Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Leukoc Biol ; 116(4): 779-792, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38652699

RESUMEN

The mammalian target of rapamycin (mTOR) pathway plays a key role in determining immune cells function through modulation of their metabolic status. By specific deletion of Rictor in CD11c+ myeloid cells (referred to here as CD11cRicΔ/Δ), we investigated the role of mTOR complex 2 (mTORC2) signaling in dendritic cells (DCs) function in mice. We showed that upon dextran sulfate sodium-induced colitis, the lack of mTORC2 signaling CD11c+ cells diminishes the colitis score and abrogates DC migration to the mesenteric lymph nodes, thereby diminishing the infiltration of T helper 17 cells in the lamina propria and subsequent inflammation. These findings corroborate with the abrogation of cytoskeleton organization and the decreased activation of Rac1 and Cdc42 GTPases observed in CD11c+-mTORC2-deficient cells. Meta-analysis on colonic samples from ulcerative colitis patients revealed increased gene expression of proinflammatory cytokines, which coincided with augmented expression of the mTOR pathway, a positive correlation between the DC marker ITGAX and interleukin-6, the expression of RICTOR, and CDC42. Together, this work proposes that targeting mTORC2 on DCs offers a key to hamper inflammatory responses, and this way, ameliorates the progression and severity of intestinal inflammatory diseases.


Asunto(s)
Movimiento Celular , Colitis , Células Dendríticas , Sulfato de Dextran , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Mieloides , Transducción de Señal , Animales , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Colitis/patología , Colitis/inducido químicamente , Colitis/inmunología , Células Mieloides/metabolismo , Células Mieloides/inmunología , Sulfato de Dextran/toxicidad , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Antígeno CD11c/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Humanos , Proteína de Unión al GTP rac1/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Ratones Noqueados , Neuropéptidos , Antígenos CD11
2.
Clin Transl Oncol ; 26(8): 2020-2024, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38478261

RESUMEN

OBJECTIVE: The primary goal of this study was to investigate the expressions of TUFT1 (Tuftelin) and Rac1-GTP in the cancerous tissues of individuals with triple-negative breast cancer (TNBC). Additionally, we aimed to explore the correlation between TUFT1 and Rac1-GTP expressions and examine the associations of TUFT1 and Rac1-GTP expressions with the clinical and pathological indicators of the patients. METHODS: Ninety-six patients diagnosed with TNBC, scheduled for surgery between May 2022 and November 2022, were enrolled in this study. Cancerous tissue specimens were collected from these patients, and immunohistochemistry was employed to evaluate the levels of TUFT1 and Rac1-GTP expressions in the cancerous tissues. Subsequent to data collection, a comprehensive analysis was conducted to examine the correlation between TUFT1 and Rac1-GTP expressions. Furthermore, we sought to assess the associations of TUFT1 and Rac1-GTP expressions with the clinical and pathological indicators of the patients. RESULTS: The TUFT1 protein was expressed in both the membrane and cytoplasm of TNBC cancer cells, with notably higher expression observed in the cytoplasm. Rac1-GTP was primarily expressed in the cytoplasm. There was a positive correlation between the levels of TUFT1 and Rac1-GTP expressions (χ2 = 9.816, P < 0.05). The levels of TUFT1 and Rac1-GTP protein expressions showed no correlation with patient age (χ2 = 2.590, 2.565, P > 0.05); however, they demonstrated a positive correlation with tumor size (χ2 = 5.592,5.118), histological grading (χ2 = 6.730, 5.443), and lymph node metastasis (χ2 = 8.221, 5.180) (all with a significance level of P < 0.05). CONCLUSION: A significant correlation was identified between the levels of TUFT1 and Rac1-GTP expressions in the cancerous tissues of patients with TNBC, suggesting a close association with the progression of TNBC. The two molecules play significant roles in facilitating an early diagnosis and treatment of TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Proteína de Unión al GTP rac1 , Humanos , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Femenino , Persona de Mediana Edad , Adulto , Anciano , Metástasis Linfática , Biomarcadores de Tumor/metabolismo , Inmunohistoquímica , Citoplasma/metabolismo
3.
Biol Res ; 56(1): 26, 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37211600

RESUMEN

Previous studies have shown that peripheral nerve injury can lead to abnormal dendritic spine remodeling in spinal dorsal horn neurons. Inhibition of abnormal dendritic spine remodeling can relieve neuropathic pain. Electroacupuncture (EA) has a beneficial effect on the treatment of neuropathic pain, but the specific mechanism remains unclear. Evidence has shown that slit-robo GTPase activating protein 3 (srGAP3) and Rho GTPase (Rac1) play very important roles in dendritic spine remodeling. Here, we used srGAP3 siRNA and Rac1 activator CN04 to confirm the relationship between SrGAP3 and Rac1 and their roles in improving neuropathic pain with EA. Spinal nerve ligation (SNL) was used as the experimental model, and thermal withdrawal latency (TWL), mechanical withdrawal threshold (MWT), Western blotting, immunohistochemistry and Golgi-Cox staining were used to examine changes in behavioral performance, protein expression and dendritic spines. More dendritic spines and higher expression levels of srGAP3 were found in the initial phase of neuropathic pain. During the maintenance phase, dendritic spines were more mature, which was consistent with lower expression levels of srGAP3 and higher expression levels of Rac1-GTP. EA during the maintenance phase reduced the density and maturity of dendritic spines of rats with SNL, increased the levels of srGAP3 and reduced the levels of Rac1-GTP, while srGAP3 siRNA and CN04 reversed the therapeutic effects of EA. These results suggest that dendritic spines have different manifestations in different stages of neuropathic pain and that EA may inhibit the abnormal dendritic spine remodeling by regulating the srGAP3/Rac1 signaling pathway to alleviate neuropathic pain.


Asunto(s)
Electroacupuntura , Neuralgia , Animales , Ratas , Espinas Dendríticas/metabolismo , GTP Fosfohidrolasas/metabolismo , Guanosina Trifosfato/metabolismo , Neuralgia/metabolismo , Neuralgia/terapia , Proteína de Unión al GTP rac1/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Nervios Espinales/metabolismo
4.
Biol. Res ; 56: 26-26, 2023. ilus, graf
Artículo en Inglés | LILACS | ID: biblio-1513738

RESUMEN

Previous studies have shown that peripheral nerve injury can lead to abnormal dendritic spine remodeling in spinal dorsal horn neurons. Inhibition of abnormal dendritic spine remodeling can relieve neuropathic pain. Electroacupuncture (EA) has a beneficial effect on the treatment of neuropathic pain, but the specific mechanism remains unclear. Evidence has shown that slit-robo GTPase activating protein 3 (srGAP3) and Rho GTPase (Rac1) play very important roles in dendritic spine remodeling. Here, we used srGAP3 siRNA and Rac1 activator CN04 to confirm the relationship between SrGAP3 and Rac1 and their roles in improving neuropathic pain with EA. Spinal nerve ligation (SNL) was used as the experimental model, and thermal withdrawal latency (TWL), mechanical withdrawal threshold (MWT), Western blotting, immunohistochemistry and Golgi-Cox staining were used to examine changes in behavioral performance, protein expression and dendritic spines. More dendritic spines and higher expression levels of srGAP3 were found in the initial phase of neuropathic pain. During the maintenance phase, dendritic spines were more mature, which was consistent with lower expression levels of srGAP3 and higher expression levels of Rac1-GTP. EA during the maintenance phase reduced the density and maturity of dendritic spines of rats with SNL, increased the levels of srGAP3 and reduced the levels of Rac1-GTP, while srGAP3 siRNA and CN04 reversed the therapeutic effects of EA. These results suggest that dendritic spines have different manifestations in different stages of neuropathic pain and that EA may inhibit the abnormal dendritic spine remodeling by regulating the srGAP3/Rac1 signaling pathway to alleviate neuropathic pain.


Asunto(s)
Animales , Ratas , Electroacupuntura , Neuralgia/metabolismo , Neuralgia/terapia , Nervios Espinales/metabolismo , Transducción de Señal , Ratas Sprague-Dawley , Proteína de Unión al GTP rac1/metabolismo , Espinas Dendríticas/metabolismo , GTP Fosfohidrolasas/metabolismo , Guanosina Trifosfato/metabolismo
5.
Sci Rep ; 11(1): 20946, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686741

RESUMEN

Cell spreading and phagocytosis are notably regulated by small GTPases and GAP proteins. TBC1D10C is a dual inhibitory protein with GAP activity. In immune cells, TBC1D10C is one of the elements regulating lymphocyte activation. However, its specific role in macrophages remains unknown. Here, we show that TBC1D10C engages in functions dependent on the cytoskeleton and plasma membrane reorganization. Using ex vivo and in vitro assays, we found that elimination and overexpression of TBC1D10C modified the cytoskeletal architecture of macrophages by decreasing and increasing the spreading ability of these cells, respectively. In addition, TBC1D10C overexpression contributed to higher phagocytic activity against Burkholderia cenocepacia and to increased cell membrane tension. Furthermore, by performing in vitro and in silico analyses, we identified 27 TBC1D10C-interacting proteins, some of which were functionally classified as protein complexes involved in cytoskeletal dynamics. Interestingly, we identified one unreported TBC1D10C-intrinsically disordered region (IDR) with biological potential at the cytoskeleton level. Our results demonstrate that TBC1D10C shapes macrophage activity by inducing reorganization of the cytoskeleton-plasma membrane in cell spreading and phagocytosis. We anticipate our results will be the basis for further studies focused on TBC1D10C. For example, the specific molecular mechanism in Burkholderia cenocepacia phagocytosis and functional analysis of TBC1D10C-IDR are needed to further understand its role in health and disease.


Asunto(s)
Citoesqueleto/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiología , Fagocitosis/fisiología , Animales , Burkholderia cenocepacia/patogenicidad , Membrana Celular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de Unión al GTP rac1/metabolismo
6.
Cells ; 10(8)2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34440613

RESUMEN

Sepsis and septic shock are associated with acute and sustained impairment in the function of the cardiovascular system, kidneys, lungs, liver, and brain, among others. Despite the significant advances in prevention and treatment, sepsis and septic shock sepsis remain global health problems with elevated mortality rates. Rho proteins can interact with a considerable number of targets, directly affecting cellular contractility, actin filament assembly and growing, cell motility and migration, cytoskeleton rearrangement, and actin polymerization, physiological functions that are intensively impaired during inflammatory conditions, such as the one that occurs in sepsis. In the last few decades, Rho proteins and their downstream pathways have been investigated in sepsis-associated experimental models. The most frequently used experimental design included the exposure to bacterial lipopolysaccharide (LPS), in both in vitro and in vivo approaches, but experiments using the cecal ligation and puncture (CLP) model of sepsis have also been performed. The findings described in this review indicate that Rho proteins, mainly RhoA and Rac1, are associated with the development of crucial sepsis-associated dysfunction in different systems and cells, including the endothelium, vessels, and heart. Notably, the data found in the literature suggest that either the inhibition or activation of Rho proteins and associated pathways might be desirable in sepsis and septic shock, accordingly with the cellular system evaluated. This review included the main findings, relevance, and limitations of the current knowledge connecting Rho proteins and sepsis-associated experimental models.


Asunto(s)
Sepsis/enzimología , Choque Séptico/enzimología , Proteínas de Unión al GTP rho/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Terapia Molecular Dirigida , Sepsis/tratamiento farmacológico , Sepsis/patología , Choque Séptico/tratamiento farmacológico , Choque Séptico/patología , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
7.
Sci Rep ; 11(1): 14984, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294745

RESUMEN

Fatty acids (FA) have a multitude of biological actions on living cells. A target of their action is cell motility, a process of critical importance during cancer cell dissemination. Here, we studied the effect of unsaturated FA on ovarian cancer cell migration in vitro and its role in regulating cytoskeleton structures that are essential for cell motility. Scratch wound assays on human ovary cancer SKOV-3 cell monolayers revealed that low doses (16 µM) of linoleic acid (LA, 18:2 ω6) and oleic acid (OA; 18:1 ω9) promoted migration, while α-linolenic acid (ALA, 18:3 ω3), showed a migration rate similar to that of the control group. Single cell tracking demonstrated that LA and OA-treated cells migrated faster and were more orientated towards the wound closure than control. In vitro addition of those FA resulted in an increased number, length and protrusion speed of filopodia and also in a prominent and dynamic lamellipodia at the cell leading edge. Using time-lapse video-microscopy and FRAP we observed an increase in both the speed and frequency of actin waves associated with more mobile actin and augmented Rac1 activity. We also observed that FA induced microtubule-organizing center (MTOC)-orientation towards the cell front and affected the dynamics of microtubules (MT) in the direction of cell migration. We propose that environmental cues such as OA and LA present in ascitic fluid, should be taken into account as key factors for the regulation of cell migration.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Ácido Linoleico/farmacología , Microtúbulos/efectos de los fármacos , Ácido Oléico/farmacología , Neoplasias Ováricas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Líquido Ascítico/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Microtúbulos/metabolismo , Análisis de la Célula Individual , Imagen de Lapso de Tiempo , Regulación hacia Arriba
8.
Microbes Infect ; 23(8): 104837, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33957277

RESUMEN

Cell invasion by Trypanosoma cruzi extracellular amastigotes (EAs) relies significantly upon the host cell actin cytoskeleton. In past decades EAs have been established as a reliable model for phagocytosis inducer in non-phagocytic cells. Our current hypothesis is that EAs engage a phagocytosis-like mechanism in non-professional phagocytic cells; however, the molecular mechanisms in professional phagocytes still remain unexplored. In this work, we evaluated the involvement of Rac1 and Cdc42 in the actin-dependent internalization of EAs in RAW 264.7 macrophages. Kinetic assays showed similar internalization of EAs in unstimulated RAW and non-phagocytic HeLa cells but increased in LPS/IFN-γ stimulated RAW cells. However, depletion of Rac1, Cdc42 or RhoA inhibited EA internalization similarly in both unstimulated and stimulated RAW cells. Overexpression of active, but not the dominant-negative, construct of Rac1 increased EA internalization. Remarkably, for Cdc42, both the active and the inactive mutants decreased EA internalization when compared to wild type groups. Despite that, both Rac1 and Cdc42 activation mutants were similarly recruited to and colocalized with actin at the EA-macrophage contact sites when compared to their native isoforms. Altogether, these results corroborate that EAs engage phagocytic processes to invade both professional and non-professional phagocytic cells providing evidences of converging actin mediated mechanisms induced by intracellular pathogens in both cell types.


Asunto(s)
Trypanosoma cruzi , Actinas/metabolismo , Células HeLa , Humanos , Macrófagos/metabolismo , Fagocitosis/fisiología , Trypanosoma cruzi/fisiología , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
9.
Biochem J ; 478(2): 407-422, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33393983

RESUMEN

Insulin stimulates glucose uptake in muscle cells by rapidly redistributing vesicles containing GLUT4 glucose transporters from intracellular compartments to the plasma membrane (PM). GLUT4 vesicle fusion requires the formation of SNARE complexes between vesicular VAMP and PM syntaxin4 and SNAP23. SNARE accessory proteins usually regulate vesicle fusion processes. Complexins aide in neuro-secretory vesicle-membrane fusion by stabilizing trans-SNARE complexes but their participation in GLUT4 vesicle fusion is unknown. We report that complexin-2 is expressed and homogeneously distributed in L6 rat skeletal muscle cells. Upon insulin stimulation, a cohort of complexin-2 redistributes to the PM. Complexin-2 knockdown markedly inhibited GLUT4 translocation without affecting proximal insulin signalling of Akt/PKB phosphorylation and actin fiber remodelling. Similarly, complexin-2 overexpression decreased maximal GLUT4 translocation suggesting that the concentration of complexin-2 is finely tuned to vesicle fusion. These findings reveal an insulin-dependent regulation of GLUT4 insertion into the PM involving complexin-2.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/farmacología , Mioblastos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Transportador de Glucosa de Tipo 4/genética , Insulina/genética , Insulina/metabolismo , Músculo Esquelético/citología , Mioblastos/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
10.
Methods Mol Biol ; 2174: 31-43, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32813243

RESUMEN

Molecular docking is a useful and powerful computational method for the identification of potential interactions between small molecules and pharmacological targets. In reverse docking, the ability of one or a few compounds to bind a large dataset of proteins is evaluated in silico. This strategy is useful for identifying molecular targets of orphan bioactive compounds, proposing new molecular mechanisms, finding alternative indications of drugs, or predicting drug toxicity. Herein, we describe a detailed reverse docking protocol for the identification of potential targets for 4-hydroxycoumarin (4-HC). Our results showed that RAC1 is a target of 4-HC, which partially explains the biological activities of 4-HC on cancer cells. The strategy reported here can be easily applied to other compounds and protein datasets.


Asunto(s)
4-Hidroxicumarinas/farmacología , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Simulación del Acoplamiento Molecular/métodos , 4-Hidroxicumarinas/química , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Simulación por Computador , Bases de Datos de Proteínas , Humanos , Ligandos , Terapia Molecular Dirigida , Conformación Proteica , Programas Informáticos , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/metabolismo
11.
ChemMedChem ; 16(6): 1011-1021, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33284505

RESUMEN

The Rho GTPase Rac1 is involved in the control of cytoskeleton reorganization and other fundamental cellular functions. Aberrant activity of Rac1 and its regulators is common in human cancer. In particular, deregulated expression/activity of Rac GEFs, responsible for Rac1 activation, has been associated to a metastatic phenotype and drug resistance. Thus, the development of novel Rac1-GEF interaction inhibitors is a promising strategy for finding new preclinical candidates. Here, we studied structure-activity relationships within a new family of N,N'-disubstituted guanidine as Rac1 inhibitors. We found that compound 1D-142, presents superior antiproliferative activity in human cancer cell lines and higher potency as Rac1-GEF interaction inhibitor in vitro than parental compounds. In addition, 1D-142 reduces Rac1-mediated TNFα-induced NF-κB nuclear translocation during cell proliferation and migration in NSCLC. Notably, 1D-142 allowed us to show for the first time the application of a Rac1 inhibitor in a lung cancer animal model.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Desarrollo de Medicamentos , Guanidina/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Guanidina/síntesis química , Guanidina/química , Humanos , Hidroxilación , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad , Proteína de Unión al GTP rac1/metabolismo
12.
Oral Oncol ; 112: 105038, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33129055

RESUMEN

OBJECTIVES: To analyze the inflammatory millieu in oral squamous cell carcinoma (OSCC) tumors and the influence of macrophages related-cytokines on the tumor cell migration. MATERIALS AND METHODS: Inflammatory protein profile and macrophage population (M2/M1 ratio) of human OSCC fragments were analyzed by proteomic analysis and flow cytometry assay respectively. To evaluate the effects of inflammation on OSCC behavior, we analyzed the role of polarized macrophages and cytokines (IL-6, IL-1ß and TNF-α) on OSCC cell lines (SCC25 and Cal27) responsiveness by western blotting (cell signaling) and time-lapse (cell migration). Also, it was addressed the crosstalk of IL-6-STAT3 axis with cell migration signaling using a STAT3 inhibitor (Stattic®) and a pull down assay for the RhoGTPase Rac1 activity. RESULTS: It was observed a ~2 fold predominance of M2 over M1 macrophages and a pro-inflammatory state in OSCC fragments. The M2 conditioned media increased migration speed and directionality of highly invasive OSCC cells (SCC25). OSCC cell lines were responsive to cytokine stimuli (IL6, IL-1ß and TNF-α), but only IL-6 increased migration properties of OSCC cells. This effect was dependent on STAT3-phosphorylation levels, which interfered with Rac1 activation levels. CONCLUSION: Our results suggest that the inflammatory milieu might favor invasion and metastasis of OSCC by the direct effect of macrophage-related cytokines on tumor migration.


Asunto(s)
Movimiento Celular , Citocinas/metabolismo , Neoplasias de la Boca , Carcinoma de Células Escamosas de Cabeza y Cuello , Microambiente Tumoral , Macrófagos Asociados a Tumores , Análisis de Varianza , Cadherinas/metabolismo , Comunicación Celular , Línea Celular Tumoral , Forma de la Célula , Medios de Cultivo Condicionados/farmacología , Citometría de Flujo , Humanos , Inflamación , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Invasividad Neoplásica , Fosforilación , Proteómica , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Factor de Necrosis Tumoral alfa/metabolismo , Macrófagos Asociados a Tumores/citología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/fisiología , Proteína de Unión al GTP rac1/metabolismo
13.
Cells ; 9(10)2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053687

RESUMEN

Cell migration is critical for several physiological and pathophysiological processes. It depends on the coordinated action of kinases, phosphatases, Rho-GTPases proteins, and Ca2+ signaling. Interestingly, ubiquitination events have emerged as regulatory elements of migration. Thus, the role of proteins involved in ubiquitination processes could be relevant to a complete understanding of pro-migratory mechanisms. KCTD5 is a member of Potassium Channel Tetramerization Domain (KCTD) proteins that have been proposed as a putative adaptor for Cullin3-E3 ubiquitin ligase and a novel regulatory protein of TRPM4 channels. Here, we study whether KCTD5 participates in cell migration-associated mechanisms, such as focal adhesion dynamics and cellular spreading. Our results show that KCTD5 CRISPR/Cas9- and shRNA-based depletion in B16-F10 cells promoted an increase in cell migration and cell spreading, and a decrease in the focal adhesion area, consistent with an increased focal adhesion disassembly rate. The expression of a dominant-negative mutant of Rho-GTPases Rac1 precluded the KCTD5 depletion-induced increase in cell spreading. Additionally, KCTD5 silencing decreased the serum-induced Ca2+ response, and the reversion of this with ionomycin abolished the KCTD5 knockdown-induced decrease in focal adhesion size. Together, these data suggest that KCTD5 acts as a regulator of cell migration by modulating cell spreading and focal adhesion dynamics through Rac1 activity and Ca2+ signaling, respectively.


Asunto(s)
Señalización del Calcio/fisiología , Canales de Potasio/metabolismo , Animales , Calcio/metabolismo , Adhesión Celular/genética , Línea Celular , Movimiento Celular/genética , Adhesiones Focales/genética , Humanos , Ratones , Canales de Potasio/fisiología , Transducción de Señal , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo
14.
Biochem Pharmacol ; 177: 113941, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32240650

RESUMEN

In advanced stages of cancer disease, caveolin-1 (CAV1) expression increases and correlates with increased migratory and invasive capacity of the respective tumor cells. Previous findings from our laboratory revealed that specific ECM-integrin interactions and tyrosine-14 phosphorylation of CAV1 are required for CAV1-enhanced melanoma cell migration, invasion and metastasis in vivo. In this context, CAV1 phosphorylation on tyrosine-14 mediated by non-receptor Src-family tyrosine kinases seems to be important; however, the effect of Src-family kinase inhibitors on CAV1-enhanced metastasis in vivo has not been studied. Here, we evaluated the effect of CAV1 and c-Abl overexpression, as well as the use of the Src-family kinase inhibitors, PP2 and dasatinib (more specific for Src/Abl) in lung metastasis of B16F10 melanoma cells. Overexpression of CAV1 and c-Abl enhanced CAV1 phosphorylation and the metastatic potential of the B16F10 murine melanoma cells. Alternatively, treatment with PP2 or dasatinib for 2 h reduced CAV1 tyrosine-14 phosphorylation and levels recovered fully within 12 h of removing the inhibitors. Nonetheless, pre-treatment of cells with these inhibitors for 2 h sufficed to prevent migration, invasion and trans-endothelial migration in vitro. Importantly, the transient decrease in CAV1 phosphorylation by these kinase inhibitors prevented early steps of CAV1-enhanced lung metastasis by B16F10 melanoma cells injected into the tail vein of mice. In conclusion, this study underscores the relevance of CAV1 tyrosine-14 phosphorylation by Src-family kinases during the first steps of the metastatic sequence promoted by CAV1. These findings open up potential options for treatment of metastatic tumors in patients in which Src-family kinase activation and CAV1 overexpression favor dissemination of cancer cells to secondary sites.


Asunto(s)
Caveolina 1/metabolismo , Dasatinib/farmacología , Neoplasias Pulmonares/secundario , Melanoma Experimental/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Neoplasias Cutáneas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Animales , Caveolina 1/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dasatinib/uso terapéutico , Femenino , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Neoplasias Cutáneas/patología , Transfección , Tirosina/metabolismo , Proteína de Unión al GTP rac1/metabolismo
15.
Toxicol Appl Pharmacol ; 380: 114694, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31356930

RESUMEN

Lead (Pb) exposure at high concentrations is associated with poor sperm quality, acrosome alterations, and low fertilization rate. Sperm capacitation and the acrosome reaction (AR) are required for successful fertilization. Actin polymerization is crucial for correct capacitation, and small GTPases, such as RhoA, Rac1, and Cdc42, are involved. This study aimed to evaluate the effects of Pb on sperm fertilization ability, capacitation, AR, and the mechanisms involved in mice exposed to low Pb concentrations. CD1 mice were exposed to 0.01% Pb2+ for 45 days through their drinking water and their spermatozoa were collected from the cauda epididymis-vas deferens to evaluate the following: AR (oAR: initial, sAR: spontaneous, and iAR: induced) using the PNA-FITC assay, sperm capacitation (P-Tyr levels), actin polymerization (phalloidin-TRITC), MDA production (stress oxidative marker), the RhoA, Rac1, and Cdc42 protein levels, and the in vitro fertilization (IVF). After the treatment, the blood Pb (PbB) concentration was 9.4 ±â€¯1.6 µg/dL. Abnormal sperm morphology and the oAR increased (8 and 19%, respectively), whereas the iAR decreased (15%) after a calcium ionophore challenge, and the actin polymerization decreased in the sperm heads (59%) and tails (42%). Rac1 was the only Rho protein to significantly decrease (33%). Spermatozoa from the Pb-treated mice showed a significant reduction in the fertilization rate (19%). Our data suggest that Pb exposure at environmental concentrations (PbB < 10 µg/dL) decreases the acrosome function and affects the sperm fertilization ability; this is probably a consequence of the low Rac1 levels, which did not allow adequate actin polymerization to occur.


Asunto(s)
Contaminantes Ambientales/toxicidad , Plomo/toxicidad , Capacitación Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Actinas/metabolismo , Animales , Femenino , Masculino , Ratones Endogámicos ICR , Neuropéptidos/metabolismo , Espermatozoides/anomalías , Espermatozoides/fisiología , Proteína de Unión al GTP rac1/metabolismo
16.
Toxicon ; 162: 32-39, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30849455

RESUMEN

Lonomia obliqua is a caterpillar of potential therapeutic interest whose venom is able to induce severe blood leakage and modulate leukocyte migration. Since both phenotypes are associated with changes in cytoskeleton dynamics and cell adhesion properties, the aim of this study was to analyze the effects of Lonomia obliqua bristle extract (LOBE) in cell adhesion and migration signaling. Proteomic analysis revealed that epithelial cells (CHO-K1) exposed to LOBE (30 µg/mL, 30 min) exhibited changes in levels of actin regulatory proteins, including RhoGTPases. These changes correlated with an increase in the activity of the RhoGTPase family member Rac as measured by Förster resonance energy transfer (FRET). When plated in migration promoting conditions, CHO-K1 cells exposed to LOBE (10 µg/mL) showed an increase in membrane ruffling after short (30 min) period of incubation that was accompanied by changes in the distribution of the adhesion markers paxillin, vinculin and an increase of focal adhesion kinase autophosphorylation levels (Y397), suggesting changes in cell-extracellular matrix (ECM) adhesion properties and signaling. These data suggest that LOBE possesses bioactive molecules that are capable to modulated cell migration signaling, cytoskeletal dynamics and cell-ECM properties of several cell types.


Asunto(s)
Venenos de Artrópodos/toxicidad , Adhesión Celular/efectos de los fármacos , Mariposas Nocturnas/química , Proteína de Unión al GTP rac1/metabolismo , Actinas/metabolismo , Animales , Células CHO , Movimiento Celular/efectos de los fármacos , Cricetulus , Citoesqueleto/fisiología , Larva/química , Paxillin/metabolismo , Fosforilación , Proteoma/análisis , Vinculina/metabolismo
17.
Cell Death Dis ; 10(2): 143, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760703

RESUMEN

Protein disulfide isomerases including PDIA1 are implicated in cancer progression, but underlying mechanisms are unclear. PDIA1 is known to support vascular Nox1 NADPH oxidase expression/activation. Since deregulated reactive oxygen species (ROS) production underlies tumor growth, we proposed that PDIA1 is an upstream regulator of tumor-associated ROS. We focused on colorectal cancer (CRC) with distinct KRas activation levels. Analysis of RNAseq databanks and direct validation indicated enhanced PDIA1 expression in CRC with constitutive high (HCT116) vs. moderate (HKE3) and basal (Caco2) Ras activity. PDIA1 supported Nox1-dependent superoxide production in CRC; however, we first reported a dual effect correlated with Ras-level activity: in Caco2 and HKE3 cells, loss-of-function experiments indicate that PDIA1 sustains Nox1-dependent superoxide production, while in HCT116 cells PDIA1 restricted superoxide production, a behavior associated with increased Rac1 expression/activity. Transfection of Rac1G12V active mutant into HKE3 cells induced PDIA1 to become restrictive of Nox1-dependent superoxide, while in HCT116 cells treated with Rac1 inhibitor, PDIA1 became supportive of superoxide. PDIA1 silencing promoted diminished cell proliferation and migration in HKE3, not detectable in HCT116 cells. Screening of cell signaling routes affected by PDIA1 silencing highlighted GSK3ß and Stat3. Also, E-cadherin expression after PDIA1 silencing was decreased in HCT116, consistent with PDIA1 support of epithelial-mesenchymal transition. Thus, Ras overactivation switches the pattern of PDIA1-dependent Rac1/Nox1 regulation, so that Ras-induced PDIA1 bypass can directly activate Rac1. PDIA1 may be a crucial regulator of redox-dependent adaptive processes related to cancer progression.


Asunto(s)
Neoplasias del Colon/metabolismo , NADPH Oxidasa 1/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células CACO-2 , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HCT116 , Humanos , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transfección , Proteína de Unión al GTP rac1/metabolismo
18.
Clin Transl Oncol ; 21(4): 404-411, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30182212

RESUMEN

BACKGROUND: The oxidative pentose phosphate pathway (PPP) is essential for cancer metabolism and growth. However, the contribution of 6-phosphogluconate dehydrogenase (6PGD), a key enzyme of PPP, to cervical cancer development remains largely unknown. METHODS: mRNA and protein levels of 6PGD were analyzed in cervical cancer cells and tissues derived from patients and compared to normal counterparts. Using cell culture system and xenograft mouse model, the functions of 6PGD in cervical cancer are determined and its molecular mechanism is analyzed. 6PGD inhibitor physcion and siRNA knockdown were used. RESULTS: In this work, we demonstrate that 6PGD is aberrantly upregulated and activated in cervical cancer cells and patient tissues compared to normal counterparts. Using different approaches and preclinical models, we show that 6PGD inhibition decreases growth and migration, and enhances chemosensitivity in cervical cancer. Mechanistically, inhibition of 6PGD activates AMP-activated protein kinase (AMPK) and decreases RhoA and Rac1 activities. AMPK depletion significantly reduces the effects of 6PGD inhibition in decreasing RhoA and Rac1 activities, growth and migration in cervical cancer cells. CONCLUSIONS: Our work is the first to demonstrate the aberrant expression of 6PGD and its predominant roles in cervical cancer cell growth and migration, via a AMPK-dependent activation. Our findings suggest 6PGD as a potential therapeutic target to enhance chemosensitivity in cervical cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Biomarcadores de Tumor/antagonistas & inhibidores , Fosfogluconato Deshidrogenasa/antagonistas & inhibidores , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimioterapia Adyuvante , Emodina/análogos & derivados , Emodina/farmacología , Emodina/uso terapéutico , Femenino , Expresión Génica , Humanos , Ratones SCID , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Fosfogluconato Deshidrogenasa/genética , Fosfogluconato Deshidrogenasa/metabolismo , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Toxicology ; 394: 35-44, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29197552

RESUMEN

Methylmercury (MeHg) is an environmental neurotoxicant that inhibits neuronal migration. This process requires several cyclic steps involving the formation of membrane protrusions (lamellipodia and filopodia) and focal adhesion turnover. FAK and Src are critical proteins that regulate both processes. The FAK-Src complex promotes the activation of Rac1 and Cdc42, two GTPases involved in the remodeling of the actin cytoskeletal network. Here, we studied the effect of MeHg (1, 10, 100, 500 and 1000nM) on cell migration, the formation of cell protrusions, focal adhesion location and the activation of FAK, Src, Rac1 and Cdc42 using the SH-SY5Y neuroblastoma cell line stimulated with PDGF-BB (PDGF). The data show that MeHg (1-500nM) inhibited PDGF-stimulated cell migration. In PDGF-stimulated cells, MeHg (100-1000nM) decreased protrusions and increased the size of the p-FAKY397 clusters. MeHg also inhibited PDGF-induced FAK and Src activation and, at 100nM, MeHg inhibited the activation of Rac1 and Cdc42. Altogether, the findings show that low concentrations of MeHg inhibit SH-SY5Y cell migration by disrupting the activation and disassembly of FAK. This negatively affects the activation of Src, Rac1 and Cdc42, all of which are critical proteins for the regulation of cell movement. These effects could be related to the MeHg-mediated inhibition of PDGF-induced formation of lamellipodia and filopodia, focal adhesion disassembly and PDGF-induced movement.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Compuestos de Metilmercurio/farmacología , Neuroblastoma/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Familia-src Quinasas/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Neuroblastoma/enzimología , Neuroblastoma/patología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/metabolismo
20.
FASEB J ; 31(11): 4946-4958, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28751526

RESUMEN

Saliva is a key factor that contributes to the high efficiency of wound healing in the oral mucosa. This is not only attributed to physical cues but also to the presence of specific peptides in the saliva, such as histatins. Histatin-1 is a 38 aa antimicrobial peptide, highly enriched in human saliva, which has been previously reported to promote the migration of oral keratinocytes and fibroblasts in vitro However, the participation of histatin-1 in other crucial events required for wound healing, such as angiogenesis, is unknown. Here we demonstrate that histatin-1 promotes angiogenesis, as shown in vivo, using the chick chorioallantoic membrane model, and by an in vitro tube formation assay, using both human primary cultured endothelial cells (HUVECs) and the EA.hy926 cell line. Specifically, histatin-1 promoted endothelial cell adhesion and spreading onto fibronectin, as well as endothelial cell migration in the wound closure and Boyden chamber assays. These actions required the activation of the Ras and Rab interactor 2 (RIN2)/Rab5/Rac1 signaling axis, as histatin-1 increased the recruitment of RIN2, a Rab5-guanine nucleotide exchange factor (GEF) to early endosomes, leading to sequential Rab5/Rac1 activation. Accordingly, interfering with either Rab5 or Rac1 activities prevented histatin-1-dependent endothelial cell migration. Finally, by immunodepletion assays, we showed that salivary histatin-1 is required for the promigratory effects of saliva on endothelial cells. In conclusion, we report that salivary histatin-1 is a novel proangiogenic factor that may contribute to oral wound healing.-Torres, P., Díaz, J., Arce, M., Silva, P., Mendoza, P., Lois, P., Molina-Berríos, A., Owen, G. I., Palma, V., Torres, V. A. The salivary peptide histatin-1 promotes endothelial cell adhesion, migration, and angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Histatinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Proteínas y Péptidos Salivales/farmacología , Inductores de la Angiogénesis/metabolismo , Proteínas Portadoras/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular , Células Endoteliales/patología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Histatinas/metabolismo , Humanos , Mucosa Bucal/lesiones , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Proteínas y Péptidos Salivales/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Proteínas de Unión al GTP rab5/metabolismo , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA