vav/genética
4.
Cell Biol Int ; 47(11): 1835-1842, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37545183

RESUMEN

Hypoxia has been shown to induce gemcitabine (GEM) resistance in pancreatic ductal adenocarcinoma (PDAC) cells, however, the underlying mechanisms remain to be clarified. In the present study, we investigated whether activation of Vav1/Rac1/HIF-1α axis is responsible for hypoxia-induced GEM resistance in PDAC cells. Our results showed that Rac1 activation contributed to hypoxia-induced GEM resistance in PANC-1 cells. Hypoxia treatment led to an increased expression level of Vav1, which was responsible for Rac1 activation and GEM resistance in PDAC cells. Furthermore, Rac1 mediated hypoxia-induced GEM resistance by upregulating HIF-1α in PDAC cells. Taken together, these findings suggest that hypoxia induces GEM resistance in PDAC cells by activating the Vav1/Rac1/HIF-1α signaling pathway.


Asunto(s)
Carcinoma Ductal Pancreático , Resistencia a Antineoplásicos , Gemcitabina , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Hipoxia , Neoplasias Pancreáticas/metabolismo , Regulación hacia Arriba , Neoplasias Pancreáticas
5.
J Biol Chem ; 296: 100527, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33705794

RESUMEN

Adult mammalian astrocytes are sensitive to inflammatory stimuli in the context of neuropathology or mechanical injury, thereby affecting functional outcomes of the central nervous system (CNS). In contrast, glial cells residing in the spinal cord of regenerative vertebrates exhibit a weak astroglial reaction similar to those of mammals in embryonic stages. Macrophage migration inhibitory factor (MIF) participates in multiple neurological disorders by activation of glial and immune cells. However, the mechanism of astrocytes from regenerative species, such as gecko astrocytes (gAS), in resistance to MIF-mediated inflammation in the severed cords remains unclear. Here, we compared neural stem cell markers among gAS, as well as adult (rAS) and embryonic (eAS) rat astrocytes. We observed that gAS retained an immature phenotype resembling rat eAS. Proinflammatory activation of gAS with gecko (gMIF) or rat (rMIF) recombinant protein was unable to induce the production of inflammatory cytokines, despite its interaction with membrane CD74 receptor. Using cross-species screening of inflammation-related mediators from models of gMIF- and rMIF-induced gAS and rAS, we identified Vav1 as a key regulator in suppressing the inflammatory activation of gAS. The gAS with Vav1 deficiency displayed significantly restored sensitivity to inflammatory stimuli. Meanwhile, gMIF acts to promote the migration of gAS through regulation of CXCL8 following cord lesion. Taken together, our results suggest that Vav1 contributes to the regulation of astrocyte-mediated inflammation, which might be beneficial for the therapeutic development of neurological diseases.


Asunto(s)
Astrocitos/inmunología , Mediadores de Inflamación/metabolismo , Inflamación/inmunología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Médula Espinal/inmunología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Biomarcadores/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Inflamación/metabolismo , Inflamación/patología , Factores Inhibidores de la Migración de Macrófagos/genética , Proteínas Proto-Oncogénicas c-vav/genética , Ratas , Reptiles , Médula Espinal/citología , Médula Espinal/metabolismo
6.
J Cell Sci ; 133(5)2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31974114

RESUMEN

Vav family guanine nucleotide exchange factors (GEFs) are essential regulators of immune function. Despite their structural similarity, Vav1 promotes and Vav2 opposes T cell receptor (TCR)-induced Ca2+ entry. By using a Vav1-deficient Jurkat T cell line, we find that Vav1 facilitates Ca2+ entry via non-catalytic scaffolding functions that are encoded by the catalytic core of Vav1 and flanking linker regions. We implicate, in this scaffolding function, a previously undescribed polybasic motif that is strictly conserved in Vav1 and absent from Vav2 in tetrapods. Conversely, the catalytic activity of Vav2 contributes to the suppression of TCR-mediated Ca2+ entry. By performing an in vivo 'GEF trapping' assay in intact cells, we demonstrate that Cdc42 interacts with the catalytic surface of Vav2 but not Vav1, and that Vav1 discriminates Cdc42 from Rac1 via F56 (W56 in Rac1). Finally, the Cdc42-specific inhibitor ZCL278 and the shRNA-mediated suppression of Cdc42 each prevent the inhibition of TCR-induced Ca2+ entry by Vav2. These findings define stark differences in the functions of Vav1 and Vav2, and provide an explanation for the differential usage of these Vav isoforms by immune subpopulations.


Asunto(s)
Activación de Linfocitos , Proteínas Proto-Oncogénicas c-vav , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos T , Linfocitos T
7.
Can J Physiol Pharmacol ; 100(6): 562-572, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35413215

RESUMEN

Mecobalamin is commonly used in the adjuvant intervention of various peripheral nerve injuries. Actin cytoskeleton plays a role in the regeneration of myelin and axon. Therefore, the purpose of this study was to explore the possibility of mecobalamin regulating actin cytoskeleton in repairing nerve injury. In this study, a crush injury on the right sciatic nerve of two groups of rats (12 in each group) was established. The control group was only given normal saline (i.g.), and the intervention group was given mecobalamin 1 mg/kg (i.g.). The rats were sacrificed on 28th day and the injured nerves were collected for proteomics. The result shows that regulation of actin cytoskeleton pathway changed significantly. The expression of protein Vav1 was verified by Western blot and immunofluorescence. In the intervention group, the nerve fiber structure was complete, the axons were dense and symmetrical, and the myelin sheath was compact and uniform in thickness. The positive rate of myelin basic protein and ßⅢ-tubulin was higher than that in the control group. The findings of the study show that mecobalamin regulates the actin cytoskeleton in the repair of nerve damage and upregulates Vav1 in the regulation of actin cytoskeleton pathway.


Asunto(s)
Traumatismos de los Nervios Periféricos , Proteómica , Animales , Axones/metabolismo , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-vav/metabolismo , Ratas , Nervio Ciático/metabolismo , Vitamina B 12/análogos & derivados
8.
Rinsho Ketsueki ; 63(12): 1657-1667, 2022.
Artículo en Japonés | MEDLINE | ID: mdl-36653140

RESUMEN

Peripheral T-cell lymphomas (PTCLs) are mature T-cell and natural killer (NK) cell neoplasms with poor prognosis and no available standard treatment. The 2016 revision of the World Health Organization classification of hematological malignancies recognized a new group of nodal T-cell lymphoma with T follicular helper cell phenotype due to the presence of a group of PTCLs, not otherwise specified. This new subgroup of tumors showed clinical and molecular similarities with angioimmunoblastic T-cell lymphoma (AITL). AITL patients have mutations in the Vav guanine nucleotide exchange factor-1 (VAV1) gene. We established a transgenic mouse model harboring VAV1 gene mutation. These mice developed tumors that mimic human T-lymphoblastic lymphoma and the newly proposed PTCL-GATA3 subtype. To develop personalized treatment strategies by analyzing subgroup-specific mouse models, our study supports the establishment of PTCL subgroups based on genetic alterations or gene expression profiles.


Asunto(s)
Linfadenopatía Inmunoblástica , Linfoma de Células T Periférico , Linfoma de Células T , Humanos , Animales , Ratones , Linfoma de Células T Periférico/genética , Linfoma de Células T/genética , Mutación , Fenotipo
9.
Biotechnol Appl Biochem ; 68(4): 849-855, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32767384

RESUMEN

Immunotherapy based on adoptive transfer of genetically engineered T- and NK-cells is an area of active ongoing research and has proven highly efficacious for patients with certain B-cell malignancies. Use of NK cells and NK cell lines as carriers of chimeric antigen receptors (CARs) appears particularly promising, as this opens an opportunity for moving the therapy from autologous to the allogeneic (universal) format. This "off-the-shelf" approach is thought to significantly reduce the price of the treatment and make it available to many more patients in need. Yet, the efficacy of CAR-NK cells in vivo presently remains low, and boosting the activity of CAR NK cells via stronger tumor homing, resistance to tumor microenvironment, as well as greater cytotoxicity may translate into improved patient outcomes. Here, we established a derivative of a human NK cell line YT overexpressing a positive regulator of cytotoxicity, VAV1. Activity of YT-VAV1 cells obtained was assayed in vitro against several cancer cell lines and primary patient-derived cancer cells. YT-VAV1 cells outperform parental YT cells in terms of cytotoxicity.


Asunto(s)
Inmunidad Celular , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Proteínas Proto-Oncogénicas c-vav/inmunología , Células CACO-2 , Células HEK293 , Humanos , Inmunoterapia , Células PC-3 , Proteínas Proto-Oncogénicas c-vav/genética
10.
J Cell Physiol ; 235(1): 480-493, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31385301

RESUMEN

Alzheimer's disease (AD) is a progressive and age-related neurological dysfunction. Abundant data have profiled microRNA (miR) patterns in healthy, aging brain, and in the moderate and late-stages of AD. Herein, this study aimed to explore whether miR-326 could influence neuron apoptosis in AD mice and how miR-326 functions in this process. The candidate differentially expressed gene VAV1 was obtained by microarray analysis, and miRNAs that could regulate VAV1 candidate gene were predicted. Luciferase activity determination confirmed VAV1 as a target gene of miR-326. AD mice models were established for investigating the effect of miR-326 on AD mice. The overexpression of miR-326 contributed to decreased time of the mice to find the platform and the escape latency and increased residence time on the target area. Besides, elevation of miR-326 decreased Aß deposition and contents of Aß1-40 and Aß1-42 . Moreover, miR-326 overexpression increased neuron cell ability, mediated cell entry, and inhibited neuron apoptosis via JNK signaling pathway. Of crucial importance, miR-326 negatively regulated the expression of VAV1, inhibited tau phosphorylation, and blocked the activation of the JNK signaling pathway. Taken together these observations, we demonstrate that miR-326 improves cognitive function of AD mice and inhibits neuron apoptosis in AD mice through inactivation of the JNK signaling pathway by targeting VAV1. Based on those findings, miR-326 might exert promise as target for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Apoptosis/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-vav/genética , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Sistema de Señalización de MAP Quinasas/genética , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Fosforilación , Proteínas tau/metabolismo
11.
Proc Natl Acad Sci U S A ; 114(4): 764-769, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28062691

RESUMEN

Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of non-Hodgkin lymphomas frequently associated with poor prognosis and for which genetic mechanisms of transformation remain incompletely understood. Using RNA sequencing and targeted sequencing, here we identify a recurrent in-frame deletion (VAV1 Δ778-786) generated by a focal deletion-driven alternative splicing mechanism as well as novel VAV1 gene fusions (VAV1-THAP4, VAV1-MYO1F, and VAV1-S100A7) in PTCL. Mechanistically these genetic lesions result in increased activation of VAV1 catalytic-dependent (MAPK, JNK) and non-catalytic-dependent (nuclear factor of activated T cells, NFAT) VAV1 effector pathways. These results support a driver oncogenic role for VAV1 signaling in the pathogenesis of PTCL.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Guanina/metabolismo , Linfoma de Células T Periférico/genética , Mutación/genética , Proteínas Proto-Oncogénicas c-vav/genética , Translocación Genética/genética , Empalme Alternativo/genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular Tumoral , Humanos , Células Jurkat , Eliminación de Secuencia/genética
12.
Int J Mol Sci ; 21(4)2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32079227

RESUMEN

Vav1 is a Rho/Rac (Ras-related C3 botulinum toxin substrate) guanine nucleotide exchange factor expressed in hematopoietic and endothelial cells that are involved in a wide range of cellular functions. It is also stabilized under hypoxic conditions when it regulates the accumulation of the transcription factor HIF (Hypoxia Inducible Factor)-1α, which activates the transcription of target genes to orchestrate a cellular response to low oxygen. One of the genes induced by HIF-1α is GLUT (Glucose Transporter)-1, which is the major glucose transporter expressed in vessels that supply energy to the brain. Here, we identify a role for Vav1 in providing glucose to the brain. We found that Vav1 deficiency downregulates HIF-1α and GLUT-1 levels in endothelial cells, including blood-brain barrier cells. This downregulation of GLUT-1, in turn, reduced glucose uptake to endothelial cells both in vitro and in vivo, and reduced glucose levels in the brain. Furthermore, endothelial cell-specific Vav1 knock-out in mice, which caused glucose uptake deficiency, also led to a learning delay in fear conditioning experiments. Our results suggest that Vav1 promotes learning by activating HIF-1α and GLUT-1 and thereby distributing glucose to the brain. We further demonstrate the importance of glucose transport by endothelial cells in brain functioning and reveal a potential new axis for targeting GLUT-1 deficiency syndromes and other related brain diseases.


Asunto(s)
Glucemia/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 1/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas Proto-Oncogénicas c-vav/genética , Animales , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-vav/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transfección
13.
Biochem Biophys Res Commun ; 511(4): 787-793, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30833082

RESUMEN

Vav1 is a guanine nucleotide exchange factor (GEF) predominantly expressed in hematopoietic cells, and functions in the development and antigen-stimulated response of lymphocytes. Burkitt's lymphoma (BL) is characterized as transformed B cell lymphoma, and is highly associated with Epstein-Barr virus (EBV). EBV nuclear antigen 1 (EBNA1) is the only viral protein expressed across all three types of latency and essential for the persistence of EBV genome. It is not clear yet how EBNA1 contributes to the growth advantage of latently infected cells such as in EBV+ lymphoma B cells. Here, we reported that Vav1 interacts with EBNA1 via its C-terminal SH3 domain. This interaction suppresses the expression of a pro-apoptotic Bcl-2 family member, Bim, resulting in the resistance of the BL cells to apoptotic inductions. Our data uncovered Vav1 as a novel target for EBNA1, and suggested a pro-survival role of Vav1 in the pathogenesis of EBV associated BLs.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Linfoma de Burkitt/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Linfoma de Burkitt/genética , Linfoma de Burkitt/virología , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Humanos , Mapas de Interacción de Proteínas
14.
J Cell Mol Med ; 22(6): 3149-3158, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29532991

RESUMEN

It has been recently demonstrated that high pre-treatment levels of miR-29b positively correlated with the response of patients with acute myeloid leukaemia (AML) to hypomethylating agents. Upmodulation of miR-29b by restoring its transcriptional machinery appears indeed a tool to improve therapeutic response in AML. In cells from acute promyelocytic leukaemia (APL), miR-29b is regulated by PU.1, in turn upmodulated by agonists currently used to treat APL. We explored here the ability of PU.1 to also regulate miR-29b in non-APL cells, in order to identify agonists that, upmodulating PU.1 may be beneficial in hypomethylating agents-based therapies. We found that PU.1 may regulate miR-29b in the non-APL Kasumi-1 cells, showing the t(8;21) chromosomal rearrangement, which is prevalent in AML and correlated with a relatively low survival. We demonstrated that the PU.1-mediated contribution of the 2 miR-29b precursors is cell-related and almost completely dependent on adequate levels of Vav1. Nuclear PU.1/Vav1 association accompanies the transcription of miR-29b but, at variance with the APL-derived NB4 cells, in which the protein is required for the association of PU.1 with both miRNA promoters, Vav1 is part of molecular complexes to the PU.1 consensus site in Kasumi-1. Our results add new information on the transcriptional machinery that regulates miR-29b expression in AML-derived cells and may help in identifying drugs useful in upmodulation of this miRNA in pre-treatment of patients with non-APL leukaemia who can take advantage from hypomethylating agent-based therapies.


Asunto(s)
Leucemia Mieloide Aguda/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Regulación Leucémica de la Expresión Génica/genética , Humanos , Leucemia Mieloide Aguda/patología , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Regiones Promotoras Genéticas
15.
J Cell Biochem ; 119(7): 5437-5448, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29369410

RESUMEN

This study aims to explore the effect of miR-330 targeting VAV1 on amyloid ß-protein (Aß) production, oxidative stress (OS), and mitochondrial dysfunction in Alzheimer's disease (AD) mice through the MAPK signaling pathway. Putative targeted gene of miR-330 was performed by a miRNA target prediction website and dual-luciferase reporter gene assay. AD mouse model was successfully established. Fourteen C57 mice were randomized into AD and control groups. The positive protein expression rate of VAV1 was measured by immunohistochemistry. Neuron cells were assigned into control, blank, negative control (NC), miR-330 mimics, miR-330 inhibitors, siRNA-VAV1, and miR-330 inhibitors + siRNA-VAV1 groups. Expression of miR-330, VAV1, ERK1, JNK1, P38MAPK, Aß, COX, and lipoprotein receptor-related protein-1 (LRP-1) were determined using RT-qPCR and Western blotting. Colorimetry was applied to measure the levels of OS parameters of superoxide dismutase (SOD) and malondialdehyde (MDA). Aß production in brain tissue was detected using ELISA, while that in neuron cell was measured by radioimmunoassay. MiR-330 was down-regulated in neuron cells of AD mice and VAV1 was negatively regulated by miR-330. Compared with the control group, the positive protein expression rate of VAV1 was significantly elevated in the AD group. Overexpression of miR-330 decreased the expression of VAV1, ERK1, JNK1, P38MAPK, and Aß, but increased the expression of COX and LRP-1. AD mice revealed elevated Aß production and MDA with decreased SOD level. The result indicates that overexpressed miR-330 targeting VAV1 through the MAPK signaling pathway reduces Aß production and alleviates OS and mitochondrial dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Sistema de Señalización de MAP Quinasas , MicroARNs/farmacología , Mitocondrias/genética , Estrés Oxidativo/genética , Sustancias Protectoras/farmacología , Proteínas Proto-Oncogénicas c-vav/antagonistas & inhibidores , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal
16.
Biochem Biophys Res Commun ; 486(2): 571-576, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28336434

RESUMEN

Recently, Vav1 has been suggested to play an essential role in the progression of human cancers. However, the correlation between Vav1 expression and prognosis of esophageal squamous cell carcinoma (ESCC) is still unknown. The aim of this study was to investigate Vav1 expression and its prognostic value in ESCC. The expression of Vav1 was detected by real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting in ESCC tissues and matched nontumorous tissues. Immunohistochemistry (IHC) was carried out to detect Vav1 expression in paraffin samples from 112 primary ESCC patients. Survival analysis was performed using Kaplan-Meier method. Univariate and multivariate Cox regression analyses were performed to evaluate the correlation of Vav1 expression with prognosis of ESCC patients. The expression levels of Vav1 mRNA and protein in ESCC tissues were both significantly higher than those in adjacent nontumorous tissues. High Vav1 expression was significantly correlated with larger tumor size (P = 0.015), depth of tumor invasion (P = 0.023), lymph node metastasis (P = 0.008) and TNM stage (P < 0.001). The rate of overall survival (OS) was significantly lower in patients with high Vav1 expression than those with low Vav1 expression (P = 0.014). Multivariate Cox analysis indicated that Vav1 expression is an independent prognostic factor for OS (HR = 1.660, 95%CI = 1.058-2.607, P = 0.028). In summary, our findings demonstrate that Vav1 may be a candidate molecular prognostic marker for patients with ESCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/diagnóstico , Neoplasias Esofágicas/diagnóstico , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-vav/genética , ARN Mensajero/genética , Adulto , Anciano , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Análisis Multivariante , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Análisis de Supervivencia
17.
Stem Cells ; 34(7): 1934-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26990002

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent stromal cells residing in the bone marrow. MSCs have the potential to differentiate to adipocytes, chondrocytes, and other types of cells. In this study, we investigated the molecular mechanism that controls MSC cell fate decisions for differentiation. We found that Vav1, a guanine nucleotide exchange factor for Rho GTPase, was highly expressed in MSCs. Interestingly, loss of Vav1 in MSCs led to spontaneous adipogenic but impaired chondrogenic differentiation, and accordingly Vav1 null mice displayed an increase in fat content and a decrease in cartilage. Conversely, ectopic expression of Vav1 in MSCs reversed this phenotype, and led to enhanced MSC differentiation into chondrocyte but retarded adipogenesis. Mechanistically, loss of Vav1 reduced the level of Sirt1, which was responsible for an increase of acetylated PPARγ. As acetylation activates PPARγ, it increased C/EBPα expression and promoted adipogenesis. On the other hand, loss of Vav1 resulted in an increase of acetylated Sox9, a target of Sirt1. As acetylation represses Sox9 activity, it led to a dramatic reduction of collagen 2α1, a key regulator in chondrocyte differentiation. Finally, we found that Vav1 regulates Sirt1 in MSCs through Creb. Together this study reveals a novel function of Vav1 in regulating MSC cell fate decisions for differentiation through Sirt1. Sirt1 deacetylates PPARγ and Sox9, two key mediators that control adipocyte and chondrocyte differentiation. The acetylation status of PPARγ and Sox9 has opposite effects on its activity, thereby controlling cell fate decision. Stem Cells 2016;34:1934-1946.


Asunto(s)
Adipocitos/citología , Diferenciación Celular , Condrocitos/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Sirtuina 1/metabolismo , Adipocitos/metabolismo , Adipogénesis/genética , Adiposidad , Animales , Diferenciación Celular/genética , Condrocitos/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Eliminación de Gen , Ratones Endogámicos C57BL , Factores de Transcripción/metabolismo
18.
Blood Cells Mol Dis ; 59: 58-62, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27282569

RESUMEN

Imatinib (IM) resistant Chronic Myeloid Leukemia (CML) is an important issue to be addressed while treating CML patients. The present study analyzes the role of EphB4-VAV1 signaling in IM-resistant CML. EphB4 and VAV1 were overexpressed in IM-resistant CML patients and K562-R cell line (K562-R). Then, we established stable under-expressing EphB4 cell line K562-R-EphB4-sh. VAV1 was down-regulated in K562-R-EphB4-sh cells. K562-R-EphB4-sh cells gained re-sensitivity to IM and K562-R cells showed mild IM resistance. However, EphB4 was no changed when the VAV1 was down-regulated. EphB4 and VAV1 were overexpressed in IM-resistant CML, VAV1might be the downstream moleculars of EphB4. These results suggest a potential role of EphB4-VAV1 signaling as therapeutic target of IM-resistant CML.


Asunto(s)
Resistencia a Antineoplásicos , Mesilato de Imatinib/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptor EphB4/metabolismo , Humanos , Mesilato de Imatinib/uso terapéutico , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Transducción de Señal
19.
J Proteome Res ; 14(7): 2963-75, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26043137

RESUMEN

Vav1, a Rac/Rho guanine nucleotide exchange factor and a critical component of the T-cell receptor (TCR) signaling cascade is tyrosine phosphorylated rapidly in response to T-cell activation. Vav1 has established roles in proliferation, cytokine secretion, Ca(2+) responses, and actin cytoskeleton regulation; however, its function in the regulation of phosphorylation of TCR components, including the ζ chain, the CD3 δ, ε, γ chains, and the associated kinases Lck and ZAP-70, is not well established. To obtain a more comprehensive picture of the role of Vav1 in receptor proximal signaling, we performed a wide-scale characterization of Vav1-dependent tyrosine phosphorylation events using quantitative phosphoproteomic analysis of Vav1-deficient T cells across a time course of TCR stimulation. Importantly, this study revealed a new function for Vav1 in the negative feedback regulation of the phosphorylation of immunoreceptor tyrosine-based activation motifs within the ζ chains, CD3 δ, ε, γ chains, as well as activation sites on the critical T cell tyrosine kinases Itk, Lck, and ZAP-70. Our study also uncovered a previously unappreciated role for Vav1 in crosstalk between the CD28 and TCR signaling pathways.


Asunto(s)
Antígenos CD28/metabolismo , Activación de Linfocitos/fisiología , Proteínas Proto-Oncogénicas c-vav/fisiología , Receptor Cross-Talk , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología , Humanos , Células Jurkat , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal , Proteína Tirosina Quinasa ZAP-70/metabolismo
20.
Protein Pept Lett ; 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39301900

RESUMEN

OBJECTIVE: The aim was to investigate the potential relationship between Vav1 protein and prognosis in patients with hepatocellular carcinoma (HCC). METHODS: Samples were collected from 96 patients with HCC. For each patient, cancerous tissue and adjacent non-cancerous tissue were obtained. The Vav1 expression levels in these tissues were determined using immunohistochemistry. Chi-square and Fisher's exact tests were used to analyse the associations between Vav1 expression and clinicopathological characteristics. Kaplan- Meier analysis was used to assess the relationship between Vav1 expression and 5-year overall survival (OS). RESULTS: The expression level of Vav1 protein in primary tumour samples (64.46%; 59/96) was higher (33.33%; 32/96; P<0.001). Moreover, the high expression rate of Vav1 was correlated with tumour differentiation, TNM stage, and tumour recurrence (P<0.05). Univariate and multivariate Cox analysis further demonstrated that tumour differentiation, TNM stage, vascular invasion, tumour recurrence and Vav1 expression were independent prognostic factors for 5-year OS. Notably, follow-up analysis determined that patients with HCC with higher Vav1 expression levels have lower survival rates (P<0.05). CONCLUSION: Vav1 may serve as a promising molecular prognostic biomarker for patients diagnosed with HCC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA