Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Med Res Rev ; 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39132876

RESUMEN

Pyroptosis, a newly characterized type of inflammatory programmed cell death (PCD), is usually triggered by multiple inflammasomes which can recognize different danger or damage-associated molecular patterns (DAMPs), leading to the activation of caspase-1 and the cleavage of gasdermin D (GSDMD). Gasdermin family pore-forming proteins are the executers of pyroptosis and are normally maintained in an inactive state through auto-inhibition. Upon caspases mediated cleavage of gasdermins, the pro-pyroptotic N-terminal fragment is released from the auto-inhibition of C-terminal fragment and oligomerizes, forming pores in the plasma membrane. This results in the secretion of interleukin (IL)-1ß, IL-18, and high-mobility group box 1 (HMGB1), generating osmotic swelling and lysis. Current therapeutic approaches including chemotherapy, radiotherapy, molecularly targeted therapy and immunotherapy for lung cancer treatment efficiently force the cancer cells to undergo pyroptosis, which then generates local and systemic antitumor immunity. Thus, pyroptosis is recognized as a new therapeutic regimen for the treatment of lung cancer. In this review, we briefly describe the signaling pathways involved in pyroptosis, and endeavor to discuss the antitumor effects of pyroptosis and its potential application in lung cancer therapy, focusing on the contribution of pyroptosis to microenvironmental reprogramming and evocation of antitumor immune response.

2.
Exp Cell Res ; 403(2): 112615, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33894221

RESUMEN

IRS4 is a member of the insulin receptor substrate (IRS) protein family. It acts as a cytoplasmic adaptor protein, integrating and transmitting signals from receptor protein tyrosine kinases to the intracellular environment. IRS4 can induce mammary tumorigenesis and is usually overexpressed in non-small cell lung cancer (NSCLC). However, little is known about the role of IRS4 in the development and progression of lung cancer. In this study, we show that IRS4 knockout suppresses the proliferation, colony formation, migration, and invasion of A549 lung cancer cells, as well as tumor growth in a nude mouse xenograft model. In contrast, stable expression of IRS4 showed the opposite effects. As expected, IRS4 was found to activate the PI3K/Akt and Ras-MAPK pathways, and we also showed that IRS4 depletion significantly enhanced the sensitivity of EGFR tyrosine kinase inhibitor (EGFR-TKI)-resistant cells to gefitinib. Taken together, these results show that IRS4 promotes NSCLC progression and may represent a potential therapeutic target for EGFR-TKI-resistant NSCLC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Gefitinib/uso terapéutico , Proteínas Sustrato del Receptor de Insulina/genética , Neoplasias Pulmonares/genética , Fosfatidilinositol 3-Quinasas/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Am J Physiol Cell Physiol ; 321(3): C429-C442, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34161152

RESUMEN

The phosphatidylinositol 3-kinase-Akt signaling pathway plays an essential role in regulating cell proliferation and apoptosis. Akt kinase is at the center of this signaling pathway and interacts with a variety of proteins. Akt is overexpressed in almost 80% of tumors. However, inhibiting Akt has serious clinical side effects so is not a suitable treatment for cancer. During recent years, Akt scaffold proteins have received increasing attention for their ability to regulate Akt signaling and have emerged as potential targets for cancer therapy. In this paper, we categorize Akt kinase scaffold proteins into four groups based on their cellular location: membrane-bound activator and inhibitor, cytoplasm, and endosome. We describe how these scaffolds interact with Akt kinase, how they affect Akt activity, and how they regulate the specificity of Akt signaling. We also discuss the clinical application of Akt scaffold proteins as targets for cancer therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
4.
Carcinogenesis ; 42(4): 587-600, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33151304

RESUMEN

c-Met hyperactivity has been observed in numerous neoplasms. Several researchers have shown that the abnormal activation of c-Met is mainly caused by transcriptional activation. However, the molecular mechanism behind this transcriptional regulation is poorly understood. Here, we suggest that Smad3 negatively regulates the expression and activation of c-Met via a transcriptional mechanism. We explore the molecular mechanisms that underlie Smad3-induced c-Met transcription inhibition. We found in contrast to the high expression of c-Met, Smad3 showed low protein and mRNA levels. Smad3 and c-Met expressions were inconsistent between lung cancer tissues and cell lines. We also found that Smad3 overexpression suppresses whereas Smad3 knockdown significantly promotes Epithelial-Mesenchymal Transition and production of the angiogenic factors VEGF, CTGF and COX-2 through the ERK1/2 pathway. In addition, Smad3 overexpression decreases whereas Smad3 knockdown significantly increases protein and mRNA levels of invasion-related ß-catenin and FAK through the PI3K/Akt pathway. Furthermore, using the chromatin immunoprecipitation analysis method, we demonstrate that a transcriptional regulatory complex consisting of HDAC1, Smad3 and mSin3A binds to the promoter of the c-Met gene. By either silencing endogenous mSin3A expression with siRNA or by pretreating cells with a specific HDAC1 inhibitor (MS-275), Smad3-induced transcriptional suppression of c-Met could be effectively attenuated. These results demonstrate that Smad3-induced inhibition of c-Met transcription depends on of a functional transcriptional regulatory complex that includes Smad3, mSin3A and HDAC1 at the c-Met promoter. Collectively, our findings reveal a new regulatory mechanism of c-Met signaling, and suggest a potential molecular target for the development of anticancer drugs.


Asunto(s)
Histona Desacetilasa 1/genética , Neoplasias Pulmonares/genética , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Proteína smad3/genética , Línea Celular Tumoral , Factor de Crecimiento del Tejido Conjuntivo/genética , Ciclooxigenasa 2/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/patología , Fosfatidilinositol 3-Quinasas/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-met/genética , Activación Transcripcional/genética , Factor A de Crecimiento Endotelial Vascular/genética , beta Catenina/genética
5.
Biosci Biotechnol Biochem ; 85(5): 1128-1139, 2021 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-33693487

RESUMEN

The C-terminal of G protein-coupled receptors is now recognized as being important for G protein activation and signaling function. To detect the role of C-terminal tail in receptor activation, we used the α1b-AR, which has a long C-terminal of 164 amino acids. We constructed the intramolecular FRET sensors, in which the C-terminal was truncated to 10 (∆C-10), 20 (∆C-20), 30 (∆C-30), 50 (∆C-50), 70 (∆C-70), or 90 (∆C-90). The truncated mutants of ∆C-10, ∆C-20, or ∆C-30 cannot induce FRET signal changes and downstream ERK1/2 phosphorylation. However, the truncated mutants of ∆C-50, ∆C-70, or ∆C-90 induce significant FRET signal changes and downstream ERK1/2 phosphorylation, especially ∆C-90. This is particularly true in the case of the ∆C-90, ∆C-70, or ∆C-50 which retained the potential phosphorylation sites (Ser401, Ser404, Ser408, or Ser410). The ∆C-90 showed an increase in agonist-induced FRET signal changes and ERK1/2 phosphorylation in PKC- or endocytosis-dependent and EGFR-, src-, or ß-arrestin2-independent.


Asunto(s)
Técnicas Biosensibles , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Procesamiento Proteico-Postraduccional , Receptores Adrenérgicos alfa 1/química , Arrestina beta 2/genética , Animales , Transferencia Resonante de Energía de Fluorescencia , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Mesocricetus , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Fenilefrina/farmacología , Fosforilación/efectos de los fármacos , Plásmidos/química , Plásmidos/metabolismo , Dominios Proteicos , Ingeniería de Proteínas/métodos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/metabolismo , Arrestina beta 2/antagonistas & inhibidores , Arrestina beta 2/metabolismo
6.
Biochem Biophys Res Commun ; 514(4): 1217-1223, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31109650

