Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 544
Filtrar
1.
Respir Res ; 25(1): 295, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095798

RESUMEN

Shortening of airway smooth muscle and bronchoconstriction are pathognomonic for asthma. Airway shortening occurs through calcium-dependent activation of myosin light chain kinase, and RhoA-dependent calcium sensitization, which inhibits myosin light chain phosphatase. The mechanism through which pro-contractile stimuli activate calcium sensitization is poorly understood. Our review of the literature suggests that pro-contractile G protein coupled receptors likely signal through G12/13 to activate RhoA and mediate calcium sensitization. This hypothesis is consistent with the effects of pro-contractile agonists on RhoA and Rho kinase activation, actin polymerization and myosin light chain phosphorylation. Recognizing the likely role of G12/13 signaling in the pathophysiology of asthma rationalizes the effects of pro-contractile stimuli on airway hyperresponsiveness, immune activation and airway remodeling, and suggests new approaches for asthma treatment.


Asunto(s)
Asma , Transducción de Señal , Asma/metabolismo , Asma/fisiopatología , Asma/tratamiento farmacológico , Humanos , Transducción de Señal/fisiología , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Músculo Liso/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/fisiología
2.
Nat Immunol ; 25(9): 1718-1730, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39025963

RESUMEN

Germinal centers (GCs) that form in mucosal sites are exposed to gut-derived factors that have the potential to influence homeostasis independent of antigen receptor-driven selective processes. The G-protein Gα13 confines B cells to the GC and limits the development of GC-derived lymphoma. We discovered that Gα13-deficiency fuels the GC reaction via increased mTORC1 signaling and Myc protein expression specifically in the mesenteric lymph node (mLN). The competitive advantage of Gα13-deficient GC B cells (GCBs) in mLN was not dependent on T cell help or gut microbiota. Instead, Gα13-deficient GCBs were selectively dependent on dietary nutrients likely due to greater access to gut lymphatics. Specifically, we found that diet-derived glutamine supported proliferation and Myc expression in Gα13-deficient GCBs in the mLN. Thus, GC confinement limits the effects of dietary glutamine on GC dynamics in mucosal tissues. Gα13 pathway mutations coopt these processes to promote the gut tropism of aggressive lymphoma.


Asunto(s)
Linfocitos B , Proliferación Celular , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Centro Germinal , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Noqueados , Centro Germinal/inmunología , Centro Germinal/metabolismo , Animales , Ratones , Linfocitos B/inmunología , Linfocitos B/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/inmunología , Nutrientes/metabolismo , Transducción de Señal , Glutamina/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/inmunología
3.
Breast Cancer Res ; 26(1): 113, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965558

RESUMEN

GNA13 (Gα13) is one of two alpha subunit members of the G12/13 family of heterotrimeric G-proteins which mediate signaling downstream of GPCRs. It is known to be essential for embryonic development and vasculogenesis and has been increasingly shown to be involved in mediating several steps of cancer progression. Recent studies found that Gα13 can function as an oncogene and contributes to progression and metastasis of multiple tumor types, including ovarian, head and neck and prostate cancers. In most cases, Gα12 and Gα13, as closely related α-subunits in the subfamily, have similar cellular roles. However, in recent years their differences in signaling and function have started to emerge. We previously identified that Gα13 drives invasion of Triple Negative Breast Cancer (TNBC) cells in vitro. As a highly heterogenous disease with various well-defined molecular subtypes (ER+ /Her2-, ER+ /Her2+, Her2+, TNBC) and subtype associated outcomes, the function(s) of Gα13 beyond TNBC should be explored. Here, we report the finding that low expression of GNA13 is predictive of poorer survival in breast cancer, which challenges the conventional idea of Gα12/13 being universal oncogenes in solid tumors. Consistently, we found that Gα13 suppresses the proliferation in multiple ER+ breast cancer cell lines (MCF-7, ZR-75-1 and T47D). Loss of GNA13 expression drives cell proliferation, soft-agar colony formation and in vivo tumor formation in an orthotopic xenograft model. To evaluate the mechanism of Gα13 action, we performed RNA-sequencing analysis on these cell lines and found that loss of GNA13 results in the upregulation of MYC signaling pathways in ER+ breast cancer cells. Simultaneous silencing of MYC reversed the proliferative effect from the loss of GNA13, validating the role of MYC in Gα13 regulation of proliferation. Further, we found Gα13 regulates the expression of MYC, at both the transcript and protein level in an ERα dependent manner. Taken together, our study provides the first evidence for a tumor suppressive role for Gα13 in breast cancer cells and demonstrates for the first time the direct involvement of Gα13 in ER-dependent regulation of MYC signaling. With a few exceptions, elevated Gα13 levels are generally considered to be oncogenic, similar to Gα12. This study demonstrates an unexpected tumor suppressive role for Gα13 in ER+ breast cancer via regulation of MYC, suggesting that Gα13 can have subtype-dependent tumor suppressive roles in breast cancer.


Asunto(s)
Proliferación Celular , Receptor alfa de Estrógeno , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-myc , Humanos , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Femenino , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética , Animales , Línea Celular Tumoral , Ratones , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Transducción de Señal , Regulación hacia Arriba
4.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39000269

RESUMEN

Gα13 and Gα12, encoded by the GNA13 and GNA12 genes, respectively, are members of the G12 family of Gα proteins that, along with their associated Gßγ subunits, mediate signaling from specific G protein-coupled receptors (GPCRs). Advanced prostate cancers have increased expression of GPCRs such as CXC Motif Chemokine Receptor 4 (CXCR4), lysophosphatidic acid receptor (LPAR), and protease activated receptor 1 (PAR-1). These GPCRs signal through either the G12 family, or through Gα13 exclusively, often in addition to other G proteins. The effect of Gα13 can be distinct from that of Gα12, and the role of Gα13 in prostate cancer initiation and progression is largely unexplored. The oncogenic effect of Gα13 on cell migration and invasion in prostate cancer has been characterized, but little is known about other biological processes such as mitochondrial function and oxidative stress. Current knowledge on the link between Gα13 and oxidative stress is based on animal studies in which GPCR-Gα13 signaling decreased superoxide levels, and the overexpression of constitutively active Gα13 promoted antioxidant gene activation. In human samples, mitochondrial superoxide dismutase 2 (SOD2) correlates with prostate cancer risk and prognostic Gleason grade. However, overexpression of SOD2 in prostate cancer cells yielded conflicting results on cell growth and survival under basal versus oxidative stress conditions. Hence, it is necessary to explore the effect of Gα13 on prostate cancer tumorigenesis, as well as the effect of Gα13 on SOD2 in prostate cancer cell growth under oxidative stress conditions.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Mitocondrias , Estrés Oxidativo , Neoplasias de la Próstata , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Mitocondrias/metabolismo , Mitocondrias/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Animales , Transducción de Señal , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/genética
5.
Sci Rep ; 14(1): 11119, 2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750247

RESUMEN

G-protein-coupled receptors (GPCRs) transduce diverse signals into the cell by coupling to one or several Gα subtypes. Of the 16 Gα subtypes in human cells, Gα12 and Gα13 belong to the G12 subfamily and are reported to be functionally different. Notably, certain GPCRs display selective coupling to either Gα12 or Gα13, highlighting their significance in various cellular contexts. However, the structural basis underlying this selectivity remains unclear. Here, using a Gα12-coupled designer receptor exclusively activated by designer drugs (DREADD; G12D) as a model system, we identified residues in the α5 helix and the receptor that collaboratively determine Gα12-vs-Gα13 selectivity. Residue-swapping experiments showed that G12D distinguishes differences between Gα12 and Gα13 in the positions G.H5.09 and G.H5.23 in the α5 helix. Molecular dynamics simulations observed that I378G.H5.23 in Gα12 interacts with N1032.39, S1693.53 and Y17634.53 in G12D, while H364G.H5.09 in Gα12 interact with Q2645.71 in G12D. Screening of mutations at these positions in G12D identified G12D mutants that enhanced coupling with Gα12 and to an even greater extent with Gα13. Combined mutations, most notably the dual Y17634.53H and Q2645.71R mutant, further enhanced Gα12/13 coupling, thereby serving as a potential Gα12/13-DREADD. Such novel Gα12/13-DREADD may be useful in future efforts to develop drugs that target Gα12/13 signaling as well as to identify their therapeutic indications.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Simulación de Dinámica Molecular , Receptores Acoplados a Proteínas G , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/química , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Células HEK293 , Drogas de Diseño/química , Drogas de Diseño/metabolismo , Unión Proteica
6.
Sci Rep ; 14(1): 6335, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38491049

RESUMEN

Inflammatory bowel disease (IBD) pathogenesis involves complex inflammatory events and cell death. Although IBD involves mainly necrosis in the digestive tract, pyroptosis has also been recognized. Nonetheless, the underlying basis is elusive. Gα12/13 overexpression may affect endoplasmic reticulum (ER) stress. This study examined how Gα12/13 and ER stress affect pyroptosis using dextran sulfate sodium (DSS)-induced colitis models. Gα12/13 levels were increased in the distal and proximal colons of mice exposed to a single cycle of DSS, as accompanied by increases of IRE1α, ATF6, and p-PERK. Moreover, Il-6, Il-1ß, Ym1, and Arg1 mRNA levels were increased with caspase-1 and IL-1ß activation, supportive of pyroptosis. In the distal colon, RIPK1/3 levels were enhanced to a greater degree, confirming necroptosis. By contrast, the mice subjected to three cycles of DSS treatments showed decreases of Gα12/13, as accompanied by IRE1α and ATF6 suppression, but increases of RIPK1/3 and c-Cas3. AZ2 treatment, which inhibited Gα12, has an anti-pyroptotic effect against a single cycle of colitis. These results show that a single cycle of DSS-induced colitis may cause ER stress-induced pyroptosis as mediated by Gα12 overexpression in addition to necroptosis, but three cycles model induces only necroptosis, and that AZ2 may have an anti-pyroptotic effect.


Asunto(s)
Colitis , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Animales , Ratones , Colitis/metabolismo , Colitis/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piroptosis
7.
Scand J Gastroenterol ; 59(6): 710-721, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38357893

RESUMEN

BACKGROUND: The lncRNA TRG-AS1 and its co-expressed gene P2RY10 are important for colorectal cancer (CRC) occurrence and development. The purpose of our research was to explore the roles of TRG-AS1 and P2RY10 in CRC progression. METHODS: The abundance of TRG-AS1 and P2RY10 in CRC cell lines (HT-29 and LoVo) and normal colon cells FHC was determined and difference between CRC cells and normal cells was compared. LoVo cells were transfected with si-TRG-AS1 and si-P2RY10 constructs. Subsequently, the viability, colony formation, and migration of the transfected cells were analyzed using cell counting kit-8, clonogenicity, and scratch-wound/Transwell® assays, respectively. Cells overexpressing GNA13 were used to further explore the relationship between TRG-AS1 and P2RY10 along with their downstream functions. Finally, nude mice were injected with different transfected cell types to observe tumor formation in vivo. RESULTS: TRG-AS1 and P2RY10 were significantly upregulated in HT-29 and LoVo compared to FHC cells. TRG-AS1 knockdown and P2RY10 silencing suppressed the viability, colony formation, and migration of LoVo cells. TRG-AS1 knockdown downregulated the expression of P2RY10, GNA12, and GNA13, while P2RY10 silencing downregulated the expression of TRG-AS1, GNA12, and GNA13. Additionally, GNA13 overexpression reversed the cell growth and gene expression changes in LoVo cells induced by TRG-AS1 knockdown or P2RY10 silencing. In vivo experiments revealed that CRC tumor growth was suppressed by TRG-AS1 knockdown and P2RY10 silencing. CONCLUSIONS: TRG-AS1 knockdown repressed the growth of HT-29 and LoVo by regulating P2RY10 and GNA13 expression.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , ARN Largo no Codificante , Animales , Humanos , Ratones , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Células HT29 , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba
8.
Nature ; 626(7997): 128-135, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38233523

RESUMEN

The assembly and specification of synapses in the brain is incompletely understood1-3. Latrophilin-3 (encoded by Adgrl3, also known as Lphn3)-a postsynaptic adhesion G-protein-coupled receptor-mediates synapse formation in the hippocampus4 but the mechanisms involved remain unclear. Here we show in mice that LPHN3 organizes synapses through a convergent dual-pathway mechanism: activation of Gαs signalling and recruitment of phase-separated postsynaptic protein scaffolds. We found that cell-type-specific alternative splicing of Lphn3 controls the LPHN3 G-protein-coupling mode, resulting in LPHN3 variants that predominantly signal through Gαs or Gα12/13. CRISPR-mediated manipulation of Lphn3 alternative splicing that shifts LPHN3 from a Gαs- to a Gα12/13-coupled mode impaired synaptic connectivity as severely as the overall deletion of Lphn3, suggesting that Gαs signalling by LPHN3 splice variants mediates synapse formation. Notably, Gαs-coupled, but not Gα12/13-coupled, splice variants of LPHN3 also recruit phase-transitioned postsynaptic protein scaffold condensates, such that these condensates are clustered by binding of presynaptic teneurin and FLRT ligands to LPHN3. Moreover, neuronal activity promotes alternative splicing of the synaptogenic Gαs-coupled variant of LPHN3. Together, these data suggest that activity-dependent alternative splicing of a key synaptic adhesion molecule controls synapse formation by parallel activation of two convergent pathways: Gαs signalling and clustered phase separation of postsynaptic protein scaffolds.


Asunto(s)
Empalme Alternativo , Receptores Acoplados a Proteínas G , Receptores de Péptidos , Sinapsis , Animales , Ratones , Empalme Alternativo/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gs , Ligandos , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Sinapsis/metabolismo , Transducción de Señal
9.
Sci Rep ; 13(1): 22412, 2023 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-38104152

RESUMEN

In silico interrogation of glioblastoma (GBM) in The Cancer Genome Atlas (TCGA) revealed upregulation of GNA12 (Gα12), encoding the alpha subunit of the heterotrimeric G-protein G12, concomitant with overexpression of multiple G-protein coupled receptors (GPCRs) that signal through Gα12. Glioma stem cell lines from patient-derived xenografts also showed elevated levels of Gα12. Knockdown (KD) of Gα12 was carried out in two different human GBM stem cell (GSC) lines. Tumors generated in vivo by orthotopic injection of Gα12KD GSC cells showed reduced invasiveness, without apparent changes in tumor size or survival relative to control GSC tumor-bearing mice. Transcriptional profiling of GSC-23 cell tumors revealed significant differences between WT and Gα12KD tumors including reduced expression of genes associated with the extracellular matrix, as well as decreased expression of stem cell genes and increased expression of several proneural genes. Thrombospondin-1 (THBS1), one of the genes most repressed by Gα12 knockdown, was shown to be required for Gα12-mediated cell migration in vitro and for in vivo tumor invasion. Chemogenetic activation of GSC-23 cells harboring a Gα12-coupled DREADD also increased THBS1 expression and in vitro invasion. Collectively, our findings implicate Gα12 signaling in regulation of transcriptional reprogramming that promotes invasiveness, highlighting this as a potential signaling node for therapeutic intervention.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Animales , Ratones , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Glioblastoma/genética , Glioblastoma/patología , Transducción de Señal , Procesos Neoplásicos , Regulación hacia Arriba , Línea Celular Tumoral , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proliferación Celular
10.
J Neurosci ; 43(25): 4559-4579, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37225434

RESUMEN

Previous studies have shown the essential roles of O-GlcNAc transferase (Ogt) and O-GlcNAcylation in neuronal development, function and neurologic diseases. However, the function of Ogt and O-GlcNAcylation in the adult cerebellum has not been well elucidated. Here, we have found that cerebellum has the highest level of O-GlcNAcylation relative to cortex and hippocampus of adult male mice. Specific deletion of Ogt in granule neuron precursors (GNPs) induces abnormal morphology and decreased size of the cerebellum in adult male Ogt deficient [conditional knock-out (cKO)] mice. Adult male cKO mice show the reduced density and aberrant distribution of cerebellar granule cells (CGCs), the disrupted arrangement of Bergman glia (BG) and Purkinje cells. In addition, adult male cKO mice exhibit aberrant synaptic connection, impaired motor coordination, and learning and memory abilities. Mechanistically, we have identified G-protein subunit α12 (Gα12) is modified by Ogt-mediated O-GlcNAcylation. O-GlcNAcylation of Gα12 facilitates its binding to Rho guanine nucleotide exchange factor 12 (Arhgef12) and consequently activates RhoA/ROCK signaling. RhoA/ROCK pathway activator LPA can rescue the developmental deficits of Ogt deficient CGCs. Therefore, our study has revealed the critical function and related mechanisms of Ogt and O-GlcNAcylation in the cerebellum of adult male mice.SIGNIFICANCE STATEMENT Cerebellar function are regulated by diverse mechanisms. To unveil novel mechanisms is critical for understanding the cerebellar function and the clinical therapy of cerebellum-related diseases. In the present study, we have shown that O-GlcNAc transferase gene (Ogt) deletion induces abnormal cerebellar morphology, synaptic connection, and behavioral deficits of adult male mice. Mechanistically, Ogt catalyzes O-GlcNAcylation of Gα12, which promotes the binding to Arhgef12, and regulates RhoA/ROCK signaling pathway. Our study has uncovered the important roles of Ogt and O-GlcNAcylation in regulating cerebellar function and cerebellum-related behavior. Our results suggest that Ogt and O-GlcNAcylation could be potential targets for some cerebellum-related diseases.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Transducción de Señal , Ratones , Masculino , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , N-Acetilglucosaminiltransferasas/genética , Ratones Noqueados
11.
s.l; CONETEC; 22 mayo 2023.
No convencional en Español | BRISA/RedTESA | ID: biblio-1444580

RESUMEN

INTRODUCCIÓN: El cáncer colorrectal (CCR) representa la segunda causa de mortalidad por tumores en la Argentina. Según la Agencia Internacional de Investigación sobre el Cáncer la incidencia en nuestro país durante el año 2020 se calculó en aproximadamente 15.600 casos, registrándose en el mismo período de tiempo unas 8.600 muertes. que otros pacientes diagnosticados en estadios iniciales progresarán en el transcurso de la enfermedad requiriendo quimioterapia sistémica. Sin tratamiento, el promedio de sobrevida de estos pacientes es de unos 6 meses. Los esquemas de quimioterapia utilizados frecuentemente están basados en la combinación de fluoropirimidinas con oxaliplatino o irinotecan asociados con anticuerpos monoclonales contra el factor de crecimiento endotelial vascular (bevacizumab) o el receptor del factor de crecimiento epidérmico (cetuximab, panitunumab) en tumores sin mutaciones en la vía RAS (wildtipe o "salvajes"). En aquellos tumores con deficiencia del sistema de reparación de apareamientos erróneos o alta inestabilidad de microsatélites (por sus siglas en inglés, dMMR o MSI-H, respectivamente) se listan dentro de las opciones terapéuticas el uso de inmunoterapia. En aquellos pacientes en los cuales la enfermedad progresa a pesar de los esquemas mencionados se pueden utilizar regorafenib o trifluridina/tipiracilo y en ciertos subtipos moleculares encorafenib (mutación V600E en el gen BRAF); trastuzumab +/-pertuzumab/lapatinib/tucatinib (HER2 amplificado y sin mutaciones en los genes RAS y BRAF), entre otras. Se debe tener en cuenta que alguna de estas opciones no cuentan con aprobación o no se encuentran disponibles aún en nuestro país. En este documento se plantea evaluar la eficacia y seguridad del uso de adagrasib en pacientes con carcinoma colorectal metastásico portadores de la mutación G12C en el gen KRAS. TECNOLOGÍA: Adagrasib (Krasati®) es un inhibidor irreversible y selectivo de la proteína mutante KRAS G12C (homólogo del oncogén vírico de sarcoma de rata Kirsten). La proteína pertenece a la subfamilia de proteínas RAS (KRAS, HRAS y NRAS) que actúan como GTPasas y se desempeñan como reguladores moleculares, controlando un amplio espectro de actividades celulares, como la proliferación y la sobrevida celular. Su inactivación por parte de adagrasib bloquea la transmisión de señales inhibiendo el crecimiento celular y favoreciendo la apoptosis de manera selectiva en tumores portadores de la mutación KRAS G12C. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo de adagrasib en pacientes con diagnóstico de carcinoma colorectal metastásico. MÉTODOS Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. La fecha de búsqueda de información fue hasta el 19 de mayo de 2023. Para la búsqueda en Pubmed se utilizó la siguiente estrategia de búsqueda: (adagrasib [Supplementary Concept] OR adagrasib [tiab] OR MRTX849 [tiab]) AND ("Colonic Neoplasms"[Mesh] OR ¨Colorectal Cancer¨ [tiab]). CONCLUSIONES: La evidencia sobre el uso de adagrasib para el tratamiento de pacientes con diagnóstico de carcinoma colorectal metastásico progresados a múltiples líneas de tratamiento se basa en un único ensayo clínico no aleatorizado de fase I-II. Este estudio mostró que aquellos pacientes, progresados generalmente a tres líneas de tratamiento y con tumores que presentan mutaciones G12C en el gen KRAS, que utilizaron adagrasib con cetuximab reportaron una mediana de sobrevida global de 13,4 meses y libre de progresión de 6,9 meses, y una tasa de respuesta del 46%. Los eventos adversos severos fueron reportados en el 16% de los pacientes. Actualmente se encuentra en curso un ensayo clínico aleatorizado de fase III que cuenta con centros en Argentina y tratará de establecer la seguridad y eficacia de la adición de adagrasib a un esquema de poliquimioterapia como segunda línea de tratamiento La Administración de Alimentos y Medicamentos de Estados Unidos y la Agencia Europea de Medicamentos aún no ha autorizado la comercialización del medicamento en la indicación antes mencionada. En referencias a las recomendaciones relevadas, no se hallaron guías que mencionen los esquemas de adagrasib combinado con cetuximab o en monoterapia como una opción para el tratamiento del carcinoma colorectal avanzado. Utilizando precios de referencia internacionales, el costo de adquisición para un ciclo de tratamiento fue estimado en aproximadamente 4,8 millones de pesos argentinos.


Asunto(s)
Humanos , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas ras/uso terapéutico , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Metástasis de la Neoplasia/tratamiento farmacológico , Argentina , Eficacia , Análisis Costo-Beneficio
12.
J Alzheimers Dis ; 93(2): 545-560, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37038813

RESUMEN

BACKGROUND: Our previous studies indicated that anesthesia and surgery could aggravate cognitive impairment of 5XFAD transgenic (Tg) mice, and this aggravation was associated with tau hyperphosphorylation. We previously identified that GNA13 (the gene encoding Gα13) was a hub gene with tau hyperphosphorylation. OBJECTIVE: This study aims to further investigate the mechanism that whether the Gα13-mediated signaling pathway acts as an instigator to regulate cofilin activation and autophagy impairment in this process. METHODS: 5XFAD Tg mice and their littermate (LM) mice were randomly allocated into four groups: LM Control group, LM Anesthesia/Surgery group, AD Control group, and AD Anesthesia/Surgery group. For mice in the Anesthesia/Surgery groups, abdominal surgery was performed under 1.4% isoflurane anesthesia followed by sustaining anesthetic inhalation for up to 2 h. RESULTS: Compared with the AD Control group, protein levels of Gα13, ROCK2, LPAR5, and p-tau/tau46 ratio were increased, while p-cofilin/cofilin protein expression ratio was decreased in the AD Anesthesia/Surgery group. However, the differences in these protein levels were not significant among LM groups. CONCLUSION: This study demonstrated that anesthesia and surgery might exacerbate p-tau accumulation in 5XFAD Tg mice but not in LM mice. And this might be closely related to cofilin activation via Gα13-mediated signaling cascade.


Asunto(s)
Enfermedad de Alzheimer , Anestesia , Ratones , Animales , Ratones Transgénicos , Enfermedad de Alzheimer/patología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Modelos Animales de Enfermedad
13.
Hypertension ; 80(2): 403-415, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36448462

RESUMEN

BACKGROUND: Postsynaptic density 95/disk-large/ZO-1 Rho guanine nucleotide exchange factor (PDZ-RhoGEF, PRG) functions as a RhoGEF for activated Gα13 and transmits activation signals to downstream signaling pathways in various pathological processes. Although the prohypertrophic effect of activated Gα13 (guanine nucleotide binding protein alpha 13; a heterotrimeric G protein) is well-established, the role of PDZ-RhoGEF in pathological cardiac hypertrophy is still obscure. METHODS: Genetically engineered mice and neonatal rat ventricular myocytes were generated to investigate the function of PRG in pathological myocardial hypertrophy. The prohypertrophic stimuli-induced alternations in the morphology and intracellular signaling were measured in myocardium and neonatal rat ventricular myocytes. Furthermore, multiple molecular methodologies were used to identify the precise molecular mechanisms underlying PDZ-RhoGEF function. RESULTS: Increased PDZ-RhoGEF expression was documented in both hypertrophied hearts and neonatal rat ventricular myocytes. Upon prohypertrophic stimuli, the PDZ-RhoGEF-deficient hearts displayed alleviated cardiomyocyte enlargement and attenuated collagen deposition with improved cardiac function, whereas the adverse hypertrophic responses in hearts and neonatal rat ventricular myocytes were markedly exaggerated by PDZ-RhoGEF overexpression. Mechanistically, RhoA (ras homolog family member A)-dependent signaling pathways may function as the downstream effectors of PDZ-RhoGEF in hypertrophic remodeling, as confirmed by rescue experiments using a RhoA inhibitor and dominant-negative RhoA. Furthermore, PDZ-RhoGEF is associated with activated Gα13 and contributes to Gα13-mediated activation of RhoA-dependent signaling. CONCLUSIONS: Our data provide the first evidence that PDZ-RhoGEF promotes pathological cardiac hypertrophy by linking activated Gα13 to RhoA-dependent signaling pathways. Therefore, PDZ-RhoGEF has the potential to be a diagnostic marker or therapeutic target for pathological cardiac hypertrophy.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Transducción de Señal , Animales , Ratones , Ratas , Cardiomegalia , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Proteína de Unión al GTP rhoA/metabolismo , Dominios PDZ
14.
Cell Signal ; 102: 110534, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36442589

RESUMEN

Gα13, a heterotrimeric G protein α subunit of the G12/13 subfamily, is an oncogenic driver in multiple cancer types. Unlike other G protein subfamilies that contribute to cancer progression via amino acid substitutions that abolish their deactivating, intrinsic GTPase activity, Gα13 rarely harbors such mutations in tumors and instead appears to stimulate aberrant cell growth via overexpression as a wildtype form. It is not known why this effect is exclusive to the G12/13 subfamily, nor has a mechanism been elucidated for overexpressed Gα13 promoting tumor progression. Using a reporter gene assay for serum response factor (SRF)-mediated transcription in HEK293 cells, we found that transiently expressed, wildtype Gα13 generates a robust SRF signal, approximately half the amplitude observed for GTPase-defective Gα13. When epitope-tagged, wildtype Gα13 was titrated upward in cells, a sharp increase in SRF stimulation was observed coincident with a "spillover" of Gα13 from membrane-associated to a soluble fraction. Overexpressing G protein ß and γ subunits caused both a decrease in this signal and a shift of wildtype Gα13 back to the membranous fraction, suggesting that stoichiometric imbalance in the αßγ heterotrimer results in aberrant subcellular localization and signalling by overexpressed Gα13. We also examined the acylation requirements of wildtype Gα13 for signalling to SRF. Similar to GTPase-defective Gα13, S-palmitoylation of the wildtype α subunit was necessary for SRF activation but could be replaced functionally by an engineered site for N-terminal myristoylation. However, a key difference was observed between wildtype and GTPase-defective Gα13: whereas the latter protein lacking palmitoylation sites was rescued in its SRF signalling by either an engineered polybasic sequence or a C-terminal isoprenylation site, these motifs failed to restore signalling by wildtype, non-palmitoylated Gα13. These findings illuminate several components of the mechanism in which overexpressed, wildtype Gα13 contributes to growth and tumorigenic signalling, and reveal greater stringency in its requirements for post-translational modification in comparison to GTPase-defective Gα13.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Neoplasias , Humanos , Citoplasma/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Factor de Respuesta Sérica/metabolismo
15.
Mediators Inflamm ; 2022: 1818758, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36248188

RESUMEN

Lysophosphatidic acid (LPA) has disruptive effects on lumbar spinal stenosis (LSS). Recently, LPA has been reported to be involved in spinal cord neuronal injury and toxicity, promoting the pathogenesis of LSS. However, the exact effects of LPA on spinal cord neurons remain unknown. The purpose of this study is to investigate the effects of LPA (18 : 1) on spinal cord neuronal cytotoxicity, apoptosis, DNA damage, and oxidative stress. After clinical detection of LPA secretion, spinal cord neurons were treated with LPA (18 : 1); cell viability was analyzed by MTT assay, and LDH leakage was detected by LDH kit; cell apoptosis was detected by flow cytometry; ROS production was measured by DCFDA staining and MitoSOX Red Staining; the activation of the Gα12/Gα13 signaling pathway was detected by serum response factor response element (SRF-RE) luciferase reporter gene; the relationship among LPA, LPA4/6, and ROCK was examined by western blotting. In spinal cord neurons treated with LPA (18 : 1), cellular activity decreased and LDH release increased. The Rho kinase inhibitor (Y-27632) can attenuate LPA-induced apoptosis, DNA damage, and oxidative stress in spinal cord neurons. Moreover mechanistic investigation indicated that LPA (18 : 1) activates Gα12/13-Rho-ROCK2-induced apoptosis, DNA damage, and oxidative stress in spinal cord neurons by upregulating LPA4/LPA6 receptors. Further, the Rho kinase inhibitor Y-27632 attenuates the effects of LPA by downregulating LPA4/LPA6 receptors. Taken together, the possible mechanism by which LPA secretion in LSS patients aggravates patient injury was further elucidated using an LPA-induced spinal cord neuronal injury cell model in vitro.


Asunto(s)
Receptores del Ácido Lisofosfatídico , Traumatismos de la Médula Espinal , Amidas , Apoptosis , Daño del ADN , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/farmacología , Humanos , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Neuronas/metabolismo , Estrés Oxidativo , Piridinas , Especies Reactivas de Oxígeno/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores Purinérgicos P2/metabolismo , Factor de Respuesta Sérica/metabolismo , Factor de Respuesta Sérica/farmacología , Médula Espinal/patología , Traumatismos de la Médula Espinal/metabolismo , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/farmacología
16.
Cells ; 11(16)2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-36010639

RESUMEN

Despite fully functional primary hemostasis, platelets of healthy neonates exhibit hypoaggregability and secretion defects, which may be adaptations to specific requirements in this developmental stage. The etiologies for reduced signal transduction vary with the type of agonist. The discovered peculiarities are lower receptor densities, reduced calcium mobilization, and functional impairments of G proteins. Reduced secretion of dense granules has been attributed to lower numbers of granules. Signaling studies with adult platelets have shown a regulating effect of the G12/13 signaling pathway on dense granule secretion via RhoA. We comparatively analyzed secretion profiles using flow cytometry and expression levels of Gq, Gi, and G12/13 using Western blot analysis in platelets from cord blood and adults. Furthermore, we evaluated Rho activation after in vitro platelet stimulation with thrombin using a pulldown assay. We observed a markedly reduced expression of the dense granule marker CD63 on neonatal platelets after thrombin stimulation. Gα12/13 expression was significantly decreased in neonatal platelets and correlated with lower Rho activation after thrombin stimulation. We conclude that lower expression of G12/13 in neonatal platelets results in attenuated activation of Rho and may contribute to reduced secretion of dense granules after exposure to thrombin.


Asunto(s)
Plaquetas , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Trombina , Plaquetas/metabolismo , Proteínas de Unión al GTP/metabolismo , Hemostasis , Humanos , Recién Nacido , Transducción de Señal , Trombina/farmacología
17.
J Biol Chem ; 298(9): 102294, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35872018

RESUMEN

Promiscuous G protein-coupled receptors (GPCRs) engage multiple Gα subtypes with different efficacies to propagate signals in cells. A mechanistic understanding of Gα selectivity by GPCRs is critical for therapeutic design, since signaling can be restrained by ligand-receptor complexes to preferentially engage specific G proteins. However, details of GPCR selectivity are unresolved. Here, we investigated cognate G protein selectivity using the prototypical promiscuous Gαq/11 and Gα12/13 coupling receptors, angiotensin II type I receptor (AT1R) and prostaglandin F2α receptor (FP), bioluminescence resonance energy transfer-based G protein and pathway-selective sensors, and G protein knockout cells. We determined that competition between G proteins for receptor binding occurred in a receptor- and G protein-specific manner for AT1R and FP but not for other receptors tested. In addition, we show that while Gα12/13 competes with Gαq/11 for AT1R coupling, the opposite occurs for FP, and Gαq-mediated signaling regulated G protein coupling only at AT1R. In cells, the functional modulation of biased ligands at FP and AT1R was contingent upon cognate Gα availability. The efficacy of AT1R-biased ligands, which poorly signal through Gαq/11, increased in the absence of Gα12/13. Finally, we show that a positive allosteric modulator of Gαq/11 signaling that also allosterically decreases FP-Gα12/13 coupling, lost its negative modulation in the absence of Gαq/11 coupling to FP. Together, our findings suggest that despite preferential binding of similar subsets of G proteins, GPCRs follow distinct selectivity rules, which may contribute to the regulation of ligand-mediated G protein bias of AT1R and FP.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Receptor de Angiotensina Tipo 1 , Receptores de Prostaglandina , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Ligandos , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Prostaglandina/metabolismo
18.
Biochem Pharmacol ; 201: 115069, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35525325

RESUMEN

We could previously show that thromboxane A2 receptor (TP) activation inhibits the angiogenic capacity of human endothelial cells, but the underlying mechanisms remained unclear. Therefore, the aim of this study was to elucidate TP signal transduction pathways relevant to angiogenic sprouting of human endothelial cells. To clarify this matter, we used RNAi-mediated gene silencing as well as pharmacological inhibition of potential TP downstream targets in human umbilical vein endothelial cells (HUVEC) and VEGF-induced angiogenic sprouting of HUVEC spheroids in vitro as a functional read-out. In this experimental set-up, the TP agonist U-46619 completely blocked VEGF-induced angiogenic sprouting of HUVEC spheroids. Moreover, in live-cell analyses TP activation induced endothelial cell contraction, sprout retraction as well as endothelial cell tension and focal adhesion dysregulation of HUVEC. These effects were reversed by pharmacological TP inhibition or TP knockdown. Moreover, we identified a TP-Gα13-RhoA/C-ROCK-LIMK2-dependent signal transduction pathway to be relevant for U-46619-induced inhibition of VEGF-mediated HUVEC sprouting. In line with these results, U-46619-mediated TP activation potently induced RhoA and RhoC activity in live HUVEC as measured by FRET biosensors. Interestingly, pharmacological inhibition of ROCK and LIMK2 also normalized U-46619-induced endothelial cell tension and focal adhesion dysregulation of HUVEC. In summary, our work reveals mechanisms by which the TP may disturb angiogenic endothelial function in disease states associated with sustained endothelial TP activation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Células Endoteliales de la Vena Umbilical Humana , Quinasas Lim , Receptores de Tromboxano A2 y Prostaglandina H2 , Proteína de Unión al GTP rhoA , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Quinasas Lim/metabolismo , Neovascularización Fisiológica , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Proteína rhoC de Unión a GTP
19.
Mol Psychiatry ; 27(5): 2425-2438, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35393556

RESUMEN

Latrophilin-3 (Lphn3; also known as ADGRL3) is a member of the adhesion G Protein Coupled Receptor subfamily, which participates in the stabilization and maintenance of neuronal networks by mediating intercellular adhesion through heterophilic interactions with transmembrane ligands. Polymorphisms modifying the Lphn3 gene are associated with attention-deficit/hyperactivity disorder (ADHD) in children and its persistence into adulthood. How these genetic alterations affect receptor function remains unknown. Here, we conducted the functional validation of distinct ADHD-related Lphn3 variants bearing mutations in the receptor's adhesion motif-containing extracellular region. We found that all variants tested disrupted the ability of Lphn3 to stabilize intercellular adhesion in a manner that was distinct between ligands classes, but which did not depend on ligand-receptor interaction parameters, thus pointing to altered intrinsic receptor signaling properties. Using G protein signaling biosensors, we determined that Lphn3 couples to Gαi1, Gαi2, Gαs, Gαq, and Gα13. However, all ADHD-related receptor variants consistently lacked intrinsic as well as ligand-dependent Gα13 coupling efficiency while maintaining unaltered coupling to Gαi, Gαs, and Gαq. Consistent with these alterations, actin remodeling functions as well as actin-relevant RhoA signaling normally displayed by the constitutively active Lphn3 receptor were impeded by select receptor variants, thus supporting additional signaling defects. Taken together, our data point to Gα13 selective signaling impairments as representing a disease-relevant pathogenicity pathway that can be inherited through Lphn3 gene polymorphisms. This study highlights the intricate interplay between Lphn3 GPCR functions and the actin cytoskeleton in modulating neurodevelopmental cues related to ADHD etiology.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Actinas , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Niño , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Humanos , Ligandos , Receptores Acoplados a Proteínas G/genética , Virulencia
20.
Tissue Cell ; 76: 101795, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35413491

RESUMEN

The purpose of this study aimed to figure out the role of GNA13 in lung squamous cell carcinoma (LUSC) and the underlying mechanism. Male BALB/c mice were used to construct LUSC mouse model. Cell growth was examined using MTT and colony formation assay. Cell migration and invasion was determined using wound healing and transwell assay. The expression and phosphorylation of protein was detected by Western blotting assay. Immunohistochemistry staining was used to observe the tumor growth and metastasis. GNA13 was overexpressed in both LUSC tissues and LUSC cell lines. Knockdown of GNA13 in LUSC cells reduced cell viability and inhibited the formation of colonies in the SK-MES-1 and NCI-H520 cells. Cell migration and invasion was also prevented by inhibition of GNA13 in the LUSC cells. Phosphorylation of PI3K and AKT was downregulated by silencing GNA13 and upregulated by overexpression of GNA13 in the LUSC cells. In LUSC mouse model, tumor size and tumor weight were significantly decreased in si-GNA13 mice compared to control group. The expression of GNA13, Ki67, MMP2 and phosphorylation of AKT were significantly inhibited in si-GNA13 mice compared to control group. This study has demonstrated that knockdown of GNA13 could inhibit cell survival, migration and metastasis in LUSC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Neoplasias Pulmonares , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA