Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
1.
Cancer Lett ; 600: 217153, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39102940

RESUMEN

The transforming growth factor-ß (TGF-ß) signaling pathway is pivotal in inducing epithelial-mesenchymal transition (EMT) and promoting cancer metastasis. Long non-coding RNAs (lncRNAs) have emerged as significant players in these processes, yet their precise mechanisms remain elusive. Here, we demonstrate that TGF-ß-upregulated lncRNA 1 (TBUR1) is significantly activated by TGF-ß via Smad3/4 signaling in lung adenocarcinoma (LUAD) cells. Functionally, TBUR1 triggers EMT, enhances LUAD cell migration and invasion in vitro, and promotes metastasis in nude mice. Mechanistically, TBUR1 interacts with heterogeneous nuclear ribonucleoprotein C (hnRNPC) to stabilize GRB2 mRNA in an m6A-dependent manner. Clinically, TBUR1 is upregulated in LUAD tissues and correlates with poor prognosis, highlighting its potential as a prognostic biomarker and therapeutic target for LUAD. Taken together, our findings underscore the crucial role of TBUR1 in mediating TGF-ß-induced EMT and metastasis in LUAD, providing insights for future therapeutic interventions.


Asunto(s)
Adenocarcinoma del Pulmón , Transición Epitelial-Mesenquimal , Proteína Adaptadora GRB2 , Neoplasias Pulmonares , Ratones Desnudos , ARN Largo no Codificante , Factor de Crecimiento Transformador beta , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Animales , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular , Línea Celular Tumoral , Estabilidad del ARN , Transducción de Señal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células A549 , Masculino , Femenino , Metástasis de la Neoplasia
2.
J Gen Virol ; 105(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39136113

RESUMEN

Porcine deltacoronavirus (PDCoV), an enteropathogenic coronavirus, causes severe watery diarrhoea, dehydration and high mortality in piglets, which has the potential for cross-species transmission in recent years. Growth factor receptor-bound protein 2 (Grb2) is a bridging protein that can couple cell surface receptors with intracellular signal transduction events. Here, we investigated the reciprocal regulation between Grb2 and PDCoV. It is found that Grb2 regulates PDCoV infection and promotes IFN-ß production through activating Raf/MEK/ERK/STAT3 pathway signalling in PDCoV-infected swine testis cells to suppress viral replication. PDCoV N is capable of interacting with Grb2. The proline-rich motifs in the N- or C-terminal region of PDCoV N were critical for the interaction between PDCoV-N and Grb2. Except for Deltacoronavirus PDCoV N, the Alphacoronavirus PEDV N protein could interact with Grb2 and affect the regulation of PEDV replication, while the N protein of Betacoronavirus PHEV and Gammacoronavirus AIBV could not interact with Grb2. PDCoV N promotes Grb2 degradation by K48- and K63-linked ubiquitin-proteasome pathways. Overexpression of PDCoV N impaired the Grb2-mediated activated effect on the Raf/MEK/ERK/STAT3 signal pathway. Thus, our study reveals a novel mechanism of how host protein Grb2 protein regulates viral replication and how PDCoV N escaped natural immunity by interacting with Grb2.


Asunto(s)
Proteína Adaptadora GRB2 , Proteínas de la Nucleocápside , Replicación Viral , Animales , Porcinos , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Proteínas de la Nucleocápside/metabolismo , Proteínas de la Nucleocápside/genética , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/metabolismo , Deltacoronavirus/metabolismo , Deltacoronavirus/genética , Sistema de Señalización de MAP Quinasas , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/metabolismo , Humanos , Transducción de Señal , Línea Celular , Quinasas raf/metabolismo , Quinasas raf/genética , Células HEK293
3.
Biochem Biophys Res Commun ; 728: 150325, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-38959529

RESUMEN

RHOV and RHOU are considered atypical Rho-family small GTPases because of the existence of N- and C-terminal extension regions, abnormal GDP/GTP cycling, and post-translational modification. Particularly, RHOV and RHOU both have a proline-rich (PR) motif in the N-terminal region. It has been reported that the PR motif of RHOU interacts with GRB2, a SH3 domain-containing adaptor protein, and regulates its activity through EGF receptor signaling. However, it is unknown whether RHOV, like RHOU, interacts with SH3 domain-containing adaptor proteins. In this study, we investigated the interactions between RHOV and SH3 domain-containing adaptor proteins, including GRB2 and NCK2. The RHOV-induced serum response factor (SRF)-dependent gene transcriptional activity was attenuated in cells co-expressing either GRB2 or NCK2 compared to cells expressing RHOV alone. From the results of experiments using various gene mutants of RHOV and GRB2, it appears that the PR motif of the N-terminal region of RHOV is the crucial binding site for the SH3 domain-containing proteins. Furthermore, we found that Ser25 in the N-terminal region of RHOV is phosphorylated by PKA and that its phosphorylation is suppressed by interaction with NCK2 but not GRB2. We have found a novel regulatory mechanism for the phosphorylation of RHOV and its interaction with SH3 domain-containing adaptor proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Quinasas Dependientes de AMP Cíclico , Proteína Adaptadora GRB2 , Transducción de Señal , Dominios Homologos src , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Células HEK293 , Proteínas Oncogénicas/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Fosforilación , Unión Proteica
4.
Mol Med ; 30(1): 82, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862918

RESUMEN

BACKGROUND: Programmed cell death is an important mechanism for the development of hepatic ischemia and reperfusion (IR) injury, and multiple novel forms of programmed cell death are involved in the pathological process of hepatic IR. ERRFI1 is involved in the regulation of cell apoptosis in myocardial IR. However, the function of ERRFI1 in hepatic IR injury and its modulation of programmed cell death remain largely unknown. METHODS: Here, we performed functional and molecular mechanism studies in hepatocyte-specific knockout mice and ERRFI1-silenced hepatocytes to investigate the significance of ERRFI1 in hepatic IR injury. The histological severity of livers, enzyme activities, hepatocyte apoptosis and ferroptosis were determined. RESULTS: ERRFI1 expression increased in liver tissues from mice with IR injury and hepatocytes under oxygen-glucose deprivation/reoxygenation (OGD/R) conditions. Hepatocyte-specific ERRFI1 knockout alleviated IR-induced liver injury in mice by reducing cell apoptosis and ferroptosis. ERRFI1 knockdown reduced apoptotic and ferroptotic hepatocytes induced by OGD/R. Mechanistically, ERRFI1 interacted with GRB2 to maintain its stability by hindering its proteasomal degradation. Overexpression of GRB2 abrogated the effects of ERRFI1 silencing on hepatocyte apoptosis and ferroptosis. CONCLUSIONS: Our results revealed that the ERRFI1-GRB2 interaction and GRB2 stability are essential for ERRFI1-regulated hepatic IR injury, indicating that inhibition of ERRFI1 or blockade of the ERRFI1-GRB2 interaction may be potential therapeutic strategies in response to hepatic IR injury.


Asunto(s)
Apoptosis , Ferroptosis , Proteína Adaptadora GRB2 , Hepatocitos , Ratones Noqueados , Daño por Reperfusión , Animales , Humanos , Masculino , Ratones , Apoptosis/genética , Modelos Animales de Enfermedad , Ferroptosis/genética , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Hepatocitos/metabolismo , Hígado/metabolismo , Hígado/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/genética
5.
Vet Microbiol ; 295: 110149, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38909417

RESUMEN

Avian reovirus (ARV) is a significant pathogen that causes various clinical diseases in chickens, including viral arthritis, chronic respiratory diseases, retarded growth, and malabsorption syndrome. These conditions result in substantial economic losses for the global poultry industry. MicroRNAs (miRNAs), a type of small noncoding RNAs that regulate gene expression post transcriptionally by silencing or degrading their RNA targets, play crucial roles in response to pathogenic infections. In this study, transfection of DF-1 cells with gga-miR-200a-3p, an upregulated miRNA observed in ARV-infected cells, significantly suppressed ARV-induced apoptosis by directly targeting GRB2 and impeded ARV replication. Conversely, knockdown of endogenous gga-miR-200a-3p in DF-1 cells using a specific miRNA inhibitor enhanced ARV-induced apoptosis and promoted GRB2 expression, thereby facilitating viral growth within cells. Consistently, inhibition of GRB2 activity through siRNA-mediated knockdown reduced viral titers. Therefore, gga-miR-200a-3p plays a vital antiviral role in the host response to ARV infection by suppressing apoptosis via direct targeting of GRB2 protein. This information enhances our understanding of the mechanisms by which host cells combat against ARV infection through self-encoded small RNA molecules and expands our knowledge regarding the involvement of microRNAs in the host response to pathogenic infections.


Asunto(s)
Apoptosis , Pollos , Proteína Adaptadora GRB2 , MicroARNs , Orthoreovirus Aviar , Replicación Viral , Animales , MicroARNs/genética , MicroARNs/metabolismo , Orthoreovirus Aviar/fisiología , Orthoreovirus Aviar/genética , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Línea Celular , Enfermedades de las Aves de Corral/virología , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/veterinaria
6.
Proc Natl Acad Sci U S A ; 121(23): e2322359121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38805286

RESUMEN

Rearranged during transfection (RET) rearrangement oncoprotein-mediated Ras/MAPK signaling cascade is constitutively activated in cancers. Here, we demonstrate a unique signal niche. The niche is a ternary complex based on the chimeric RET liquid-liquid phase separation. The complex comprises the rearranged kinase (RET fusion); the adaptor (GRB2), and the effector (SHC1). Together, they orchestrate the Ras/MAPK signal cascade, which is dependent on tyrosine kinase. CCDC6-RET fusion undergoes LLPS requiring its kinase domain and its fusion partner. The CCDC6-RET fusion LLPS promotes the autophosphorylation of RET fusion, with enhanced kinase activity, which is necessary for the formation of the signaling niche. Within the signal niche, the interactions among the constituent components are reinforced, and the signal transduction efficiency is amplified. The specific RET fusion-related signal niche elucidates the mechanism of the constitutive activation of the Ras/MAPK signaling pathway. Beyond just focusing on RET fusion itself, exploration of the ternary complex potentially unveils a promising avenue for devising therapeutic strategies aimed at treating RET fusion-driven diseases.


Asunto(s)
Proteína Adaptadora GRB2 , Sistema de Señalización de MAP Quinasas , Proteínas de Fusión Oncogénica , Proteínas Proto-Oncogénicas c-ret , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Proteínas ras , Humanos , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Células HEK293 , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Fusión Oncogénica/genética , Fosforilación , Proteínas Proto-Oncogénicas c-ret/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas ras/metabolismo , Proteínas ras/genética , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética
7.
J Biol Chem ; 300(4): 107129, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432639

RESUMEN

The adaptor protein Grb2, or growth factor receptor-bound protein 2, possesses a pivotal role in the transmission of fundamental molecular signals in the cell. Despite lacking enzymatic activity, Grb2 functions as a dynamic assembly platform, orchestrating intracellular signals through its modular structure. This study delves into the energetic communication of Grb2 domains, focusing on the folding and binding properties of the C-SH3 domain linked to its neighboring SH2 domain. Surprisingly, while the folding and stability of C-SH3 remain robust and unaffected by SH2 presence, significant differences emerge in the binding properties when considered within the tandem context compared with isolated C-SH3. Through a double mutant cycle analysis, we highlighted a subset of residues, located at the interface with the SH2 domain and far from the binding site, finely regulating the binding of a peptide mimicking a physiological ligand of the C-SH3 domain. Our results have mechanistic implications about the mechanisms of specificity of the C-SH3 domain, indicating that the presence of the SH2 domain optimizes binding to its physiological target, and emphasizing the general importance of considering supramodular multidomain protein structures to understand the functional intricacies of protein-protein interaction domains.


Asunto(s)
Proteína Adaptadora GRB2 , Unión Proteica , Pliegue de Proteína , Dominios Homologos src , Humanos , Sitios de Unión , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/química , Proteína Adaptadora GRB2/genética , Modelos Moleculares , Estructura Terciaria de Proteína
8.
Nat Commun ; 15(1): 2132, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459011

RESUMEN

Growth factor receptor-bound protein 2 (GRB2) is a cytoplasmic adapter for tyrosine kinase signaling and a nuclear adapter for homology-directed-DNA repair. Here we find nuclear GRB2 protects DNA at stalled replication forks from MRE11-mediated degradation in the BRCA2 replication fork protection axis. Mechanistically, GRB2 binds and inhibits RAD51 ATPase activity to stabilize RAD51 on stalled replication forks. In GRB2-depleted cells, PARP inhibitor (PARPi) treatment releases DNA fragments from stalled forks into the cytoplasm that activate the cGAS-STING pathway to trigger pro-inflammatory cytokine production. Moreover in a syngeneic mouse metastatic ovarian cancer model, GRB2 depletion in the context of PARPi treatment reduced tumor burden and enabled high survival consistent with immune suppression of cancer growth. Collective findings unveil GRB2 function and mechanism for fork protection in the BRCA2-RAD51-MRE11 axis and suggest GRB2 as a potential therapeutic target and an enabling predictive biomarker for patient selection for PARPi and immunotherapy combination.


Asunto(s)
Replicación del ADN , Neoplasias , Animales , Humanos , Ratones , ADN , Inestabilidad Genómica , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Inmunidad Innata , Proteína Homóloga de MRE11/metabolismo , Neoplasias/genética , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
9.
Biol Open ; 12(7)2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37421147

RESUMEN

FGF activation is known to engage canonical signals, including ERK/MAPK and PI3K/AKT, through various effectors including FRS2 and GRB2. Fgfr2FCPG/FCPG mutants that abrogate canonical intracellular signaling exhibit a range of mild phenotypes but are viable, in contrast to embryonic lethal Fgfr2-/- mutants. GRB2 has been reported to interact with FGFR2 through a non-traditional mechanism, by binding to the C-terminus of FGFR2 independently of FRS2 recruitment. To investigate whether this interaction provides functionality beyond canonical signaling, we generated mutant mice harboring a C-terminal truncation (T). We found that Fgfr2T/T mice are viable and have no distinguishable phenotype, indicating that GRB2 binding to the C-terminal end of FGFR2 is not required for development or adult homeostasis. We further introduced the T mutation on the sensitized FCPG background but found that Fgfr2FCPGT/FCPGT mutants did not exhibit significantly more severe phenotypes. We therefore conclude that, although GRB2 can bind to FGFR2 independently of FRS2, this binding does not have a critical role in development or homeostasis.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Transducción de Señal , Animales , Ratones , Desarrollo Embrionario/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética
10.
Sci Rep ; 13(1): 9784, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37328606

RESUMEN

Protein interactions with the microRNA (miRNA)-mediated gene silencing protein Argonaute 2 (AGO2) control miRNA expression. miRNA biogenesis starts with the production of precursor transcripts and culminates with the loading of mature miRNA onto AGO2 by DICER1. Here we reveal an additional component to the regulatory mechanism for miRNA biogenesis involving the adaptor protein, growth factor receptor-bound protein 2 (GRB2). The N-terminal SH3 domain of GRB2 is recruited to the PAZ domain of AGO2 forming a ternary complex containing GRB2, AGO2 and DICER1. Using small-RNA sequencing we identified two groups of miRNAs which are regulated by the binding of GRB2. First, mature and precursor transcripts of mir-17~92 and mir-221 miRNAs are enhanced. Second, mature, but not precursor, let-7 family miRNAs are diminished suggesting that GRB2 directly affects loading of these miRNAs. Notably, the resulting loss of let-7 augments expression of oncogenic targets such as RAS. Thus, a new role for GRB2 is established with implications for cancer pathogenesis through regulation of miRNA biogenesis and oncogene expression.


Asunto(s)
MicroARNs , MicroARNs/metabolismo , Silenciador del Gen , Secuencia de Bases , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo
11.
Sci Rep ; 13(1): 3505, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36864087

RESUMEN

GRB2 is an adaptor protein required for facilitating cytoplasmic signaling complexes from a wide array of binding partners. GRB2 has been reported to exist in either a monomeric or dimeric state in crystal and solution. GRB2 dimers are formed by the exchange of protein segments between domains, otherwise known as "domain-swapping". Swapping has been described between SH2 and C-terminal SH3 domains in the full-length structure of GRB2 (SH2/C-SH3 domain-swapped dimer), as well as between α-helixes in isolated GRB2 SH2 domains (SH2/SH2 domain-swapped dimer). Interestingly, SH2/SH2 domain-swapping has not been observed within the full-length protein, nor have the functional influences of this novel oligomeric conformation been explored. We herein generated a model of full-length GRB2 dimer with an SH2/SH2 domain-swapped conformation supported by in-line SEC-MALS-SAXS analyses. This conformation is consistent with the previously reported truncated GRB2 SH2/SH2 domain-swapped dimer but different from the previously reported, full-length SH2/C-terminal SH3 (C-SH3) domain-swapped dimer. Our model is also validated by several novel full-length GRB2 mutants that favor either a monomeric or a dimeric state through mutations within the SH2 domain that abrogate or promote SH2/SH2 domain-swapping. GRB2 knockdown and re-expression of selected monomeric and dimeric mutants in a T cell lymphoma cell line led to notable defects in clustering of the adaptor protein LAT and IL-2 release in response to TCR stimulation. These results mirrored similarly-impaired IL-2 release in GRB2-deficient cells. These studies show that a novel dimeric GRB2 conformation with domain-swapping between SH2 domains and monomer/dimer transitions are critical for GRB2 to facilitate early signaling complexes in human T cells.


Asunto(s)
Interleucina-2 , Dominios Homologos src , Humanos , Dimerización , Dispersión del Ángulo Pequeño , Linfocitos T , Difracción de Rayos X , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Polímeros , Proteína Adaptadora GRB2/genética
12.
Comb Chem High Throughput Screen ; 26(10): 1836-1847, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35747966

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) is associated with high morbidity and mortality. Dysregulation of lncRNAs leads to NSCLC progression. OBJECTIVE: This study aims to explore the regulatory mechanism of lncRNA LINC01234 in NSCLC. MATERIALS AND METHODS: LINC01234 expression in NSCLC cells was determined. Cell proliferation was detected using CCK-8, colony formation, and EDU assays after transfection of siRNA LINC01234 into H1299 cells and transfection of pcDNA3.1-LINC01234 into H1975 cells. Subcellular localization of LINC01234 was predicted and the binding relations between LINC01234 and miR-433-3p as well as miR-433-3p and GRB2 were verified. The expression levels of miR-433-3p and GRB2 in NSCLC cells were determined. Joint experiments of miR-433-3p inhibitor + si- LINC01234-1 or oe-GRB2 + si-LINC01234-1 were conducted to verify the role of miR-433-3p and GRB2 in NSCLC cell malignant proliferation. RESULTS: LINC01234 was abundantly expressed in NSCLC cells. LINC01234 silencing reduced NSCLC cell proliferation while LINC01234 overexpression enhanced cell proliferation. LINC01234 competitively bound to miR-433-3p and miR-433-3p directly targeted GRB2. miR- 433-3p knockdown or GRB2 overexpression counteracted the repressive effect of LINC01234 silencing on NSCLC cell malignant proliferation. CONCLUSION: LINC01234 competitively bound to miR-433-3p and promoted GRB2 transcription to augment NSCLC cell malignant proliferation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , MicroARNs/genética , MicroARNs/metabolismo , Proliferación Celular/genética , Línea Celular Tumoral , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo
13.
Melanoma Res ; 33(1): 27-37, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36545920

RESUMEN

Malignant melanoma is the most aggressive form of skin cancer, and it is characterized by poor prognosis in patients with metastatic diseases. Accurate prediction of prognosis is crucial for therapeutic decisions. In this study, bioinformatics analysis was used to explore the prognostic value of growth factor receptor-bound protein 2-associated binding protein 3 (GAB3) mRNA. RNA transcriptome sequencing data and clinical data from The Cancer Genome Atlas and genotype-tissue expression (GTEx) were analyzed for differentially expressed genes in high and low GAB3 mRNA expression groups in melanoma. Performing gene enrichment analysis and constructing protein-protein interaction networks. High expression of GAB3 was significantly correlated with a lower T stage, melanoma Clark level, Breslow depth, and melanoma ulceration. And high GAB3 expression was also associated with better progression-free interval in T1 and T2 stages and N0 stage and longer overall survival in T1 and T2 stages, N0 stage, and N1 stage. GAB3 promoted high levels of infiltration of macrophages and activated natural killer cells in melanoma. High expression of GAB3 predicted a positive prognosis in early-stage melanoma that may be mediated by the anticancer immune response.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/patología , Neoplasias Cutáneas/patología , ARN Mensajero/genética , Pronóstico , Transcriptoma , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo
14.
Commun Biol ; 5(1): 1029, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36171279

RESUMEN

Activation of RAS is crucial in driving cellular outcomes including proliferation, differentiation, migration and apoptosis via the MAPK pathway. This is initiated on recruitment of Grb2, as part of a Grb2-Sos complex, to an up-regulated receptor tyrosine kinase (RTK), enabling subsequent interaction of Sos with the plasma membrane-localised RAS. Aberrant regulation at this convergence point for RTKs in MAPK signalling is a key driver of multiple cancers. Splicing of the GRB2 gene produces a deletion variant, Grb3-3, that is incapable of binding to RTKs. We show that, despite maintaining the ability to bind to Sos, the Grb3-3-Sos complex remains in the cytoplasm, unable to engage with RAS. Competition between Grb2 and Grb3-3 for binding to C-terminal proline-rich sequences on Sos modulates MAPK signalling. Additionally, we demonstrate that splicing is regulated by heterogenous nuclear riboproteins C1/C2, and that normal and malignant colon tissue show differential Grb3-3 expression.


Asunto(s)
Apoptosis , Proteínas Tirosina Quinasas , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Mutación , Prolina , Proteínas Tirosina Quinasas/metabolismo , Transfección
15.
Eur J Obstet Gynecol Reprod Biol ; 277: 12-15, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35970002

RESUMEN

BACKGROUND: Endometriosis is a common gynecological condition with a substantial economic burden on society. It is known that both genetic and environmental factors are contributing to the phenotypic development of the disease. MicroRNAs have a vital role in the pathogenesis of endometriosis. miR-1271 and its direct target gene, GRB2 (growth factor receptor-bound protein 2), expression have been studied in gynecologic cancers, while their role in endometriosis has not been studied. OBJECTIVE: We measured miR-1271 and GRB2 gene expression in the eutopic and ectopic tissues of patients (endometrial tissues) in contrast to the control samples from healthy women. MATERIALS AND METHODS: In this study, a total of 45 samples (15 control samples, 15 eutopic samples and 15 ectopic samples) were collected. We used qRT-PCR (quantitative polymerase chain reaction) to evaluate the expression levels of the miR-1271 and GRB2 gene. RESULTS: We observed inverse expression of miR-1271 and GRB2 gene. MiR-1271 expression was significantly reduced in patients with endometriosis compared with healthy women. While there was a noticeable increase in the expression level of its target gene, GRB2, in tissues of endometriosis patients compared with normal control samples. CONCLUSION: We have shown an inverse relationship between the reduction of miR-1271 expression level and increase in the expression level of GRB2, therefore, increased GRB2 expression in endometriosis tissues can be due to decreased expression of this microRNA. Our findings suggested that miR-1271 maybe play a role as a biomarker in the diagnosis of patients with endometriosis.


Asunto(s)
Endometriosis , Proteína Adaptadora GRB2/genética , MicroARNs , Endometriosis/patología , Endometrio/patología , Femenino , Proteína Adaptadora GRB2/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Receptores de Factores de Crecimiento/metabolismo
16.
Oncotarget ; 13: 659-676, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35574218

RESUMEN

Translocation of Fibroblast Growth Factor Receptors (FGFRs) often leads to aberrant cell proliferation and cancer. The BCR-FGFR1 fusion protein, created by chromosomal translocation t(8;22)(p11;q11), contains Breakpoint Cluster Region (BCR) joined to Fibroblast Growth Factor Receptor 1 (FGFR1). BCR-FGFR1 represents a significant driver of 8p11 myeloproliferative syndrome, or stem cell leukemia/lymphoma, which progresses to acute myeloid leukemia or T-cell lymphoblastic leukemia/lymphoma. Mutations were introduced at Y177F, the binding site for adapter protein Grb2 within BCR; and at Y766F, the binding site for the membrane associated enzyme PLCγ1 within FGFR1. We examined anchorage-independent cell growth, overall cell proliferation using hematopoietic cells, and activation of downstream signaling pathways. BCR-FGFR1-induced changes in protein phosphorylation, binding partners, and signaling pathways were dissected using quantitative proteomics to interrogate the protein interactome, the phosphoproteome, and the interactome of BCR-FGFR1. The effects on BCR-FGFR1-stimulated cell proliferation were examined using the PLCγ1 inhibitor U73122, and the irreversible FGFR inhibitor futibatinib (TAS-120), both of which demonstrated efficacy. An absolute requirement is demonstrated for the dual binding partners Grb2 and PLCγ1 in BCR-FGFR1-driven cell proliferation, and new proteins such as ECSIT, USP15, GPR89, GAB1, and PTPN11 are identified as key effectors for hematopoietic transformation by BCR-FGFR1.


Asunto(s)
Linfoma , Trastornos Mieloproliferativos , Proliferación Celular , Cromosomas Humanos Par 8 , Proteína Adaptadora GRB2/genética , Humanos , Linfoma/genética , Trastornos Mieloproliferativos/genética , Proteómica , Pirazoles , Pirimidinas , Pirroles , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Translocación Genética , Proteasas Ubiquitina-Específicas/genética
17.
Int J Biochem Cell Biol ; 147: 106212, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35439649

RESUMEN

Loss of muscle mass can lead to diseases such as sarcopenia, diabetes, and obesity, which can worsen the quality of life and increase the incidence of disease. Therefore, understanding the mechanism underlying skeletal muscle differentiation is vital to prevent muscle diseases. We previously found that microRNA-320 (miR-320) is highly expressed in the lean muscle-type pigs, but its regulatory role in myogenesis remains unclear. The bioinformatics prediction indicated that miR-320 could bind to the 3 'untranslated region of growth factor receptor-bound protein-2 (Grb2). We hypothesized that miR-320 targets Grb2 to regulate myoblasts differentiation. To verify this, we transfected miR-320 mimic and inhibitor into C2C12 myoblasts to assess the role of miR-320 during myoblasts differentiation. We used real-time qPCR, luciferase reporter assays, and western blotting to confirm that miR-320 directly targets Grb2 to promote myoblasts differentiation. Moreover, by using a dexamethasone-induced atrophic model of myotubes, we discovered that miR-320 promotes the repair of damaged myotubes. Our findings expand understanding of miRNAs and genes related to regulating skeletal muscle differentiation, and provide insight into underlying therapeutic strategies for muscle diseases.


Asunto(s)
MicroARNs , Calidad de Vida , Regiones no Traducidas 3' , Animales , Atrofia/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Porcinos
18.
PLoS Negl Trop Dis ; 15(12): e0010027, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34879059

RESUMEN

BACKGROUND: The metacestode larval stage of the fox-tapeworm Echinococcus multilocularis causes alveolar echinococcosis by tumour-like growth within the liver of the intermediate host. Metacestode growth and development is stimulated by host-derived cytokines such as insulin, fibroblast growth factor, and epidermal growth factor via activation of cognate receptor tyrosine kinases expressed by the parasite. Little is known, however, concerning signal transmission to the parasite nucleus and cross-reaction with other parasite signalling systems. METHODOLOGY/PRINCIPAL FINDINGS: Using bioinformatic approaches, cloning, and yeast two-hybrid analyses we identified a novel mitogen-activated kinase (MAPK) cascade module that consists of E. multilocularis orthologs of the tyrosine kinase receptor interactor Growth factor receptor-bound 2, EmGrb2, the MAPK kinase kinase EmMEKK1, a novel MAPK kinase, EmMKK3, and a close homolog to c-Jun N-terminal kinase (JNK), EmMPK3. Whole mount in situ hybridization analyses indicated that EmMEKK1 and EmMPK3 are both expressed in E. multilocularis germinative (stem) cells but also in differentiated or differentiating cells. Treatment with the known JNK inhibitor SP600125 led to a significantly reduced formation of metacestode vesicles from stem cells and to a specific reduction of proliferating stem cells in mature metacestode vesicles. CONCLUSIONS/SIGNIFICANCE: We provide evidence for the expression of a MEKK1-JNK MAPK cascade module which, in mammals, is crucially involved in stress responses, cytoskeletal rearrangements, and apoptosis, in E. multilocularis stem cells. Inhibitor studies indicate an important role of JNK signalling in E. multilocularis stem cell survival and/or maintenance. Our data are relevant for molecular and cellular studies into crosstalk signalling mechanisms that govern Echinococcus stem cell function and introduce the JNK signalling cascade as a possible target of chemotherapeutics against echinococcosis.


Asunto(s)
Echinococcus multilocularis/enzimología , Proteínas del Helminto/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Células Madre/enzimología , Animales , Proliferación Celular , Echinococcus multilocularis/genética , Echinococcus multilocularis/crecimiento & desarrollo , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Proteínas del Helminto/genética , MAP Quinasa Quinasa 4/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , MAP Quinasa Quinasa Quinasa 3/genética , MAP Quinasa Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas , Células Madre/citología
19.
Mol Biol Cell ; 32(19): 1838-1848, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34260260

RESUMEN

p52SHC (SHC) and GRB2 are adaptor proteins involved in the RAS/MAPK (ERK) pathway mediating signals from cell-surface receptors to various cytoplasmic proteins. To further examine their roles in signal transduction, we studied the translocation of fluorescently labeled SHC and GRB2 to the cell surface, caused by the activation of ERBB receptors by heregulin (HRG). We simultaneously evaluated activated ERK translocation to the nucleus. Unexpectedly, the translocation dynamics of SHC were sustained when those of GRB2 were transient. The sustained localization of SHC positively correlated with the sustained nuclear localization of ERK, which became more transient after SHC knockdown. SHC-mediated PI3K activation was required to maintain the sustainability of the ERK translocation regulating MEK but not RAF. In cells overexpressing ERBB1, SHC translocation became transient, and the HRG-induced cell fate shifted from a differentiation to a proliferation bias. Our results indicate that SHC and GRB2 functions are not redundant but that SHC plays the critical role in the temporal regulation of ERK activation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Quinasas raf/metabolismo , Diferenciación Celular , Proliferación Celular , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células MCF-7 , Microscopía Fluorescente/métodos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Quinasas raf/genética
20.
Biochem J ; 478(14): 2793-2809, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34232285

RESUMEN

Growth factor receptor-bound protein 2 (GRB2) is a trivalent adaptor protein and a key element in signal transduction. It interacts via its flanking nSH3 and cSH3 domains with the proline-rich domain (PRD) of the RAS activator SOS1 and via its central SH2 domain with phosphorylated tyrosine residues of receptor tyrosine kinases (RTKs; e.g. HER2). The elucidation of structural organization and mechanistic insights into GRB2 interactions, however, remain challenging due to their inherent flexibility. This study represents an important advance in our mechanistic understanding of how GRB2 links RTKs to SOS1. Accordingly, it can be proposed that (1) HER2 pYP-bound SH2 potentiates GRB2 SH3 domain interactions with SOS1 (an allosteric mechanism); (2) the SH2 domain blocks cSH3, enabling nSH3 to bind SOS1 first before cSH3 follows (an avidity-based mechanism); and (3) the allosteric behavior of cSH3 to other domains appears to be unidirectional, although there is an allosteric effect between the SH2 and SH3 domains.


Asunto(s)
Proteína Adaptadora GRB2/química , Fosfotirosina/química , Dominios Proteicos , Proteína SOS1/química , Dominios Homologos src , Secuencia de Aminoácidos , Sitios de Unión/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Cinética , Ligandos , Modelos Moleculares , Fosfotirosina/metabolismo , Unión Proteica , Proteína SOS1/genética , Proteína SOS1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA