Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
s.l; CONETEC; 22 mayo 2023.
Non-conventional Es | BRISA | ID: biblio-1444580

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.


Humans , Colorectal Neoplasms/drug therapy , ras Proteins/therapeutic use , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , Neoplasm Metastasis/drug therapy , Argentina , Efficacy , Cost-Benefit Analysis
2.
Nat Commun ; 12(1): 3185, 2021 05 27.
Article En | MEDLINE | ID: mdl-34045461

Systemic inflammation as manifested in sepsis is an excessive, life-threatening inflammatory response to severe bacterial or viral infection or extensive injury. It is also a thrombo-inflammatory condition associated with vascular leakage/hemorrhage and thrombosis that is not effectively treated by current anti-inflammatory or anti-thrombotic drugs. Here, we show that MB2mP6 peptide nanoparticles, targeting the Gα13-mediated integrin "outside-in" signaling in leukocytes and platelets, inhibited both inflammation and thrombosis without causing hemorrhage/vascular leakage. MB2mP6 improved mouse survival when infused immediately or hours after onset of severe sepsis. Furthermore, platelet Gα13 knockout inhibited septic thrombosis whereas leukocyte Gα13 knockout diminished septic inflammation, each moderately improving survival. Dual platelet/leukocyte Gα13 knockout inhibited septic thrombosis and inflammation, further improving survival similar to MB2mP6. These results demonstrate that inflammation and thrombosis independently contribute to poor outcomes and exacerbate each other in systemic inflammation, and reveal a concept of dual anti-inflammatory/anti-thrombotic therapy without exacerbating vascular leakage.


CD18 Antigens/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , Peptide Fragments/pharmacology , Sepsis/drug therapy , Thrombosis/drug therapy , Animals , Anti-Inflammatory Agents , Blood Platelets/drug effects , Blood Platelets/immunology , Blood Platelets/metabolism , CD18 Antigens/metabolism , Chlorides/administration & dosage , Chlorides/toxicity , Disease Models, Animal , Ferric Compounds/administration & dosage , Ferric Compounds/toxicity , Fibrinolytic Agents , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Humans , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Macrophages , Mice , Mice, Knockout , Nanoparticles/therapeutic use , Peptide Fragments/therapeutic use , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Primary Cell Culture , Protein Binding/drug effects , Sepsis/blood , Sepsis/complications , Sepsis/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , THP-1 Cells , Thrombosis/blood , Thrombosis/chemically induced
3.
Circulation ; 138(21): 2395-2412, 2018 11 20.
Article En | MEDLINE | ID: mdl-29991487

BACKGROUND: Although the role of thrombin in atherothrombosis is well studied, its role in the pathogenesis of diet-induced atherosclerosis is not known. METHODS: Using a mouse model of diet-induced atherosclerosis and molecular biological approaches, here we have explored the role of thrombin and its G protein-coupled receptor signaling in diet-induced atherosclerosis. RESULTS: In exploring the role of G protein-coupled receptor signaling in atherogenesis, we found that thrombin triggers foam cell formation via inducing CD36 expression, and these events require Par1-mediated Gα12-Pyk2-Gab1-protein kinase C (PKC)θ-dependent ATF2 activation. Genetic deletion of PKCθ in apolipoprotein E (ApoE)-/- mice reduced Western diet-induced plaque formation. Furthermore, thrombin induced Pyk2, Gab1, PKCθ, and ATF2 phosphorylation, CD36 expression, and foam cell formation in peritoneal macrophages of ApoE-/- mice. In contrast, thrombin only stimulated Pyk2 and Gab1 but not ATF2 phosphorylation or its target gene CD36 expression in the peritoneal macrophages of ApoE-/-:PKCθ-/- mice, and it had no effect on foam cell formation. In addition, the aortic root cross-sections of Western diet-fed ApoE-/- mice showed increased Pyk2, Gab1, PKCθ, and ATF2 phosphorylation and CD36 expression as compared with ApoE-/-:PKCθ-/- mice. Furthermore, although the monocytes from peripheral blood and the aorta of Western diet-fed ApoE-/- mice were found to contain more of Ly6Chi cells than Ly6Clo cells, the monocytes from Western diet-fed ApoE-/-:PKCθ-/- mice were found to contain more Ly6Clo cells than Ly6Chi cells. It is interesting to note that the Ly6Chi cells showed higher CD36 expression with enhanced capacity to form foam cells as compared with Ly6Clo cells. CONCLUSIONS: These findings reveal for the first time that thrombin-mediated Par1-Gα12 signaling via targeting Pyk2-Gab1-PKCθ-ATF2-dependent CD36 expression might be playing a crucial role in diet-induced atherogenesis.


Activating Transcription Factor 2/metabolism , Atherosclerosis/pathology , CD36 Antigens/metabolism , Protein Kinase C-theta/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/veterinary , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/genetics , Cell Differentiation/drug effects , Foam Cells/cytology , Foam Cells/metabolism , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Protein Kinase C-theta/deficiency , Protein Kinase C-theta/genetics , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Thrombin/pharmacology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
4.
J Hepatol ; 68(3): 493-504, 2018 03.
Article En | MEDLINE | ID: mdl-29080810

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) have a role in liver fibrosis. Guanine nucleotide-binding α-subunit 12 (Gα12) converges signals from G-protein-coupled receptors whose ligand levels are elevated in the environment during liver fibrosis; however, information is lacking on the effect of Gα12 on HSC trans-differentiation. This study investigated the expression of Gα12 in HSCs and the molecular basis of the effects of its expression on liver fibrosis. METHODS: Gα12 expression was assessed by immunostaining, and immunoblot analyses of mouse fibrotic liver tissues and primary HSCs. The role of Gα12 in liver fibrosis was estimated using a toxicant injury mouse model with Gα12 gene knockout and/or HSC-specific Gα12 delivery using lentiviral vectors, in addition to primary HSCs and LX-2 cells using microRNA (miR) inhibitors, overexpression vectors, or adenoviruses. miR-16, Gα12, and LC3 were also examined in samples from patients with fibrosis. RESULTS: Gα12 was overexpressed in activated HSCs and fibrotic liver, and was colocalised with desmin. In a carbon tetrachloride-induced fibrosis mouse model, Gα12 ablation prevented increases in fibrosis and liver injury. This effect was attenuated by HSC-specific lentiviral delivery of Gα12. Moreover, Gα12 activation promoted autophagy accompanying c-Jun N-terminal kinase-dependent ATG12-5 conjugation. In addition, miR-16 was found to be a direct inhibitor of the de novo synthesis of Gα12. Modulations of miR-16 altered autophagy in HSCs. In a fibrosis animal model or patients with severe fibrosis, miR-16 levels were lower than in their corresponding controls. Consistently, cirrhotic patient liver tissues showed Gα12 and LC3 upregulation in desmin-positive areas. CONCLUSIONS: miR-16 dysregulation in HSCs results in Gα12 overexpression, which activates HSCs by facilitating autophagy through ATG12-5 formation. This suggests that Gα12 and its regulatory molecules could serve as targets for the amelioration of liver fibrosis. LAY SUMMARY: Guanine nucleotide-binding α-subunit 12 (Gα12) is upregulated in activated hepatic stellate cells (HSCs) as a consequence of the dysregulation of a specific microRNA that is abundant in HSCs, facilitating the progression of liver fibrosis. This event is mediated by c-Jun N-terminal kinase-dependent ATG12-5 formation and the promotion of autophagy. We suggest that Gα12 and its associated regulators could serve as new targets in HSCs for the treatment of liver fibrosis.


GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis , MicroRNAs/metabolism , Animals , Autophagy/drug effects , Cell Proliferation/drug effects , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , Gene Expression Regulation , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Serine Proteinase Inhibitors/pharmacology , Signal Transduction/drug effects , Up-Regulation
5.
Oncotarget ; 5(20): 9626-40, 2014 Oct 30.
Article En | MEDLINE | ID: mdl-25275299

Oral squamous cell carcinoma (OSCC) has a propensity to spread to the cervical lymph nodes (LN). The presence of cervical LN metastases severely impacts patient survival, whereby the two-year survival for oral cancer patients with involved LN is ~30% compared to over 80% in patients with non-involved LN. Elucidation of key molecular mechanisms underlying OSCC metastasis may afford an opportunity to target specific genes, to prevent the spread of OSCC and to improve patient survival. In this study, we demonstrated that expression of the heterotrimeric G-protein alpha-12 (Gα12) is highly up-regulated in primary tumors and LN of OSCC patients, as assessed by quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC). We also found that exogenous expression of the constitutively activated-form of Gα12 promoted cell migration and invasion in OSCC cell lines. Correspondingly, inhibition of Gα12 expression by shRNA consistently inhibited OSCC cell migration and invasion in vitro. Further, the inhibition of G12 signaling by regulator of G-protein signaling (RGS) inhibited Gα12-mediated RhoA activation, which in turn resulted in reduced LN metastases in a tongue-orthotopic xenograft mouse model of oral cancer. This study provides a rationale for future development and evaluation of drug candidates targeting Gα12-related pathways for metastasis prevention.


Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/genetics , Cell Movement/physiology , Female , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Lymphatic Metastasis , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mouth Neoplasms/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Transcriptional Activation , Up-Regulation , Xenograft Model Antitumor Assays
6.
Am J Physiol Cell Physiol ; 300(5): C1181-92, 2011 May.
Article En | MEDLINE | ID: mdl-21289285

Enhanced vascular arginase activity impairs endothelium-dependent vasorelaxation by decreasing l-arginine availability to endothelial nitric oxide (NO) synthase, thereby reducing NO production. Elevated angiotensin II (ANG II) is a key component of endothelial dysfunction in many cardiovascular diseases and has been linked to elevated arginase activity. We determined signaling mechanisms by which ANG II increases endothelial arginase function. Results show that ANG II (0.1 µM, 24 h) elevates arginase activity and arginase I expression in bovine aortic endothelial cells (BAECs) and decreases NO production. These effects are prevented by the arginase inhibitor BEC (100 µM). Blockade of ANG II AT(1) receptors or transfection with small interfering RNA (siRNA) for Gα12 and Gα13 also prevents ANG II-induced elevation of arginase activity, but siRNA for Gαq does not. ANG II also elevates active RhoA levels and induces phosphorylation of p38 MAPK. Inhibitors of RhoA activation (simvastatin, 0.1 µM) or Rho kinase (ROCK) (Y-27632, 10 µM; H1152, 0.5 µM) block both ANG II-induced elevation of arginase activity and phosphorylation of p38 MAPK. Furthermore, pretreatment of BAECs with p38 inhibitor SB-202190 (2 µM) or transfection with p38 MAPK siRNA prevents ANG II-induced increased arginase activity/expression and maintains NO production. Additionally, inhibitors of p38 MAPK (SB-203580, 5 µg·kg(-1)·day(-1)) or arginase (ABH, 8 mg·kg(-1)·day(-1)) or arginase gene knockout in mice prevents ANG II-induced vascular endothelial dysfunction and associated enhancement of arginase. These results indicate that ANG II increases endothelial arginase activity/expression through Gα12/13 G proteins coupled to AT(1) receptors and subsequent activation of RhoA/ROCK/p38 MAPK pathways leading to endothelial dysfunction.


Angiotensin II/physiology , Arginase/physiology , Endothelium, Vascular/physiopathology , p38 Mitogen-Activated Protein Kinases/physiology , rho-Associated Kinases/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Arginase/antagonists & inhibitors , Boronic Acids/pharmacology , Cattle , Cell Line , Endothelial Cells , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Imidazoles/pharmacology , Mice , Phosphorylation , Pyridines/pharmacology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Simvastatin/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors
7.
Carcinogenesis ; 31(7): 1230-7, 2010 Jul.
Article En | MEDLINE | ID: mdl-20478922

Bortezomib is a proteasome inhibitor approved for anticancer therapy. However, variable sensitivity of tumor cells exists in this therapy probably due to differences in the expression of proteasome subunits. G(alpha)(12/13) serves modulators or signal transducers in diverse pathways. This study investigated whether cancer cells display differential sensitivity to bortezomib with reference to G(alpha)(12/13) expression, and if so, whether G(alpha)(12/13) affects the expression of proteasome subunits and their activities. Bortezomib treatment exhibited greater sensitivities in Huh7 and SNU886 cells (epithelial type) than SK-Hep1 and SNU449 cells (mesenchymal type) that exhibited higher levels of G(alpha)(12/13). Overexpression of an active mutant of G(alpha)(12) (Galpha(12)QL) or G(alpha)(13) (G(alpha)(13)QL) diminished the ability of bortezomib to induce cytotoxicity in Huh7 cells. Moreover, transfection with the minigene that disturbs G protein-coupled receptor-G protein coupling (CT12 or CT13) increased it in SK-Hep1 cells. Consistently, MiaPaCa2 cells transfected with CT12 or CT13 exhibited a greater sensitivity to bortezomib. Evidence of G(alpha)(12/13)'s antagonism on the anticancer effect of bortezomib was verified in the reversal by G(alpha)(12)QL or G(alpha)(13)QL of the minigenes' enhancement of cytotoxity. Real-time polymerase chain reaction assay enabled us to identify PSMB5, multicatalytic endopeptidase complex-like-1, and proteasome activator subunit-1 repression by CT12 or CT13. Furthermore, G(alpha)(12/13) inhibition enhanced the ability of bortezomib to repress PSMB5, as shown by immunoblotting and proteasome activity assay. Moreover, this inhibitory effect on PSMB5 was attenuated by G(alpha)G(alpha)(12)QL or G(alpha)(13)QL. In conclusion, the inhibition of G(alpha)(12/13) activities may enhance the anticancer effect of bortezomib through PSMB5 repression, providing insight into the G(alpha)(12/13) pathway for the regulation of proteasomal activity.


Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/physiology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Bortezomib , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , Humans , In Situ Nick-End Labeling , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Proteasome Endopeptidase Complex/genetics , RNA, Messenger/analysis
8.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G641-54, 2009 Oct.
Article En | MEDLINE | ID: mdl-19679818

P2Y5 is a G protein-coupled receptor that binds and is activated by lysophosphatidic acid (LPA). We determined that P2Y5 transcript is expressed along the intestinal mucosa and investigated the intracellular pathways induced by P2Y5 activation, which could contribute to LPA effects on intestinal cell adhesion. P2Y5 heterologously expressed in CHO and small intestinal hBRIE 380i cells was activated by LPA resulting in an increase in intracellular calcium ([Ca(2+)](i)) when the cells concurrently expressed G(alpha)(Delta6qi5myr). P2Y5 activation also increased the phosphorylation of ERK1/2 that was sensitive to pertussis toxin. Together these indicate that P2Y5 activation by LPA induces an increase in [Ca(2+)](i) and ERK1/2 phosphorylation through G(alpha)(i). We discovered that P2Y5 was activated by farnesyl pyrophosphate (FPP) without a detectable change in [Ca(2+)](i). The activation of P2Y5 by LPA or FPP induced the activity of a serum response element (SRE)-linked luciferase reporter that was inhibited by the RGS domain of p115RhoGEF, C3 exotoxin, and Y-27632, suggesting the involvement of G(alpha)(12/13), Rho GTPase, and ROCK, respectively. However, only LPA-mediated induction of SRE reporter activity was sensitive to inhibitors targeting p38 MAPK, PI3K, PLC, and PKC. In addition, only LPA transactivated the epidermal growth factor receptor, leading to an induction of ERK1/2 phosphorylation. These observations correlate with our subsequent finding that P2Y5 activation by LPA, and not FPP, reduced intestinal cell adhesion. This study elucidates a mechanism whereby LPA can act as a luminal and/or serosal cue to alter mucosal integrity.


Cell Adhesion , Epithelial Cells/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Intestinal Mucosa/metabolism , Lysophospholipids/metabolism , Receptors, Purinergic P2/metabolism , Animals , CHO Cells , Calcium Signaling , Cell Adhesion/drug effects , Cricetinae , Cricetulus , Down-Regulation , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , ErbB Receptors/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Humans , Intestinal Mucosa/drug effects , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Polyisoprenyl Phosphates/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor Cross-Talk , Receptors, Purinergic P2/genetics , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , Serum Response Element , Sesquiterpenes/metabolism , Transfection , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
9.
FASEB J ; 22(8): 2821-31, 2008 Aug.
Article En | MEDLINE | ID: mdl-18367648

G alpha12 constitutes, along with G alpha13, one of the four families of alpha subunits of heterotrimeric G proteins. We found that the N terminus of G alpha12, but not those of other G alpha subunits, contains a predicted mitochondrial targeting sequence. Using confocal microscopy and cell fractionation, we demonstrated that up to 40% of endogenous G alpha12 in human umbilical vein endothelial cells colocalize with mitochondrial markers. N-terminal sequence of G alpha12 fused to GFP efficiently targeted the fusion protein to mitochondria. G alpha12 with mutated mitochondrial targeting sequence was still located in mitochondria, suggesting the existence of additional mechanisms for mitochondrial localization. Lysophosphatidic acid, one of the known stimuli transduced by G alpha12/13, inhibited mitochondrial motility, while depletion of endogenous G alpha12 increased mitochondrial motility. G alpha12Q229L variants uncoupled from RhoGEFs (but not fully functional activated G alpha12Q229L) induced transformation of the mitochondrial network into punctate mitochondria and resulted in a loss of mitochondrial membrane potential. All examined G alpha12Q229L variants reduced phosphorylation of Bcl-2 at Ser-70, while only mutants unable to bind RhoGEFs also decreased cellular levels of Bcl-2. These G alpha12 mutants were also more efficient Hsp90 interactors. These findings are the first demonstration of a heterotrimeric G protein alpha subunit specifically targeted to mitochondria and involved in the control of mitochondrial morphology and dynamics.


GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Mitochondria/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Guanine Nucleotide Exchange Factors/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial , Movement , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors
10.
J Cell Sci ; 121(Pt 6): 814-24, 2008 Mar 15.
Article En | MEDLINE | ID: mdl-18285450

The polarized functions of epithelia require an intact tight junction (TJ) to restrict paracellular movement and to separate membrane proteins into specific domains. TJs contain scaffolding, integral membrane and signaling proteins, but the mechanisms that regulate TJs and their assembly are not well defined. Galpha12 (GNA12) binds the TJ protein ZO-1 (TJP1), and Galpha12 activates Src to increase paracellular permeability via unknown mechanisms. Herein, we identify Src as a component of the TJ and find that recruitment of Hsp90 to activated Galpha12 is necessary for signaling. TJ integrity is disrupted by Galpha12-stimulated Src phosphorylation of ZO-1 and ZO-2 (TJP2); this phosphorylation leads to dissociation of occludin and claudin 1 from the ZO-1 protein complex. Inhibiting Hsp90 with geldanamycin blocks Galpha12-stimulated Src activation and phosphorylation, but does not affect protein levels or the Galpha12-ZO-1 interaction. Using the calcium-switch model of TJ assembly and GST-TPR (GST-fused TPR domain of PP5) pull-downs of activated Galpha12, we demonstrate that switching to normal calcium medium activates endogenous Galpha12 during TJ assembly. Thrombin increases permeability and delays TJ assembly by activating Galpha12, but not Galpha13, signaling pathways. These findings reveal an important role for Galpha12, Src and Hsp90 in regulating the TJ in established epithelia and during TJ assembly.


Epithelial Cells/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Tight Junctions/metabolism , Animals , Cell Line , Cell Membrane Permeability , Claudin-1 , Dogs , Epithelial Cells/enzymology , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mutation , Occludin , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , Tight Junctions/enzymology , Tyrosine/metabolism , Zonula Occludens-1 Protein , Zonula Occludens-2 Protein
11.
J Vasc Surg ; 46(6): 1253-1259, 2007 Dec.
Article En | MEDLINE | ID: mdl-18155002

BACKGROUND: Sphingosine-1-phosphate (S-1-P) is a bioactive sphingolipid that stimulates the migration of vascular smooth muscle cell (VSMC) through G-protein coupled receptors; it has been shown to activate reduced nicotinamide dinucleotide phosphate hydrogen (NAD[P]H) oxidase. The role of phospholipase C (PLC) in oxygen free radical generation, and the regulation of VSMC migration in response to S-1-P, are poorly understood. METHODS: Rat arterial VSMC were cultured in vitro. Oxygen free radical generation was measured by fluorescent redox indicator assays in response to S-1-P (0.1microM) in the presence and absence of the active PLC inhibitor (U73122; U7, 10nM) or its inactive analog U73343 (InactiveU7, 10nM). Activation of PLC was assessed by immunoprecipitation and Western blotting for the phosphorylated isozymes (beta and gamma). Small interfering (si) RNA to the G-proteins Galphai, Galphaq, and Galpha12/13 was used to downregulate specific proteins. Statistics were by one-way analysis of variance (n = 6). RESULTS: S-1-P induced time-dependent activation of PLC-beta and PLC-gamma; PLC-beta but not PLC-gamma activation was blocked by U7 but not by InactiveU7. PLC-beta activation was Galphai-independent (not blocked by pertussis toxin, a Galphai inhibitor, or Galphai2 and Galphai3 siRNA) and Galphaq-independent (not blocked by glycoprotein [GP] 2A, a Galphaq inhibitor, or Galphaq siRNA). PLC-beta activation and cell migration was blocked by siRNA to Galpha12/13. Oxygen free radical generation induced by S-1-P, as measured by dihydroethidium staining, was significantly inhibited by U7 but not by InactiveU7. Inhibition of oxygen free radicals with the inhibitor diphenyleneiodonium resulted in decreased cell migration to S-1-P. VSMC mitogen-activated protein kinase activation and VSMC migration in response to S-1-P was inhibited by PLC- inhibition. CONCLUSION: S-1-P induces oxygen free radical generation through a Galpha12/13, PLC-beta-mediated mechanism that facilitates VSMC migration. To our knowledge, this is the first description of PLC-mediated oxygen free radical generation as a mediator of S-1-P VSMC migration and illustrates the need for the definition of cell signaling to allow targeted strategies in molecular therapeutics for restenosis.


Cell Movement , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Lysophospholipids/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phospholipase C beta/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Cell Movement/drug effects , Cells, Cultured , Enzyme Activation , Estrenes/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Pertussis Toxin/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phospholipase C beta/antagonists & inhibitors , Phospholipase C gamma/metabolism , Phosphorylation , Pyrrolidinones/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sphingosine/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Mol Pharmacol ; 69(6): 2068-75, 2006 Jun.
Article En | MEDLINE | ID: mdl-16554409

Because phospholipase C epsilon (PLC-epsilon) is activated by Galpha(12/13) and Rho family GTPases, we investigated whether these G proteins contribute to the increased inositol lipid hydrolysis observed in COS-7 cells after activation of certain G protein-coupled receptors. Stimulation of inositol lipid hydrolysis by endogenous lysophosphatidic acid (LPA) or thrombin receptors was markedly enhanced by the expression of PLC-epsilon. Expression of the LPA(1) or PAR1 receptor increased inositol phosphate production in response to LPA or SFLLRN, respectively, and these agonist-stimulated responses were markedly enhanced by coexpression of PLC-epsilon. Both LPA(1) and PAR1 receptor-mediated activation of PLC-epsilon was inhibited by coexpression of the regulator of G protein signaling (RGS) domain of p115RhoGEF, a GTPase-activating protein for Galpha(12/13) but not by expression of the RGS domain of GRK2, which inhibits Galpha(q) signaling. In contrast, activation of the G(q)-coupled M1 muscarinic or P2Y(2) purinergic receptor was neither enhanced by coexpression with PLC-epsilon nor inhibited by the RGS domain of p115RhoGEF but was blocked by expression of the RGS domain of GRK2. Expression of the Rho inhibitor C3 botulinum toxin did not affect LPA- or SFLLRN-stimulated inositol lipid hydrolysis in the absence of PLC-epsilon but completely prevented the PLC-epsilon-dependent increase in inositol phosphate accumulation. Likewise, C3 toxin blocked the PLC-epsilon-dependent stimulatory effects of the LPA(1), LPA(2), LPA(3), or PAR1 receptor but had no effect on the agonist-promoted inositol phosphate response of the M1 or P2Y(2) receptor. Moreover, PLC-epsilon-dependent stimulation of inositol phosphate accumulation by activation of the epidermal growth factor receptor, which involves Ras- but not Rho-mediated activation of the phospholipase, was unaffected by C3 toxin. These studies illustrate that specific LPA and thrombin receptors promote inositol lipid signaling via activation of Galpha(12/13) and Rho.


GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Receptor, PAR-1/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Type C Phospholipases/metabolism , rho GTP-Binding Proteins/metabolism , ADP Ribose Transferases/pharmacology , Animals , Botulinum Toxins/pharmacology , COS Cells , Chlorocebus aethiops , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , Hydrolysis , Inositol Phosphates/metabolism , Lipid Metabolism , Peptide Fragments/pharmacology , Phosphoinositide Phospholipase C , Receptors, Thrombin/metabolism , Type C Phospholipases/antagonists & inhibitors , rho GTP-Binding Proteins/agonists , rho GTP-Binding Proteins/antagonists & inhibitors
13.
Mol Pharmacol ; 69(3): 975-82, 2006 Mar.
Article En | MEDLINE | ID: mdl-16326932

Endothelial nitric-oxide synthase (eNOS) plays a crucial role in the regulation of a variety of cardiovascular and pulmonary functions in both normal and pathological conditions. Multiple signaling inputs, including calcium, caveolin-1, phosphorylation by several kinases, and binding to the 90-kDa heat shock protein (Hsp90), regulate eNOS activity. Here, we report a novel mechanism of G protein-dependent regulation of eNOS. We demonstrate that in mammalian cells, the alpha subunit of heterotrimeric G12 protein (G alpha12) can form a complex with eNOS in an activation- and Hsp90-independent manner. Our data show that G alpha12 does not affect eNOS-specific activity, but it strongly enhances total eNOS activity by increasing cellular levels of eNOS. Experiments using inhibition of protein or mRNA synthesis show that G alpha12 increases the expression of eNOS by increasing half-life of both eNOS protein and eNOS mRNA. Small interfering RNA-mediated depletion of endogenous G alpha12 decreases eNOS levels. A quantitative correlation can be detected between the extent of down-regulation of G alpha12 and eNOS in endothelial cells after prolonged treatment with thrombin. G protein-dependent increase of eNOS expression represents a novel mechanism by which heterotrimeric G proteins can regulate the activity of downstream signaling molecules.


GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Activation , Enzyme Stability , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Nitric Oxide Synthase Type III/genetics , RNA Stability , RNA, Messenger/analysis , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Thrombin/pharmacology
14.
J Biol Chem ; 280(18): 18434-41, 2005 May 06.
Article En | MEDLINE | ID: mdl-15743761

In the present study, we examined signal transduction mechanism of reactive oxygen species (ROS) production and the role of ROS in angiotensin II-induced activation of mitogen-activated protein kinases (MAPKs) in rat neonatal cardiomyocytes. Among three MAPKs, c-Jun NH(2)-terminal kinase (JNK) and p38 MAPK required ROS production for activation, as an NADPH oxidase inhibitor, diphenyleneiodonium, inhibited the activation. The angiotensin II-induced activation of JNK and p38 MAPK was also inhibited by the expression of the Galpha(12/13)-specific regulator of G protein signaling (RGS) domain, a specific inhibitor of Galpha(12/13), but not by an RGS domain specific for Galpha(q). Constitutively active Galpha(12)- or Galpha(13)-induced activation of JNK and p38 MAPK, but not extracellular signal-regulated kinase (ERK), was inhibited by diphenyleneiodonium. Angiotensin II receptor stimulation rapidly activated Galpha(13), which was completely inhibited by the Galpha(12/13)-specific RGS domain. Furthermore, the Galpha(12/13)-specific but not the Galpha(q)-specific RGS domain inhibited angiotensin II-induced ROS production. Dominant negative Rac inhibited angiotensin II-stimulated ROS production, JNK activation, and p38 MAPK activation but did not affect ERK activation. Rac activation was mediated by Rho and Rho kinase, because Rac activation was inhibited by C3 toxin and a Rho kinase inhibitor, Y27632. Furthermore, angiotensin II-induced Rho activation was inhibited by Galpha(12/13)-specific RGS domain but not dominant negative Rac. An inhibitor of epidermal growth factor receptor kinase AG1478 did not affect angiotensin II-induced JNK activation cascade. These results suggest that Galpha(12/13)-mediated ROS production through Rho and Rac is essential for JNK and p38 MAPK activation.


GTP-Binding Protein alpha Subunits, G12-G13/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Receptors, Angiotensin/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Angiotensin Receptor Antagonists , Animals , Animals, Newborn , Benzimidazoles/pharmacology , Biphenyl Compounds , Enzyme Activation/drug effects , Enzyme Activation/physiology , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , Mice , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Tetrazoles/pharmacology
...