Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Open Biol ; 13(3): 220185, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36854376

RESUMEN

The Rho GTPase family proteins are key regulators of cytoskeletal dynamics. Deregulated activity of Rho GTPases is associated with cancers and neurodegenerative diseases, and their potential as drug targets has long been recognized. Using an economically effective drug screening workflow in fission yeast and human cells, we have identified a Rho GTPase inhibitor, O1. By a suppressor mutant screen in fission yeast, we find a point mutation in the rho1 gene that confers resistance to O1. Consistent with the idea that O1 is the direct inhibitor of Rho1, O1 reduced the cellular amount of activated, GTP-bound Rho1 in wild-type cells, but not in the O1-resistant mutant cells, in which the evolutionarily conserved Ala62 residue is mutated to Thr. Similarly, O1 inhibits activity of the human orthologue RhoA GTPase in tissue culture cells. Our studies illustrate the power of yeast phenotypic screens in the identification and characterization of drugs relevant to human cells and have identified a novel GTPase inhibitor for fission yeast and human cells.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Schizosaccharomyces , Proteína de Unión al GTP rhoA , Humanos , Citoesqueleto , Evaluación Preclínica de Medicamentos , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Schizosaccharomyces/enzimología
2.
Biochem Soc Trans ; 49(5): 2021-2035, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623375

RESUMEN

The RAS superfamily of small GTPases regulates major physiological cellular processes. Mutation or deregulation of these small GTPases, their regulators and/or their effectors are associated with many diseases including cancer. Hence, targeting these classes of proteins is an important therapeutic strategy in cancer. This has been recently achieved with the approval of the first KRASG12C covalent inhibitors for the clinic. However, many other mutants and small GTPases are still considered as 'undruggable' with small molecule inhibitors because of a lack of well-defined pocket(s) at their surface. Therefore, alternative therapeutic strategies have been developed to target these proteins. In this review, we discuss the use of intracellular antibodies and derivatives - reagents that bind their antigen inside the cells - for the discovery of novel inhibitory mechanisms, targetable features and therapeutic strategies to inhibit small GTPases and their downstream pathways. These reagents are also versatile tools used to better understand the biological mechanisms regulated by small GTPases and to accelerate the drug discovery process.


Asunto(s)
Anticuerpos/metabolismo , Proteínas de Repetición de Anquirina Diseñadas/metabolismo , Descubrimiento de Drogas/métodos , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias/enzimología , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Proteínas de Repetición de Anquirina Diseñadas/farmacología , Humanos , Terapia Molecular Dirigida/métodos , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/inmunología , Neoplasias/tratamiento farmacológico , Unión Proteica , Proteolisis/efectos de los fármacos , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología
3.
J Med Chem ; 64(14): 9677-9710, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34236862

RESUMEN

A fundamental role of pancreatic ß-cells to maintain proper blood glucose level is controlled by the Ras superfamily of small GTPases that undergo post-translational modifications, including prenylation. This covalent attachment with either a farnesyl or a geranylgeranyl group controls their localization, activity, and protein-protein interactions. Small GTPases are critical in maintaining glucose homeostasis acting in the pancreas and metabolically active tissues such as skeletal muscles, liver, or adipocytes. Hyperglycemia-induced upregulation of small GTPases suggests that inhibition of these pathways deserves to be considered as a potential therapeutic approach in treating T2D. This Perspective presents how inhibition of various points in the mevalonate pathway might affect protein prenylation and functioning of diabetes-affected tissues and contribute to chronic inflammation involved in diabetes mellitus (T2D) development. We also demonstrate the currently available molecular tools to decipher the mechanisms linking the mevalonate pathway's enzymes and GTPases with diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Hipoglucemiantes/farmacología , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Animales , Diabetes Mellitus Tipo 2/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Humanos , Hipoglucemiantes/química , Estructura Molecular , Proteínas de Unión al GTP Monoméricas/metabolismo , Relación Estructura-Actividad
4.
Bioorg Chem ; 112: 104907, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33979735

RESUMEN

The enzyme leucyl-tRNA synthetase (LRS) and the amino acid leucine regulate the mechanistic target of rapamycin (mTOR) signaling pathway. Leucine-dependent mTORC1 activation depends on GTPase activating protein events mediated by LRS. In a prior study, compound BC-LI-0186 was discovered and shown to interfere with the mTORC1 signaling pathway by inhibiting the LRS-RagD interaction. However, BC-LI-0186 exhibited poor solubility and was metabolized by human liver microsomes. In this study, in silico physicochemical properties and metabolite analysis of BC-LI-0186 are used to investigate the addition of functional groups to improve solubility and microsomal stability. In vitro experiments demonstrated that 7b and 8a had improved chemical properties while still maintaining inhibitory activity against mTORC1. The results suggest a new strategy for the discovery of novel drug candidates and the treatment of diverse mTORC1-related diseases.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Leucina-ARNt Ligasa/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Pirazolonas/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Leucina-ARNt Ligasa/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Estructura Molecular , Proteínas de Unión al GTP Monoméricas/metabolismo , Pirazolonas/síntesis química , Pirazolonas/química , Relación Estructura-Actividad
5.
Front Immunol ; 12: 564720, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679729

RESUMEN

Neutrophil granulocytes represent the first line of defense against invading pathogens. In addition to the production of Reactive Oxygen Species, degranulation, and phagocytosis, these specialized cells are able to extrude Neutrophil Extracellular Traps. Extensive work was done to elucidate the mechanism of this special form of cell death. However, the exact mechanisms are still not fully uncovered. Here we demonstrate that the small GTPase Cdc42 is a negative regulator of NET formation in primary human and murine neutrophils. We present a functional role for Cdc42 activity in NET formation that differs from the already described NETosis pathways. We show that Cdc42 deficiency induces NETs independent of the NADPH-oxidase but dependent on protein kinase C. Furthermore, we demonstrate that Cdc42 deficiency induces NETosis through activation of SK-channels and that mitochondria play a crucial role in this process. Our data therefore suggests a mechanistic role for Cdc42 activity in primary human neutrophils, and identify Cdc42 activity as a target to modulate the formation of Neutrophil Extracellular Traps.


Asunto(s)
Trampas Extracelulares/metabolismo , Mitocondrias/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neutrófilos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Células Cultivadas , Trampas Extracelulares/genética , Humanos , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Neutrófilos/citología , Canales de Potasio Calcio-Activados/metabolismo , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/genética
6.
Cancer Immunol Res ; 9(5): 554-567, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33653716

RESUMEN

Immunotherapy (IT) and targeted therapy (TT) are both effective against melanoma, but their combination is frequently toxic. Here, we investigated whether the sequence of IT (anti-PD-1)→ TT (ceritinib-trametinib or dabrafenib-trametinib) was associated with improved antitumor responses in mouse models of BRAF- and NRAS-mutant melanoma. Mice with NRAS-mutant (SW1) or BRAF-mutant (SM1) mouse melanomas were treated with either IT, TT, or the sequence of IT→TT. Tumor volumes were measured, and samples from the NRAS-mutant melanomas were collected for immune-cell analysis, single-cell RNA sequencing (scRNA-seq), and reverse phase protein analysis (RPPA). scRNA-seq demonstrated that the IT→TT sequence modulated the immune environment, leading to increased infiltration of T cells, monocytes, dendritic cells and natural killer cells, and decreased numbers of tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells. Durable responses to the IT→TT sequence were dependent on T-cell activity, with depletion of CD8+, but not CD4+, T cells abrogating the therapeutic response. An analysis of transcriptional heterogeneity in the melanoma compartment showed the sequence of IT→TT enriched for a population of melanoma cells with increased expression of MHC class I and melanoma antigens. RPPA analysis demonstrated that the sustained immune response induced by IT→TT suppressed tumor-intrinsic signaling pathways required for therapeutic escape. These studies establish that upfront IT improves the responses to TT in BRAF- and NRAS-mutant melanoma models.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Imidazoles/química , Inmunoterapia , Melanoma/genética , Melanoma/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Oximas/química , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Piridonas/química , Pirimidinas/química , Pirimidinonas/química , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Sulfonas/química , Linfocitos T Reguladores/inmunología
7.
Angew Chem Int Ed Engl ; 59(16): 6342-6366, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30869179

RESUMEN

The Ras superfamily of small GTPases are guanine-nucleotide-dependent switches essential for numerous cellular processes. Mutations or dysregulation of these proteins are associated with many diseases, but unsuccessful attempts to target the small GTPases directly have resulted in them being classed as "undruggable". The GTP-dependent signaling of these proteins is controlled by their regulators; guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in the Rho and Rab subfamilies, guanine nucleotide dissociation inhibitors (GDIs). This review covers the recent small molecule and biologics strategies to target the small GTPases through their regulators. It seeks to critically re-evaluate recent chemical biology practice, such as the presence of PAINs motifs and the cell-based readout using compounds that are weakly potent or of unknown specificity. It highlights the vast scope of potential approaches for targeting the small GTPases in the future through their regulatory proteins.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Sitios de Unión , Evaluación Preclínica de Medicamentos , Humanos , Simulación de Dinámica Molecular , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/clasificación , Péptidos/química , Péptidos/metabolismo , Filogenia , Unión Proteica , Bibliotecas de Moléculas Pequeñas/metabolismo , Relación Estructura-Actividad
8.
Hum Mol Genet ; 27(22): 3974-3985, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30137437

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by dopaminergic neuron loss. At present, there are no drugs that stop the progression of PD. As with other multifactorial genetic disorders, genome-wide association studies (GWASs) found multiple risk loci for PD, although their clinical significance remains uncertain. Here, we report the identification of candidate drugs for PD by a method using GWAS data and in silico databases. We identified 57 Food and Drug Administration-approved drug families as candidate neuroprotective drugs for PD. Among them, dabrafenib, which is known as a B-Raf kinase inhibitor and is approved for the treatment of malignant melanoma, showed remarkable cytoprotective effects in neurotoxin-treated SH-SY5Y cells and mice. Dabrafenib was found to inhibit apoptosis, and to enhance the phosphorylation of extracellular signal-regulated kinase (ERK), and inhibit the phosphorylation of c-Jun NH2-terminal kinase. Dabrafenib targets B-Raf, and we confirmed a protein-protein interaction between B-Raf and Rit2, which is coded by RIT2, a PD risk gene in Asians and Caucasians. In RIT2-knockout cells, the phosphorylation of ERK was reduced, and dabrafenib treatment improved the ERK phosphorylation. These data indicated that dabrafenib exerts protective effects against neurotoxicity associated with PD. By using animal model, we confirmed the effectiveness of this in silico screening method. Furthermore, our results suggest that this in silico drug screening system is useful in not only neurodegenerative diseases but also other common diseases such as diabetes mellitus and hypertension.


Asunto(s)
Imidazoles/administración & dosificación , Proteínas de Unión al GTP Monoméricas/genética , Fármacos Neuroprotectores/administración & dosificación , Oximas/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/genética , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Simulación por Computador , Citoprotección/efectos de los fármacos , Bases de Datos de Compuestos Químicos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Aprobación de Drogas , Evaluación Preclínica de Medicamentos/métodos , Estudio de Asociación del Genoma Completo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/tratamiento farmacológico , Ratones , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Fosforilación/efectos de los fármacos , Mapas de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores
9.
J Dairy Res ; 85(2): 204-211, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29785899

RESUMEN

This research paper addresses the hypothesis that RagD is a key signalling factor that regulates amino acid (AA) mediated-casein synthesis and cell proliferation in cow mammary epithelial cells (CMECs). The expression of RagD was analysed at different times during pregnancy and lactation in bovine mammary tissue from dairy cows. We showed that expression of RagD at lactation period was higher (P < 0·05) than that at pregnancy period. When CMECs were treated with methionine (Met) or lysine (Lys), expression of RagD, ß-casein (CSN2), mTOR and p-mTOR, and cell proliferation were increased. Further, when CMECs were treated to overexpress RagD, expression of CSN2, mTOR and p-mTOR, and cell proliferation were up-regulated. Furthermore, the increase in expression of CSN2, mTOR and p-mTOR, and cell proliferation in response to Met or Lys supply was inhibited by inhibiting RagD, and those effects were reversed in the overexpression model. When CMECs were treated with RagD overexpression together with mTOR inhibition or conversely with RagD inhibition together with mTOR overexpression, results showed that the increase in expression of CSN2 and cell proliferation in response to RagD overexpression was prevented by inhibiting mTOR, and those effects were reversed by overexpressing mTOR. The interaction of RagD with subunit proteins of mTORC1 was analysed, and the result showed that RagD interacted with Raptor. CMECs were treated with Raptor inhibition, and the result showed that the increase in expression of mTOR and p-mTOR in response to RagD overexpression was inhibited by inhibiting Raptor.In conclusion, our study showed that RagD is an important activation factor of mTORC1 in CMECs, activating AA-mediated casein synthesis and cell proliferation, potentially acting via Raptor.


Asunto(s)
Caseínas/biosíntesis , Bovinos , Glándulas Mamarias Animales/metabolismo , Proteínas de Unión al GTP Monoméricas/fisiología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos/fisiología , Animales , Proliferación Celular/fisiología , Células Cultivadas , Células Epiteliales , Femenino , Expresión Génica/efectos de los fármacos , Lactancia/fisiología , Lisina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Metionina/farmacología , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Embarazo , Proteína Reguladora Asociada a mTOR/antagonistas & inhibidores , Proteína Reguladora Asociada a mTOR/fisiología , Serina-Treonina Quinasas TOR/genética
10.
Anticancer Agents Med Chem ; 18(3): 314-322, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28356010

RESUMEN

Ras association domain family member 5 (RASSF5, also named NORE1) is an identified member of the RASSF gene family which could bind selectively to activate Ras and function as an antineoplastic effector in multiple cellular regulations. While highly expressed in majority of normal tissues, RASSF5 is epigenetically inactivated by promoter hypermethylation in numerous cancer cell lines and primary cancers, suggesting it as a potential tumor suppressor. Nevertheless, the physiologic significance of RASSF5 in tumorigenesis remains unclear. We performed a systematic literature review and assessment from PUBMED and MEDLINE databases in this article. RASSF5 is involved in a series of cellular responses including apoptosis, senescence, cell cycle regulation, differentiation and cell proliferation and the inactivation of RASSF5 has been implicated to participate in the oncogenesis, progression and poor prognosis of human cancers. In this review, we mainly elucidate the acknowledged structure, progress in the verified functions and research advances of RASSF5 and the probably relevant signaling pathways. Based on these evidences, potentiality of RASSF5 as a new therapeutic target for human cancers may play a significant role in future oncotherapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Adaptadoras Transductoras de Señales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Proliferación Celular/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/genética
11.
Genes Cells ; 22(12): 993-1003, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29193479

RESUMEN

RASSF6, a member of the tumor suppressor Ras-association domain family proteins, induces apoptosis in the caspase-dependent and caspase-independent manners. RASSF6 interacts with MDM2 and stabilizes p53. BCL-XL is a prosurvival member of BCL-2 family proteins. BCL-XL directly inhibits proapoptotic BAX and BAK. BCL-XL also traps tBID, a proapoptotic activator BH3-only protein, and sequesters p53. In addition, BCL-XL regulates the mitochondrial membrane permeability via voltage-dependent anion channel. In these manners, BCL-XL plays an antiapoptotic role. We report the interaction of BCL-XL with RASSF6. BCL-XL inhibits the interaction between RASSF6 and MDM2 and suppresses p53 expression. Consequently, BCL-XL antagonizes RASSF6-mediated apoptosis. Thus, the inhibition of RASSF6-mediated apoptosis also underlies the prosurvival role of BCL-XL.


Asunto(s)
Apoptosis , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína bcl-X/metabolismo , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Humanos , Transducción de Señal
12.
Cancer Res ; 77(23): 6513-6523, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28951457

RESUMEN

BRAF V600E colorectal cancers are insensitive to RAF inhibitor monotherapy due to feedback reactivation of receptor tyrosine kinase signaling. Combined RAF and EGFR inhibition exerts a therapeutic effect, but resistance invariably develops through undefined mechanisms. In this study, we determined that colorectal cancer progression specimens invariably harbored lesions in elements of the RAS-RAF-MEK-ERK pathway. Genetic amplification of wild-type RAS was a recurrent mechanism of resistance in colorectal cancer patients that was not seen in similarly resistant melanomas. We show that wild-type RAS amplification increases receptor tyrosine kinase-dependent activation of RAS more potently in colorectal cancer than in melanoma and causes resistance only in the former. Currently approved RAF inhibitors inhibit RAF monomers but not dimers. All the drug-resistant lesions we identified activate BRAF V600E dimerization directly or by elevating RAS-GTP. Overall, our results show that mechanisms of resistance converge on formation of RAF dimers and that inhibiting EGFR and RAF dimers can effectively suppress ERK-driven growth of resistant colorectal cancer. Cancer Res; 77(23); 6513-23. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/patología , Melanoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Quinasas raf/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/genética , Dimerización , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Células HT29 , Humanos , Melanoma/genética , Ratones , Ratones Desnudos , Ratones SCID , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/metabolismo , Quinasas raf/antagonistas & inhibidores , Proteínas ras/genética
13.
Biochem Biophys Res Commun ; 491(2): 429-435, 2017 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-28716730

RESUMEN

Connexin 43 (Cx43), a ubiquitous connexin expressed in the heart and skin, is associated with a variety of hereditary conditions. Therefore, the characterization of Cx43-interacting proteins and their dynamics is important to understand not only the molecular mechanisms underlying pathological malfunction of gap junction-mediated intercellular communication but also to identify novel and unanticipated biological functions of Cx43. In the present study, we observed potential targets of Cx43 to determine new molecular functions in cardio-protection. MALDI-TOF mass spectrometry analysis of Cx43 co-immunoprecipitated proteins showed that Cx43 interacts with several proteins related to metabolism. In GeneMANIA network analysis, SGSM3, which has not been previously associated with Cx43, was highly correlated with Cx43 in heart functions, and high levels of SGSM3 appeared to induce the turnover of Cx43 through lysosomal degradation in myocardial infarcted rat hearts. Moreover, we confirmed that lysosomal degradation of Cx43 is dependent upon the interaction between SGSM3 and Cx43 in H9c2 cardiomyocytes. The functional importance of the interaction between SGSM3 and Cx43 was confirmed by results showing that Cx43 expression was enhanced by SGSM3 siRNA knockdown in H9c2 cells. In summary, the results of this study elucidate the molecular mechanisms in which Cx43 with SGSM3 is degraded in myocardial infarcted rat hearts, which may contribute to the establishment of new therapeutic targets to modulate cardiac function in physiological and pathological conditions.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Comunicación Celular , Línea Celular , Conexina 43/genética , Vasos Coronarios/patología , Vasos Coronarios/cirugía , Uniones Comunicantes/patología , Uniones Comunicantes/ultraestructura , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ligadura , Lisosomas/metabolismo , Masculino , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Unión Proteica , Mapeo de Interacción de Proteínas , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
14.
J Biol Chem ; 292(29): 12220-12231, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28546426

RESUMEN

First messenger-dependent activation of MAP kinases in neuronal and endocrine cells is critical for cell differentiation and function and requires guanine nucleotide exchange factor (GEF)-mediated activation of downstream Ras family small GTPases, which ultimately lead to ERK, JNK, and p38 phosphorylation. Because there are numerous GEFs and also a host of Ras family small GTPases, it is important to know which specific GEF-small GTPase dyad functions in a given cellular process. Here we investigated the upstream activators and downstream effectors of signaling via the GEF Epac2 in the neuroendocrine NS-1 cell line. Three cAMP sensors, Epac2, PKA, and neuritogenic cAMP sensor-Rapgef2, mediate distinct cellular outputs: p38-dependent growth arrest, cAMP response element-binding protein-dependent cell survival, and ERK-dependent neuritogenesis, respectively, in these cells. Previously, we found that cAMP-induced growth arrest of PC12 and NS-1 cells requires Epac2-dependent activation of p38 MAP kinase, which posed the important question of how Epac2 engages p38 without simultaneously activating other MAP kinases in neuronal and endocrine cells. We now show that the small GTP-binding protein Rap2A is the obligate effector for, and GEF substrate of, Epac2 in mediating growth arrest through p38 activation in NS-1 cells. This new pathway is distinctly parcellated from the G protein-coupled receptor → Gs → adenylate cyclase → cAMP → PKA → cAMP response element-binding protein pathway mediating cell survival and the G protein-coupled receptor → Gs → adenylate cyclase → cAMP → neuritogenic cAMP sensor-Rapgef2 → B-Raf → MEK → ERK pathway mediating neuritogenesis in NS-1 cells.


Asunto(s)
AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Sistema de Señalización de MAP Quinasas , Células Neuroendocrinas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Unión al GTP rap/agonistas , Animales , Línea Celular Tumoral , Activación Enzimática , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ligandos , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Células Neuroendocrinas/citología , Neurogénesis , Fosforilación , Prenilación de Proteína , Interferencia de ARN , Ratas , Proteínas Recombinantes/metabolismo , Proteínas de Unión al GTP rap/antagonistas & inhibidores , Proteínas de Unión al GTP rap/genética , Proteínas de Unión al GTP rap/metabolismo , Proteínas ras/antagonistas & inhibidores , Proteínas ras/genética , Proteínas ras/metabolismo
15.
PLoS One ; 12(1): e0169052, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28046007

RESUMEN

SAMHD1 hydrolyzes 2'-deoxynucleoside-5'-triphosphates (dNTPs) into 2'-deoxynucleosides and inorganic triphosphate products. In this paper, we evaluated the impact of 2' sugar moiety substitution for different nucleotides on being substrates for SAMHD1 and mechanisms of actions for the results. We found that dNTPs ((2'R)-2'-H) are only permissive in the catalytic site of SAMHD1 due to L150 exclusion of (2'R)-2'-F and (2'R)-2'-OH nucleotides. However, arabinose ((2'S)-2'-OH) nucleoside-5'-triphosphates analogs are permissive to bind in the catalytic site and be hydrolyzed by SAMHD1. Moreover, when the (2'S)-2' sugar moiety is increased to a (2'S)-2'-methyl as with the SMDU-TP analog, we detect inhibition of SAMHD1's dNTPase activity. Our computational modeling suggests that (2'S)-2'-methyl sugar moiety clashing with the Y374 of SAMHD1. We speculate that SMDU-TP mechanism of action requires that the analog first docks in the catalytic pocket of SAMHD1 but prevents the A351-V378 helix conformational change from being completed, which is needed before hydrolysis can occur. Collectively we have identified stereoselective 2' substitutions that reveal nucleotide substrate specificity for SAMHD1, and a novel inhibitory mechanism for the dNTPase activity of SAMHD1. Importantly, our data is beneficial for understanding if FDA-approved antiviral and anticancer nucleosides are hydrolyzed by SAMHD1 in vivo.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Trifosfato de Arabinofuranosil Citosina , Carbohidratos/química , Pollos , Humanos , Hidrólisis , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Modelos Moleculares , Monocitos/citología , Nucleótidos/metabolismo , Multimerización de Proteína/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD , Especificidad por Sustrato/efectos de los fármacos
16.
Int J Mol Sci ; 17(4): 438, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-27043527

RESUMEN

The "target of rapamycin" (TOR) nutritional signaling pathway and juvenile hormone (JH) regulation of vitellogenesis has been known for a long time. However, the interplay between these two pathways regulating vitellogenin (Vg) expression remains obscure. Here, we first demonstrated the key role of amino acids (AAs) in activation of Vg synthesis and egg development in Nilaparvata lugens using chemically defined artificial diets. AAs induced the expression of TOR and S6K (S6 kinase), whereas RNAi-mediated silencing of these two TOR pathway genes and rapamycin application strongly inhibited the AAs-induced Vg synthesis. Furthermore, knockdown of Rheb (Ras homologue enriched in brain), TOR, S6K and application of rapamycin resulted in a dramatic reduction in the mRNA levels of jmtN (juvenile hormone acid methyltransferase, JHAMT). Application of JH III on the RNAi (Rheb and TOR) and rapamycin-treated females partially rescued the Vg expression. Conversely, knockdown of either jmtN or met (methoprene-tolerant, JH receptor) and application of JH III had no effects on mRNA levels of Rheb, TOR and S6K and phosphorylation of S6K. In summary, our results demonstrate that the TOR pathway induces JH biosynthesis that in turn regulates AAs-mediated Vg synthesis in N. lugens.


Asunto(s)
Hemípteros/metabolismo , Proteínas de Insectos/metabolismo , Hormonas Juveniles/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vitelogeninas/biosíntesis , Aminoácidos/metabolismo , Animales , Antibacterianos/farmacología , Encéfalo/metabolismo , Femenino , Hemípteros/clasificación , Hemípteros/crecimiento & desarrollo , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/genética , Hormonas Juveniles/biosíntesis , Hormonas Juveniles/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Óvulo/metabolismo , Filogenia , Interferencia de ARN , ARN Mensajero/metabolismo , Reproducción , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Vitelogénesis/efectos de los fármacos
17.
PLoS One ; 11(4): e0154280, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27101143

RESUMEN

Endoplasmic reticulum (ER)-mitochondrial contact sites play a pivotal role in exchange of lipids and ions between the two organelles. How size and function of these contact sites are regulated remains elusive. Here we report a previously unanticipated, but conserved role of the small GTPase Sar1 in the regulation of ER-mitochondrial contact site size. Activated Sar1 introduces membrane curvature through its N-terminal amphiphatic helix at the ER-mitochondria interphase and thereby reducing contact size. Conversely, the S. cerevisiae N3-Sar1 mutant, in which curvature induction is decreased, caused an increase in ER-mitochondrial contacts. As a consequence, ER tubules are no longer able to mark the prospective scission site on mitochondria, thereby impairing mitochondrial dynamics. Consistently, blocking mitochondrial fusion partially rescued, whereas deletion of the dynamin-like protein enhanced the phenotype in the sar1D32G mutant. We conclude that Sar1 regulates the size of ER-mitochondria contact sites through its effects on membrane curvature.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/genética , Mitocondrias/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Proteínas de Transporte Vesicular/genética , Sustitución de Aminoácidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/metabolismo , Secuencia Conservada , Dinaminas/genética , Dinaminas/metabolismo , Retículo Endoplásmico/química , Retículo Endoplásmico/ultraestructura , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Transporte Iónico , Metabolismo de los Lípidos , Mitocondrias/química , Mitocondrias/ultraestructura , Dinámicas Mitocondriales , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/metabolismo , Mutación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Proteínas de Transporte Vesicular/metabolismo
18.
Biochem Pharmacol ; 106: 30-45, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26851491

RESUMEN

Massive activation of infected CD4+ T cells during acute HIV-1 infection leads to reservoir seeding and T-cell destruction. During T-cell activation, the antiviral effect of the innate factor SAMHD1 is neutralized through phosphorylation at T592, allowing HIV-1 infection. Dasatinib, a tyrosine kinase inhibitor currently used for treating chronic myeloid leukemia, has been described to control HIV-1 replication through its negative effect on T-cell proliferation and viral entry. We demonstrate that Dasatinib can actually interfere with SAMHD1 phosphorylation in human peripheral blood lymphocytes, preserving its antiviral activity against HIV-1. Dasatinib prevented SAMHD1 phosphorylation in vitro and ex vivo, impairing HIV-1 reverse transcription and proviral integration. This was the major mechanism of action because the presence of Vpx, which degrades SAMHD1, in HIV-1 virions impeded the inhibitory effect of Dasatinib on HIV-1 replication. In fact, infection with VSV-pseudotyped HIV-1 virions and fusion of BlaM-Vpr-containing HIV-1 viruses with activated PBMCs in the presence of Dasatinib suggested that Dasatinib was not acting at fusion level. Finally, PBMCs from patients on chronic treatment with Dasatinib showed a lower level of SAMHD1 phosphorylation in response to activating stimuli and low susceptibility to HIV-1 infection ex vivo. Consequently, Dasatinib is a compound currently used in clinic that preserves the antiviral function of SAMHD1. Using Dasatinib as adjuvant of antiretroviral therapy during early primary HIV-1 infection would contribute to reduce viral replication and spread, prevent reservoir seeding, and preserve CD4 counts and CTL responses. These events would create a more favorable virologic and immunologic environment for future interventional studies aiming at HIV-1 eradication.


Asunto(s)
Fármacos Anti-VIH/farmacología , Antineoplásicos/farmacología , Dasatinib/farmacología , Infecciones por VIH/tratamiento farmacológico , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Replicación Viral/efectos de los fármacos , Adulto , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/virología , Proliferación Celular/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Infecciones por VIH/enzimología , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/genética , VIH-1/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Activación de Linfocitos , Masculino , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Fosforilación/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD , Transducción de Señal , Vesiculovirus/genética , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
19.
J Biol Chem ; 291(3): 1014-27, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26546679

RESUMEN

The majority of biosynthetic secretory proteins initiate their journey through the endomembrane system from specific subdomains of the endoplasmic reticulum. At these locations, coated transport carriers are generated, with the Sar1 GTPase playing a critical role in membrane bending, recruitment of coat components, and nascent vesicle formation. How these events are appropriately coordinated remains poorly understood. Here, we demonstrate that Sar1 acts as the curvature-sensing component of the COPII coat complex and highlight the ability of Sar1 to bind more avidly to membranes of high curvature. Additionally, using an atomic force microscopy-based approach, we further show that the intrinsic GTPase activity of Sar1 is necessary for remodeling lipid bilayers. Consistent with this idea, Sar1-mediated membrane remodeling is dramatically accelerated in the presence of its guanine nucleotide-activating protein (GAP), Sec23-Sec24, and blocked upon addition of guanosine-5'-[(ß,γ)-imido]triphosphate, a poorly hydrolysable analog of GTP. Our results also indicate that Sar1 GTPase activity is stimulated by membranes that exhibit elevated curvature, potentially enabling Sar1 membrane scission activity to be spatially restricted to highly bent membranes that are characteristic of a bud neck. Taken together, our data support a stepwise model in which the amino-terminal amphipathic helix of GTP-bound Sar1 stably penetrates the endoplasmic reticulum membrane, promoting local membrane deformation. As membrane bending increases, Sar1 membrane binding is elevated, ultimately culminating in GTP hydrolysis, which may destabilize the bilayer sufficiently to facilitate membrane fission.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/metabolismo , Guanosina Trifosfato/metabolismo , Modelos Biológicos , Proteínas de Unión al GTP Monoméricas/metabolismo , Sustitución de Aminoácidos , Animales , Vesículas Cubiertas por Proteínas de Revestimiento/efectos de los fármacos , Vesículas Cubiertas por Proteínas de Revestimiento/ultraestructura , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/ultraestructura , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Retículo Endoplásmico/ultraestructura , Inhibidores Enzimáticos/farmacología , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Guanilil Imidodifosfato/farmacología , Humanos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Microdominios de Membrana/ultraestructura , Microscopía de Fuerza Atómica , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Forma de los Orgánulos/efectos de los fármacos , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
20.
Cancer Chemother Pharmacol ; 77(5): 883-93, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26614508

RESUMEN

Nucleotide metabolism is the target of a large number of anticancer drugs including antimetabolites and specific enzyme inhibitors. We review scientific findings that over the last 10-15 years have allowed the identification of several intracellular nucleotide-degrading enzymes as cancer drug targets, and discuss further potential therapeutic applications for Rcl, SAMHD1, MTH1 and cN-II. We believe that enzymes involved in nucleotide metabolism represent potent alternatives to conventional cancer chemotherapy targets.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Neoplasias , Nucleótidos/metabolismo , 5'-Nucleotidasa/antagonistas & inhibidores , Animales , Antineoplásicos/química , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Humanos , Terapia Molecular Dirigida , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , N-Glicosil Hidrolasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Proteínas Nucleares/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteína 1 que Contiene Dominios SAM y HD
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...