RESUMEN

The three mammalian Raf proteins (A-Raf, B-Raf, and C-Raf) are key components of the MAPK pathway. Although diverse functions have been proposed for Raf kinases, it is still not clear how interacting proteins contribute to differences in the signaling functions of the three Raf kinases. Here, we report the comparative interactomes of the three Raf kinases under serum-starved and EGF-stimulated conditions. We identified nearly 400 novel interacting proteins; some interacted with all three isoforms while others interacted exclusively with one or two. Comparing the interactomes of the three Raf kinases under different conditions revealed Raf proteins perform distinct functions through specific interactions. Our interactome data help define the differences between the three Raf kinases and may uncover new functions or regulatory mechanisms. Knowledge of Raf kinase protein-protein interactions will help us to investigate the function of specific pathways in the future.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf/análisis , Proteínas Proto-Oncogénicas c-raf/análisis , Células HEK293 , Humanos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo
7.
Biochem Biophys Res Commun ; 498(4): 932-939, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29545175

RESUMEN

Lung cancer remains the leading cause of cancer-related deaths in the world. The RAF/MEK/ERK pathway controls many fundamental cellular functions and plays key roles in lung carcinogenesis. However, the proteins that regulate this pathway remain largely unknown. Here, we identified a novel C-RAF-binding protein, RUVBL1, which activates the RAF/MEK/ERK pathway by inhibiting phosphorylation of the C-RAF protein at serine 259. RUVBL1 expression was elevated in lung adenocarcinoma tissues. In addition, knocking out RUVBL1 effectively inhibited the proliferation and invasion of A549 cells. In vivo experiments, RUVBL1 deficiency significantly decreased the tumorigensis of lung cancer. In conclusion, we have shown that RUVBL1 could activate the RAF/MEK/ERK pathway by inhibiting phosphorylation of the C-RAF protein at serine 259, to promote lung cancer progression. Therefore, RUVBL1 could represent a novel therapeutic target for lung cancer treatment.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/fisiología , Carcinogénesis/metabolismo , Proteínas Portadoras/fisiología , ADN Helicasas/fisiología , Neoplasias Pulmonares/etiología , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal/efectos de los fármacos , Células A549 , ATPasas Asociadas con Actividades Celulares Diversas/farmacología , Carcinogénesis/efectos de los fármacos , Proteínas Portadoras/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , ADN Helicasas/farmacología , Humanos , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
8.
Mol Carcinog ; 57(10): 1396-1407, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29917268

RESUMEN

Aberrant activation of the Raf-MEK-ERK pathway has frequently been associated with various cancers, especially lung cancer. However, the key regulators of this pathway are largely unknown. Using functional proteomics screening, we found that KAP1 interacts with c-Raf. Knocking out KAP1 decreased c-Raf phosphorylation at serine 259 and increased its phosphorylation at serine 338, which activated MEK and ERK. We detected higher KAP1 expression in lung cancer tissues than in normal peri-tumoral tissues. KAP1 knockdown arrested A549 lung cancer cells in the G0/G1 phase of the cell cycle and attenuated cell growth, metastasis, the epithelial-mesenchymal transition, angiogenesis, stemness, and colony formation. Furthermore, knocking out KAP1 remarkably increased the susceptibility of A549 cells to the anti-cancer drug 5-Fluorouracil, which correlated with increasing ERK phosphorylation. In vivo xenograft experiments suggested that KAP1 deficiency significantly decreases the tumorigenicity of A549 cells. Taken together, our findings indicate that KAP1 acts as a key module in the c-Raf-interactome complex and regulates lung cancer development through the Raf-MEK-ERK pathway. Therefore, KAP1 may represent a potential diagnosis biomarker and new treatment target for lung cancer.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Células A549 , Antimetabolitos Antineoplásicos/farmacología , Carcinogénesis/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fluorouracilo/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Trasplante Heterólogo , Proteína 28 que Contiene Motivos Tripartito/genética , Quinasas raf/metabolismo
9.
Biochim Biophys Acta Proteins Proteom ; 1866(8): 849-856, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29777862

RESUMEN

A-Raf is a member of the Raf kinase family. Unlike B-Raf and C-Raf, the functions of A-Raf remain obscure. To gain more insight into the biological functions of A-Raf, we investigated the A-Raf interactome using proteomics. We found 132 proteins that interact with A-Raf and confirmed the interaction of 12 of these proteins with A-Raf by western blotting. Our data suggested that A-Raf regulates apoptosis, RNA catabolism, GTPase activity, and cell adhesion by interacting with proteins located in different cellular compartments. We identified all ten hallmarks of cancer in these interacting proteins, suggesting that A-Raf is involved in carcinogenesis. Our results also indicated that A-Raf may play a role in different diseases and signaling pathways. These findings have identified potential regulators of A-Raf and provide a systemic insight into its biological functions.


Asunto(s)
Proteómica , Proteínas Proto-Oncogénicas A-raf/metabolismo , Apoptosis , Western Blotting , Carcinogénesis/genética , Adhesión Celular , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas A-raf/genética , ARN/metabolismo , Transducción de Señal
10.
Cell Biol Int ; 42(7): 756-768, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29418027

RESUMEN

The c-Jun N-terminal kinases (JNKs) are located downstream of Ras-mitogen activated protein kinase signaling cascades. More than 20 years of study has shown that JNKs control cell fate and many cellular functions. JNKs and their interacting proteins form a complicated network with diverse biological functions and physiological effects. Members of the JNK interactome include Jun, amyloid precursor protein, and insulin receptor substrate. Recent studies have shown that the JNK interactome is involved in tumorigenesis, neuron development, and insulin resistance. In this review, we summarize the features of the JNK interactome and classify its members into three groups: upstream regulators, downstream effectors, and scaffold partners. We also highlight the unique cellular signaling mechanisms of JNKs and provide more insights into the roles of the JNK interactome in human diseases.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Transducción de Señal/fisiología , Proteínas ras/metabolismo , Animales , Salud , Humanos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Sheng Li Xue Bao ; 70(5): 565-570, 2018 Oct 25.
Artículo en Zh | MEDLINE | ID: mdl-30377696

RESUMEN

Son of sevenless homolog 1 (SOS1) protein is a ubiquitously expressed adapter. As a key protein in intracellular signaling, SOS1 plays an important role in many signal transduction pathways, such as Ras and Rac signaling pathways. The abnormal expression or mutation of SOS1 is closely related to clinical diseases. In this article, we review research progress on SOS1 functions and its roles in physiology and pathophysiology.


Asunto(s)
Proteína SOS1/fisiología , Transducción de Señal , Animales , Humanos , Mutación
12.
Crit Rev Biochem Mol Biol ; 50(6): 520-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26508523

RESUMEN

The Ras-Raf-MEK-MAPK (mitogen-activated protein kinase)-signaling pathway plays a key role in the regulation of many cellular functions, including cell proliferation, differentiation and transformation, by transmitting signals from membrane receptors to various cytoplasmic and nuclear targets. One of the key components of this pathway is the serine/threonine protein kinase, Raf. The Raf family kinases (A-Raf, B-Raf and C-Raf) have been intensively studied since being identified in the early 1980s as retroviral oncogenes, especially with respect to the discovery of activating mutations of B-Raf in a large number of tumors which led to intensified efforts to develop drugs targeting Raf kinases. This also resulted in a rapid increase in our knowledge of the biological functions of the B-Raf and C-Raf isoforms, which may in turn be contrasted with the little that is known about A-Raf. The biological functions of A-Raf remain mysterious, although it appears to share some of the basic properties of the other two isoforms. Recently, emerging evidence has begun to reveal the functions of A-Raf, of which some are kinase-independent. These include the inhibition of apoptosis by binding to MST2, acting as safeguard against oncogenic transformation by suppressing extracellular signal-regulated kinases (ERK) activation and playing a role in resistance to Raf inhibitors. In this review, we discuss the regulation of A-Raf protein expression, and the roles of A-Raf in apoptosis and cancer, with a special focus on its role in resistance to Raf inhibitors. We also describe the scaffold functions of A-Raf and summarize the unexpected complexity of Raf signaling.


Asunto(s)
Neoplasias/genética , Proteínas Proto-Oncogénicas A-raf/genética , Proteínas Proto-Oncogénicas A-raf/metabolismo , Animales , Resistencia a Antineoplásicos , Endocitosis , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Mapas de Interacción de Proteínas , Isoformas de Proteínas/análisis , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas A-raf/análisis , Proteínas Proto-Oncogénicas A-raf/antagonistas & inhibidores , Transducción de Señal
13.
Cell Physiol Biochem ; 39(1): 137-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27322838

RESUMEN

Rap, a member of the Ras-like small G-protein family, is a key node among G-protein coupled receptors (GPCR), receptor tyrosine kinases (RTKs), ion channels and many other downstream pathways. Rap plays a unique role in cell morphogenesis, adhesion, migration, exocytosis, proliferation, apoptosis and carcinogenesis. The complexity and diversity of Rap functions are tightly regulated by Rap-interacting proteins such as GEFs, GAPs, Rap effectors and scaffold proteins. These interacting proteins decide the subcellular localization of Rap, the interaction modes with downstream Rap effectors and tune Rap as an atypical molecular conductor, coupling extra- and intracellular signals to various pathways. In this review, we summarize four groups of Rap-interacting proteins, highlight their distinctions in Rap-binding properties and interactive modes and discuss their contribution to the spatiotemporal regulation of Rap as well as the implications of targeting Rap-interacting proteins in human cancer therapy.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap/metabolismo , Humanos , Modelos Biológicos , Unión Proteica , Mapas de Interacción de Proteínas , Isoformas de Proteínas/metabolismo
14.
Biochem J ; 470(2): 155-67, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26348905

RESUMEN

GPCRs (G-protein-coupled receptors) are members of a family of proteins which are generally regarded as the largest group of therapeutic drug targets. Ligands of GPCRs do not usually activate all cellular signalling pathways linked to a particular seven-transmembrane receptor in a uniform manner. The fundamental idea behind this concept is that each ligand has its own ability, while interacting with the receptor, to activate different signalling pathways (or a particular set of signalling pathways) and it is this concept which is known as biased signalling. The importance of biased signalling is that it may selectively activate biological responses to favour therapeutically beneficial signalling pathways and to avoid adverse effects. There are two levels of biased signalling. First, bias can arise from the ability of GPCRs to couple to a subset of the available G-protein subtypes: Gαs, Gαq/11, Gαi/o or Gα12/13. These subtypes produce the diverse effects of GPCRs by targeting different effectors. Secondly, biased GPCRs may differentially activate G-proteins or ß-arrestins. ß-Arrestins are ubiquitously expressed and function to terminate or inhibit classic G-protein signalling and initiate distinct ß-arrestin-mediated signalling processes. The interplay of G-protein and ß-arrestin signalling largely determines the cellular consequences of the administration of GPCR-targeted drugs. In the present review, we highlight the particular functionalities of biased signalling and discuss its biological effects subsequent to GPCR activation. We consider that biased signalling is potentially allowing a choice between signalling through 'beneficial' pathways and the avoidance of 'harmful' ones.


Asunto(s)
Receptores Odorantes/metabolismo , Transducción de Señal , Animales , Arrestinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Ligandos , Conformación Proteica , Receptores Odorantes/química , beta-Arrestinas
15.
Sheng Li Xue Bao ; 68(1): 57-64, 2016 Feb 25.
Artículo en Zh | MEDLINE | ID: mdl-26915323

RESUMEN

Gab proteins, Grb2 (growth factor receptor binding protein 2)-associated binder, are important scaffolding adapter proteins required by many signaling pathways. In mammals, the Gab proteins mainly consist of Gab1, Gab2 and Gab3, and are involved in the amplification and integration of signal transduction evoked by a variety of extracellular stimuli, including various growth factors and cytokines. They are known to play key roles in many biological processes through the two classical signal pathways, SHP2/RAS/ERK and PI3K/AKT. In this review, we provide an overview of the structure and function of the scaffolding adapter, Gab, with a special focus on its role in tumor, inflammation and cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Inflamación , Neoplasias , Proteínas Adaptadoras Transductoras de Señales , Humanos , Péptidos y Proteínas de Señalización Intercelular , Fosfatidilinositol 3-Quinasas , Fosforilación , Transducción de Señal
16.
Sheng Li Ke Xue Jin Zhan ; 47(1): 14-20, 2016 Feb.
Artículo en Zh | MEDLINE | ID: mdl-27424400

RESUMEN

Rap has different biological functions on intracellular signaling pathways, such as regulating cell polarity, cell proliferation, cell differentiation, cell adhesion and cell movement. Furthermore, at tissue and organ level, Rap controls the establishment of neural polarity, synaptic growth, synaptic plasticity, neuronal migration and so on. Rap belongs to Ras family which contains two subtypes, Rap1 and Rap2. By binding GTP or GDP Rap transform between active or inactive state, and plays an important role as a molecular switch. Moreover, in the signal pathway of tumor, Rap inhibits cell transformation induced by the oncogene Ras, therefore inhibits the proliferation, invasion and migration of certain cancer cells by interacting with its downstream target molecules. In this review, we summarized the biological functions of Rap and discussed It's significance in cancer therapy and drug treatment of neurological diseases.


Asunto(s)
Proteínas de Unión al GTP rap/fisiología , Adhesión Celular , Movimiento Celular , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Plasticidad Neuronal , Transducción de Señal
17.
Acta Biochim Biophys Sin (Shanghai) ; 47(7): 477-87, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26040315

RESUMEN

Tobacco usage is a major risk factor in the development, progression, and outcomes for lung cancer. Of the carcinogens associated with lung cancer, tobacco-specific nitrosamines 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is among the most potent ones. The oncogenic mechanisms of NNK are not entirely understood, hindering the development of effective strategies for preventing and treating smoking-associated lung cancers. Here, we introduce the NNK-induced lung cancer animal models in different species and its potential mechanisms. Finally, we summarize several chemopreventive agents developed from these animal models.


Asunto(s)
Carcinogénesis , Carcinógenos/toxicidad , Neoplasias Pulmonares/inducido químicamente , Modelos Animales , Nitrosaminas/toxicidad , Animales , Neoplasias Pulmonares/patología
18.
Biochem J ; 452(2): 303-12, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23521066

RESUMEN

The literature on GPCR (G-protein-coupled receptor) homo-oligomerization encompasses conflicting views that range from interpretations that GPCRs must be monomeric, through comparatively newer proposals that they exist as dimers or higher-order oligomers, to suggestions that such quaternary structures are rather ephemeral or merely accidental and may serve no functional purpose. In the present study we use a novel method of FRET (Förster resonance energy transfer) spectrometry and controlled expression of energy donor-tagged species to show that M(3)Rs (muscarinic M(3) acetylcholine receptors) at the plasma membrane exist as stable dimeric complexes, a large fraction of which interact dynamically to form tetramers without the presence of trimers, pentamers, hexamers etc. That M(3)R dimeric units interact dynamically was also supported by co-immunoprecipitation of receptors synthesized at distinct times. On the basis of all these findings, we propose a conceptual framework that may reconcile the conflicting views on the quaternary structure of GPCRs.


Asunto(s)
Membrana Celular/química , Receptor Muscarínico M3/química , Membrana Celular/genética , Membrana Celular/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Células HEK293 , Humanos , Modelos Moleculares , Multimerización de Proteína/genética , Estabilidad Proteica , Estructura Cuaternaria de Proteína/genética , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
J Ethnopharmacol ; 327: 117982, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38423411

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Cynanchum otophyllum C.K.Schneid.PI.Wilson, commonly referred as ''Qingyangshen'' (QYS), is a traditional folk medicine from Yunnan, renowned for its efficacy in neurological and psychiatric disorders. Glycosides isolated from QYS have shown promise in alleviating epilepsy, however, mechanisms of action and specific molecular targets remain to be elucidated. AIM OF THE STUDY: The study aimed to evaluate the anticonvulsant effects of Qingyangshen glycosides M1 (M1), a C21 steroidal glycoside from QYS, on pentylenetetrazol (PTZ)-induced convulsions in zebrafish (Danio rerio), and its neuroprotective effect on Glutamate (Glu)-induced damage to PC12 cells, and importantly to identify its potential molecular targets. MATERIALS AND METHODS: To evaluate anticonvulsant activity of M1, 7 days-post-fertilization (7-dpf) animals were pretreated (by immersion) and then exposed to PTZ (10 mM) solution. Furthermore, Glu-induced PC12 cell damage was employed to investigate the neuroprotective and anti-apoptotic capacity. Cells were pretreated with various concentrations of M1 (0-10 µM) for 12 h and then co-treated with Glu (15 mM) for an additional 24 h. The cell viability, apoptosis rate and apoptosis-related proteins (p-PI3K, PI3K, Akt, p-Akt, CREB, p-CREB, BDNF, Bax and Bcl-2) were measured using CCK-8, annexin V/PI and Western blot assays. To model the expected interaction between M1 and candidate cannabinoid receptor type 1 (CB1R), ERK phosphorylation, molecular docking, and drug affinity responsive target stability (DARTS) techniques were employed. Finally, CB1R antagonist Rimonabant (Rim) was validated by co-administration in both zebrafish and cells to confirm the requirement of CB1R for M1 efficacy. RESULTS: At a concentration of 400 µM, M1 dramatically reversed PTZ-induced convulsive-like behaviors in zebrafish, as evidenced by a significant reduction in locomotor activity. In the context of Glu-induced cytotoxicity, M1 (10 µM) demonstrated a notable increase in cell viability and suppressed apoptosis through modulation of the Bax/Bcl-2 ratio and activation of the PI3K/Akt/CREB/BDNF signaling axis. These effects were facilitated through CB1R activation. In contrast, Rim dampened the beneficial activities of M1 as a cannabinoid agonist. CONCLUSIONS: These results demonstrated that M1 as a potential CB1R activator, exhibiting anticonvulsive effects in a PTZ-induced zebrafish model and neuroprotective properties via the PI3K/Akt/CREB/BDNF signaling axis in a Glu-induced PC12 cell injury model. Notably, the observed seizure relief attenuated by CB1R chemical antagonism.


Asunto(s)
Fármacos Neuroprotectores , Proteínas Proto-Oncogénicas c-akt , Humanos , Ratas , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glicósidos/farmacología , Glicósidos/uso terapéutico , Glicósidos/química , Pez Cebra , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína X Asociada a bcl-2 , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Simulación del Acoplamiento Molecular , China , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Proteínas Reguladoras de la Apoptosis , Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2 , Pentilenotetrazol/toxicidad , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
20.
Biochem Pharmacol ; 223: 116194, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38583812

RESUMEN

Glutathione peroxidase 4 (GPX4) is a promising anticancer therapeutic target; however, the application of GPX4 inhibitors (GPX4i) is limited owing to intrinsic or acquired drug resistance. Hence, understanding the mechanisms underlying drug resistance and discovering molecules that can overcome drug resistance are crucial. Herein, we demonstrated that GPX4i killed bladder cancer cells by inducing lipid reactive oxygen species-mediated ferroptosis and apoptosis, and cisplatin-resistant bladder cancer cells were also resistant to GPX4i, representing a higher half-maximal inhibitory concentration value than that of parent bladder cancer cells. In addition, thioredoxin reductase 1 (TrxR1) overexpression was responsible for GPX4i resistance in cisplatin-resistant bladder cancer cells, and inhibiting TrxR1 restored the sensitivity of these cells to GPX4i. In vitro and in vivo studies revealed that Jolkinolide B (JB), a natural diterpenoid and previously identified as a TrxR1 inhibitor, potentiated the antiproliferative efficacy of GPX4i (RSL3 and ML162) against cisplatin-resistant bladder cancer cells. Furthermore, GPX4 knockdown and inhibition could augment JB-induced paraptosis and apoptosis. Our results suggest that inhibiting TrxR1 can effectively improve GPX4 inhibition-based anticancer therapy. A combination of JB and GPX4i, which is well-tolerated and has several anticancer mechanisms, may serve as a promising therapy for treating bladder cancer.


Asunto(s)
Compuestos de Anilina , Diterpenos , Tiofenos , Neoplasias de la Vejiga Urinaria , Humanos , Cisplatino/farmacología , Tiorredoxina Reductasa 1 , Línea Celular Tumoral , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda