Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 582: 1-7, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34678590

RESUMEN

In early stage of diabetes, insulin secretion from pancreatic ß-cells is increased to deal with the elevated blood glucose. Previous studies have reported that islet-produced carbon monoxide (CO) is associated with increased glucose-stimulated insulin secretion from ß-cells. However, this compensatory mechanism by which CO may act to enhance ß-cell function remain unclear. In this study, we revealed that CO promoted intracellular calcium ([Ca2+]i) elevation and glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells in leptin receptor deficient db/db mice but not in C57 mice. The stimulatory effects of CO on ß-cell function in db/db mice was blocked by inhibition of Phospholipase C (PLC) signaling pathway. We further demonstrated that CO triggered [Ca2+]i transients and enhanced GSIS in C57 islets when ß-cells overexpressed with PLCγ1 and PLCδ1, but not PLCß1. On the other hand, reducing PLCγ1 and PLCδ1 expressions in db/db islets dramatically attenuated the stimulatory effects of CO on ß-cell function, whereas interfering PLCß1 expression had no effects on CO-induced ß-cell function enhancement. Our findings showing that CO elevated [Ca2+]i and enhanced GSIS by activating PLC signaling through PLCγ1 and PLCδ1 isoforms in db/db pancreatic ß-cells may suggest an important mechanism by which CO promotes ß-cell function to prevent hyperglycemia. Our study may also provide new insights into the therapy for type II diabetes and offer a potential target for therapeutic applications of CO.


Asunto(s)
Calcio/metabolismo , Monóxido de Carbono/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Fosfolipasa C delta/genética , Fosfolipasa C gamma/genética , Animales , Línea Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica , Glucosa/metabolismo , Glucosa/farmacología , Insulina/biosíntesis , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Fosfolipasa C delta/antagonistas & inhibidores , Fosfolipasa C delta/metabolismo , Fosfolipasa C gamma/antagonistas & inhibidores , Fosfolipasa C gamma/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Transducción de Señal
2.
J Biol Chem ; 297(6): 101348, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34715131

RESUMEN

During signal transduction, the G protein, Gαq, binds and activates phospholipase C-ß isozymes. Several diseases have been shown to manifest upon constitutively activating mutation of Gαq, such as uveal melanoma. Therefore, methods are needed to directly inhibit Gαq. Previously, we demonstrated that a peptide derived from a helix-turn-helix (HTH) region of PLC-ß3 (residues 852-878) binds Gαq with low micromolar affinity and inhibits Gαq by competing with full-length PLC-ß isozymes for binding. Since the HTH peptide is unstructured in the absence of Gαq, we hypothesized that embedding the HTH in a folded protein might stabilize the binding-competent conformation and further improve the potency of inhibition. Using the molecular modeling software Rosetta, we searched the Protein Data Bank for proteins with similar HTH structures near their surface. The candidate proteins were computationally docked against Gαq, and their surfaces were redesigned to stabilize this interaction. We then used yeast surface display to affinity mature the designs. The most potent design bound Gαq/i with high affinity in vitro (KD = 18 nM) and inhibited activation of PLC-ß isozymes in HEK293 cells. We anticipate that our genetically encoded inhibitor will help interrogate the role of Gαq in healthy and disease model systems. Our work demonstrates that grafting interaction motifs into folded proteins is a powerful approach for generating inhibitors of protein-protein interactions.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Péptidos/farmacología , Clonación Molecular , Bases de Datos de Proteínas , Diseño de Fármacos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Péptidos/química , Péptidos/genética , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/química , Fosfolipasa C beta/metabolismo , Unión Proteica , Ingeniería de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
3.
Cell Signal ; 62: 109349, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31254604

RESUMEN

Phospholipase Cß (PLCß) enzymes regulate second messenger production following the activation of G protein-coupled receptors (GPCRs). Under basal conditions, these enzymes are maintained in an autoinhibited state by multiple elements, including an insertion within the catalytic domain known as the X-Y linker. Although the PLCß X-Y linker is variable in sequence and length, its C-terminus is conserved and features an acidic stretch, followed by a short helix. This helix interacts with residues near the active site, acting as a lid to sterically prevent substrate binding. However, deletions that remove the acidic stretch of the X-Y linker increase basal activity to the same extent as deletion of the entire X-Y linker. Thus, the acidic stretch may be the linchpin in autoinhibition mediated by the X-Y linker. We used site-directed mutagenesis and biochemical assays to investigate the importance of this acidic charge in mediating PLCß3 autoinhibition. Loss of the acidic charge in the X-Y linker increases basal activity and decreases stability, consistent with loss of autoinhibition. However, introduction of compensatory electrostatic mutations on the surface of the PLCß3 catalytic domain restore activity to basal levels. Thus, intramolecular electrostatics modulate autoinhibition by the X-Y linker.


Asunto(s)
Dominio Catalítico/genética , Fosfolipasa C beta/genética , Conformación Proteica en Hélice alfa , Electricidad Estática , Humanos , Mutagénesis Sitio-Dirigida , Fosfolipasa C beta/antagonistas & inhibidores , Fosforilación , Receptores Acoplados a Proteínas G/genética
4.
Cancer Res ; 78(8): 2052-2064, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29567857

RESUMEN

Triple-negative breast cancer (TNBC) exhibits more traits possessed by cancer stem cells (CSC) than other breast cancer subtypes and is more likely to develop brain metastases. TNBC patients usually have shorter survival time after diagnosis of brain metastasis, suggesting an innate ability of TNBC tumor cells in adapting to the brain. In this study, we establish novel animal models to investigate early tumor adaptation in brain metastases by introducing both patient-derived and cell line-derived CSC-enriched brain metastasis tumorsphere cells into mice. We discovered astrocyte-involved tumor activation of protocadherin 7 (PCDH7)-PLCß-Ca2+-CaMKII/S100A4 signaling as a mediator of brain metastatic tumor outgrowth. We further identified and evaluated the efficacy of a known drug, the selective PLC inhibitor edelfosine, in suppressing the PCDH7 signaling pathway to prohibit brain metastases in the animal models. The results of this study reveal a novel signaling pathway for brain metastases in TNBC and indicate a promising strategy of metastatic breast cancer prevention and treatment by targeting organ-adaptive cancer stem cells.Significance: These findings identify a compound to block adaptive signaling between cancer stem cells and brain astrocytes. Cancer Res; 78(8); 2052-64. ©2018 AACR.


Asunto(s)
Adaptación Fisiológica , Neoplasias Encefálicas/prevención & control , Neoplasias Encefálicas/secundario , Células Madre Neoplásicas/patología , Neoplasias de la Mama Triple Negativas/patología , Animales , Cadherinas/genética , Cadherinas/metabolismo , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular Tumoral , Proliferación Celular , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/metabolismo , Éteres Fosfolípidos/farmacología , Protocadherinas , ARN Mensajero/genética , Estudios Retrospectivos , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal
5.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L747-58, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773068

RESUMEN

Enhanced contractility of airway smooth muscle (ASM) is a major pathophysiological characteristic of asthma. Expanding the therapeutic armamentarium beyond ß-agonists that target ASM hypercontractility would substantially improve treatment options. Recent studies have identified naturally occurring phytochemicals as candidates for acute ASM relaxation. Several flavonoids were evaluated for their ability to acutely relax human and murine ASM ex vivo and murine airways in vivo and were evaluated for their ability to inhibit procontractile signaling pathways in human ASM (hASM) cells. Two members of the flavonol subfamily, galangin and fisetin, significantly relaxed acetylcholine-precontracted murine tracheal rings ex vivo (n = 4 and n = 5, respectively, P < 0.001). Galangin and fisetin also relaxed acetylcholine-precontracted hASM strips ex vivo (n = 6-8, P < 0.001). Functional respiratory in vivo murine studies demonstrated that inhaled galangin attenuated the increase in lung resistance induced by inhaled methacholine (n = 6, P < 0.01). Both flavonols, galangin and fisetin, significantly inhibited purified phosphodiesterase-4 (PDE4) (n = 7, P < 0.05; n = 7, P < 0.05, respectively), and PLCß enzymes (n = 6, P < 0.001 and n = 6, P < 0.001, respectively) attenuated procontractile Gq agonists' increase in intracellular calcium (n = 11, P < 0.001), acetylcholine-induced increases in inositol phosphates, and CPI-17 phosphorylation (n = 9, P < 0.01) in hASM cells. The prorelaxant effect retained in these structurally similar flavonols provides a novel pharmacological method for dual inhibition of PLCß and PDE4 and therefore may serve as a potential treatment option for acute ASM constriction.


Asunto(s)
Flavonoides/farmacología , Relajación Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Fosfolipasa C beta/antagonistas & inhibidores , Animales , Aorta/efectos de los fármacos , Aorta/fisiopatología , Asma/tratamiento farmacológico , Broncoconstricción/efectos de los fármacos , Señalización del Calcio , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Evaluación Preclínica de Medicamentos , Flavonoides/química , Flavonoles , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratones , Contracción Muscular , Músculo Liso/fisiología , Músculo Liso/fisiopatología , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/farmacología , Fosfolipasa C beta/fisiología
6.
FASEB J ; 30(2): 702-15, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26467792

RESUMEN

Depletion of sarcoplasmic reticulum (SR) Ca(2+) stores activates store-operated channels (SOCs) composed of canonical transient receptor potential (TRPC) 1 proteins in vascular smooth muscle cells (VSMCs), which contribute to important cellular functions. We have previously shown that PKC is obligatory for activation of TRPC1 SOCs in VSMCs, and the present study investigates if the classic phosphoinositol signaling pathway involving Gαq-mediated PLC activity is responsible for driving PKC-dependent channel gating. The G-protein inhibitor GDP-ß-S, anti-Gαq antibodies, the PLC inhibitor U73122, and the PKC inhibitor GF109203X all inhibited activation of TRPC1 SOCs, and U73122 and GF109203X also reduced store-operated PKC-dependent phosphorylation of TRPC1 proteins. Three distinct SR Ca(2+) store-depleting agents, 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester, cyclopiazonic acid, and N,N,N',N'-tetrakis(2-pyridylmethyl)ethane-1,2-diamineed, induced translocations of the fluorescent biosensor GFP-PLCδ1-PH from the cell membrane to the cytosol, which were inhibited by U73122. Knockdown of PLCß1 with small hairpin RNA reduced both store-operated PLC activity and stimulation of TRPC1 SOCs. Immunoprecipitation studies and proximity ligation assays revealed that store depletion induced interactions between TRPC1 and Gαq, and TRPC1 and PLCß1. We propose a novel activation mechanism for TRPC1 SOCs in VSMCs, in which store depletion induces formation of TRPC1-Gαq-PLCß1 complexes that lead to PKC stimulation and channel gating.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Activación del Canal Iónico/fisiología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosfolipasa C beta/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Técnicas de Silenciamiento del Gen , Activación del Canal Iónico/efectos de los fármacos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Conejos , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética
7.
PLoS One ; 10(10): e0139638, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26447765

RESUMEN

Metabolic syndrome is characterized by visceral adiposity, insulin resistance, high triglyceride (TG)- and low high-density lipoprotein cholesterol-levels, hypertension, and diabetes-all of which often cause cardiovascular and cerebrovascular diseases. It remains unclear, however, why visceral adiposity but not subcutaneous adiposity causes insulin resistance and other pathological situations. Lipoprotein lipase (LPL) catalyzes hydrolysis of TG in plasma lipoproteins. In the present study, we investigated whether the effects of angiotensin II (AngII) on TG metabolism are mediated through an effect on LPL expression. Adipose tissues were divided into visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) for comparison. AngII accelerated LPL expression in SAT but, on the contrary, suppressed its expression in VAT. In both SAT and VAT, AngII signaled through the same type 1 receptor. In SAT, AngII increased LPL expression via c-Src and p38 MAPK signaling. In VAT, however, AngII reduced LPL expression via the Gq class of G proteins and the subsequent phospholipase C ß4 (PLCß4), protein kinase C ß1, nuclear factor κB, and inducible nitric oxide synthase signaling pathways. PLCß4 small interfering RNA experiments showed that PLCß4 expression is important for the AngII-induced LPL reduction in VAT, in which PLCß4 expression increases in the evening and falls at night. Interestingly, PLCß4 expression in VAT decreased with fasting, while AngII did not decrease LPL expression in VAT in a fasting state. In conclusion, AngII reduces LPL expression through PLCß4, the expression of which is regulated by feeding in VAT, whereas AngII increases LPL expression in SAT. The different effects of AngII on LPL expression and, hence, TG metabolism in VAT and SAT may partly explain their different contributions to the development of metabolic syndrome.


Asunto(s)
Angiotensina II/farmacología , Grasa Intraabdominal/efectos de los fármacos , Lipoproteína Lipasa/metabolismo , Fosfolipasa C beta/metabolismo , Grasa Subcutánea/efectos de los fármacos , Familia-src Quinasas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína Tirosina Quinasa CSK , Células Cultivadas , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Lipoproteína Lipasa/genética , Masculino , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/genética , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo , Triglicéridos/sangre , Triglicéridos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Neurophysiol ; 112(10): 2605-15, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25185819

RESUMEN

Endocannabinoids (eCBs) released from postsynaptic neurons mediate retrograde suppression of neurotransmitter release at central synapses. eCBs are crucial for establishing proper synaptic connectivity in the developing nervous system. Mobilization of eCBs is driven either by a rise in intracellular Ca(2+) (depolarization-induced suppression of inhibition, DSI) or postsynaptic G protein-coupled receptors (GPCRs) that activate phospholipase C beta (PLCß). To determine whether eCB mobilization changes between neonatal and juvenile ages, we used whole cell voltage-clamp recordings of CA1 neurons from rat hippocampal slices at postnatal days 1-18 (neonatal) and 19-43 (juvenile), because many neurophysiological parameters change dramatically between approximately postnatal days 18-20. We found that DSI was slightly greater in juveniles than in neonates, while eCB mobilization stimulated by GPCRs was unchanged. However, when DSI was elicited during GPCR activation, its increase was much greater in juveniles, suggesting that eCB mobilization caused by the synergy between the Ca(2+) and GPCR pathways is developmentally upregulated. Western blotting revealed significant increases in both metabotropic type glutamate receptor 5 (mGluR5) and PLCß1 proteins in juveniles compared with neonates. Responses to pharmacological activation or inhibition of PLC implied that eCB upregulation is associated with a functional increase in PLC activity. We conclude that synergistic eCB mobilization in hippocampal CA1 neurons is greater in juveniles than in neonates, and that this may result from increases in the mGluR5-PLCß1 eCB pathway. The data enhance our understanding of the developmental regulation of the eCB system and may provide insight into diseases caused by improper cortical wiring, or the impact of cannabis exposure during development.


Asunto(s)
Región CA1 Hipocampal/crecimiento & desarrollo , Endocannabinoides/metabolismo , Fosfolipasa C beta/metabolismo , Células Piramidales/crecimiento & desarrollo , Receptor del Glutamato Metabotropico 5/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiología , Femenino , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Técnicas de Placa-Clamp , Fosfolipasa C beta/antagonistas & inhibidores , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/metabolismo , Receptor del Glutamato Metabotropico 5/agonistas , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo , Técnicas de Cultivo de Tejidos
9.
Am J Respir Cell Mol Biol ; 50(1): 115-24, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23962082

RESUMEN

ß-Agonists are the first-line therapy to alleviate asthma symptoms by acutely relaxing the airway. Purified components of ginger relax airway smooth muscle (ASM), but the mechanisms are unclear. By elucidating these mechanisms, we can explore the use of phytotherapeutics in combination with traditional asthma therapies. The objectives of this study were to: (1) determine if 6-gingerol, 8-gingerol, or 6-shogaol potentiate ß-agonist-induced ASM relaxation; and (2) define the mechanism(s) of action responsible for this potentiation. Human ASM was contracted in organ baths. Tissues were relaxed dose dependently with ß-agonist, isoproterenol, in the presence of vehicle, 6-gingerol, 8-gingerol, or 6-shogaol (100 µM). Primary human ASM cells were used for cellular experiments. Purified phosphodiesterase (PDE) 4D or phospholipase C ß enzyme was used to assess inhibitory activity of ginger components using fluorescent assays. A G-LISA assay was used to determine the effects of ginger constituents on Ras homolog gene family member A activation. Significant potentiation of isoproterenol-induced relaxation was observed with each of the ginger constituents. 6-Shogaol showed the largest shift in isoproterenol half-maximal effective concentration. 6-Gingerol, 8-gingerol, or 6-shogaol significantly inhibited PDE4D, whereas 8-gingerol and 6-shogaol also inhibited phospholipase C ß activity. 6-Shogaol alone inhibited Ras homolog gene family member A activation. In human ASM cells, these constituents decreased phosphorylation of 17-kD protein kinase C-potentiated inhibitory protein of type 1 protein phosphatase and 8-gingerol decreased myosin light chain phosphorylation. Isolated components of ginger potentiate ß-agonist-induced relaxation in human ASM. This potentiation involves PDE4D inhibition and cytoskeletal regulatory proteins. Together with ß-agonists, 6-gingerol, 8-gingerol, or 6-shogaol may augment existing asthma therapy, resulting in relief of symptoms through complementary intracellular pathways.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Proteínas del Citoesqueleto/metabolismo , Relajación Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Extractos Vegetales/farmacología , Zingiber officinale/química , Asma/tratamiento farmacológico , Asma/metabolismo , Catecoles/farmacología , Línea Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Alcoholes Grasos/farmacología , Proteínas del Choque Térmico HSP20/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas Musculares , Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosfatidilinositoles/antagonistas & inhibidores , Fosfatidilinositoles/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación/efectos de los fármacos , Extractos Vegetales/química , Canales de Potasio/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
10.
FASEB J ; 27(12): 5122-30, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24008754

RESUMEN

As shown in a large clinical prospective trial, inhibition of the renin-angiotensin system (RAS) can delay the onset of type 2 diabetes in high-risk individuals. We evaluated the beneficial effects of RAS inhibition on ß-cell function under glucotoxic conditions. Human islets from 13 donors were cultured in 5.5 mM (controls) or 16.7 mM glucose [high glucose (HG)] for 4 d with or without losartan (5 µM), a selective AT1R blocker, and/or U73122 (2 µM), a selective PLC inhibitor, during the last 2 d. HG induced RAS activation with overexpression of AT1R (P<0.05) and angiotensinogen (P<0.001) mRNAs. HG increased endoplasmic reticulum (ER) stress markers (P<0.001) such as GRP78, sXBP1, and ATF4 mRNAs and Grp78 protein levels (P<0.01). HG also decreased reticular calcium concentration (P<0.0001) and modified protein expressions of ER calcium pumps with reduction of SERCA2b (P<0.01) and increase of IP3R2 (P<0.05). Losartan prevented these deleterious effects and was associated with improved insulin secretion despite HG exposure. AT1R activation triggers the PLC-IP3-calcium pathway. Losartan prevented the increase of PLC ß1 and γ1 protein levels induced by HG (P<0.05). U73122 reproduced all the protective effects of losartan. AT1R blockade protects human islets from the deleterious effects of glucose through inhibition of the PLC-IP3-calcium pathway.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Glucosa/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Losartán/farmacología , Fosfolipasa C beta/metabolismo , Fosfolipasa C gamma/metabolismo , Calcio/metabolismo , Señalización del Calcio , Células Cultivadas , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Estrenos/farmacología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C gamma/antagonistas & inhibidores , Pirrolidinonas/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 305(5): L396-403, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23873842

RESUMEN

Asthma is a disease of the airways with symptoms including exaggerated airway narrowing and airway inflammation. Early asthma therapies used methylxanthines to relieve symptoms, in part, by inhibiting cyclic nucleotide phosphodiesterases (PDEs), the enzyme responsible for degrading cAMP. The classification of tissue-specific PDE subtypes and the clinical introduction of PDE-selective inhibitors for chronic obstructive pulmonary disease (i.e., roflumilast) have reopened the possibility of using PDE inhibition in the treatment of asthma. Quercetin is a naturally derived PDE4-selective inhibitor found in fruits, vegetables, and tea. We hypothesized that quercetin relaxes airway smooth muscle via cAMP-mediated pathways and augments ß-agonist relaxation. Tracheal rings from male A/J mice were mounted in myographs and contracted with acetylcholine (ACh). Addition of quercetin (100 nM-1 mM) acutely and concentration-dependently relaxed airway rings precontracted with ACh. In separate studies, pretreatment with quercetin (100 µM) prevented force generation upon exposure to ACh. In additional studies, quercetin (50 µM) significantly potentiated isoproterenol-induced relaxations. In in vitro assays, quercetin directly attenuated phospholipase C activity, decreased inositol phosphate synthesis, and decreased intracellular calcium responses to Gq-coupled agonists (histamine or bradykinin). Finally, nebulization of quercetin (100 µM) in an in vivo model of airway responsiveness significantly attenuated methacholine-induced increases in airway resistance. These novel data show that the natural PDE4-selective inhibitor quercetin may provide therapeutic relief of asthma symptoms and decrease reliance on short-acting ß-agonists.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Isoproterenol/farmacología , Relajación Muscular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Fosfodiesterasa 4/farmacología , Fosfolipasa C beta/antagonistas & inhibidores , Quercetina/farmacología , Sistema Respiratorio/efectos de los fármacos , Acetilcolina/farmacología , Agonistas Adrenérgicos beta/farmacología , Resistencia de las Vías Respiratorias/efectos de los fármacos , Animales , Antioxidantes/farmacología , Western Blotting , Broncoconstrictores/farmacología , Calcio/metabolismo , Células Cultivadas , Agonistas Colinérgicos/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Cloruro de Metacolina/farmacología , Ratones , Ratones Endogámicos A , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Fosfolipasa C beta/metabolismo , Prostaglandinas/metabolismo , Sistema Respiratorio/metabolismo , Fosfolipasas de Tipo C/metabolismo
12.
PLoS Genet ; 9(6): e1003559, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23754968

RESUMEN

Rhodopsin has been used as a prototype system to investigate G protein-coupled receptor (GPCR) internalization and endocytic sorting mechanisms. Failure of rhodopsin recycling upon light activation results in various degenerative retinal diseases. Accumulation of internalized rhodopsin in late endosomes and the impairment of its lysosomal degradation are associated with unregulated cell death that occurs in dystrophies. However, the molecular basis of rhodopsin accumulation remains elusive. We found that the novel norpA(P24) suppressor, diehard4, is responsible for the inability of endo-lysosomal rhodopsin trafficking and retinal degeneration in Drosophila models of retinal dystrophies. We found that diehard4 encodes Osiris 21. Loss of its function suppresses retinal degeneration in norpA(P24), rdgC(306), and trp(1), but not in rdgB(2), suggesting a common cause of photoreceptor death. In addition, the loss of Osiris 21 function shifts the membrane balance between late endosomes and lysosomes as evidenced by smaller late endosomes and the proliferation of lysosomal compartments, thus facilitating the degradation of endocytosed rhodopsin. Our results demonstrate the existence of negative regulation in vesicular traffic between endosomes and lysosomes. We anticipate that the identification of additional components and an in-depth description of this specific molecular machinery will aid in therapeutic interventions of various retinal dystrophies and GPCR-related human diseases.


Asunto(s)
Proteínas de Drosophila/genética , Endocitosis/genética , Proteínas de la Membrana/genética , Fosfolipasa C beta/genética , Distrofias Retinianas/genética , Rodopsina/genética , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Endosomas/genética , Endosomas/metabolismo , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Distrofias Retinianas/metabolismo , Distrofias Retinianas/patología , Rodopsina/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
13.
J Gen Virol ; 94(Pt 5): 1111-1120, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23303826

RESUMEN

Human cytomegalovirus (HCMV)-encoded G protein-coupled-receptor US28 is believed to participate in virus dissemination through modulation of cell migration and immune evasion. US28 binds different CC chemokines and the CX3C chemokine CX3CL1. Membrane-anchored CX3CL1 is expressed by immune-activated endothelial cells, causing redirection of CX3CR1-expressing leukocytes in the blood to sites of infection. Here, we used stable transfected cell lines to examine how US28 expression affects cell migration on immobilized full-length CX3CL1, to model how HCMV-infected leukocytes interact with inflamed endothelium. We observed that US28-expressing cells migrated more than CX3CR1-expressing cells when adhering to immobilized CX3CL1. US28-induced migration was G protein-signalling dependent and was blocked by the phospholipase Cß inhibitor U73122 and the intracellular calcium chelator BAPTA-AM. In addition, migration was inhibited in a dose-dependent manner by competition from CCL2 and CCL5, whereas CCL3 had little effect. Instead of migrating, CX3CR1-expressing cells performed 'dancing-on-the-spot' movements, demonstrating that anchored CX3CL1 acts as a strong tether for these cells. At low receptor expression levels, however, no significant difference in migration potential was observed when comparing the migration of CX3CR1- and US28-expressing cells. Thus, these data showed that, in contrast to CX3CR1, which promotes efficient cell capture upon binding to anchored CX3CL1, US28 acts to increase the migration of cells upon binding to the same ligand. Overall, this indicates that infected cells probably move more than uninfected cells in inflamed tissues with high CX3CL1 expression, with soluble chemokines affecting the final migration.


Asunto(s)
Movimiento Celular , Quimiocina CX3CL1/metabolismo , Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Receptor 1 de Quimiocinas CX3C , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quelantes/farmacología , Quimiocina CX3CL1/genética , Quimiocinas CC/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Células Endoteliales , Estrenos/farmacología , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa C beta/antagonistas & inhibidores , Pirrolidinonas/farmacología , Receptores de Quimiocina/genética , Transducción de Señal , Imagen de Lapso de Tiempo , Proteínas Virales/genética
14.
J Gastrointest Surg ; 17(1): 153-8; discussion p. 158, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22948835

RESUMEN

INTRODUCTION: Sweet taste receptors may enhance glucose absorption. AIM: This study aimed to explore the cell biology of sweet taste receptors on glucose uptake. HYPOTHESIS: Artificial sweeteners increase glucose uptake via activating sweet taste receptors in the enterocyte to translocate GLUT2 to the apical membrane through the PLC ßII pathway. METHODS: Caco-2, RIE-1, and IEC-6 cells, starved from glucose for 1 h were pre-incubated with 10 mM acesulfame potassium (AceK). Glucose uptake was measured by incubating cells for 1 to 10 min with 0.5-50 mM glucose with or without U-73122, chelerythrine, and cytochalasin B. RESULTS: In Caco-2 and RIE-1 cells, 10 mM AceK increased glucose uptake by 20-30 % at glucose >25 mM, but not in lesser glucose concentrations (<10 mM), nor at 1 min or 10 min incubations. U-73122 (PLC ßII inhibitor) inhibited uptake at glucose >25 mM and for 5 min incubation; chelerythrine and cytochalasin B had similar effects. No effect occurred in IEC-6 cells. Activation of sweet taste receptors had no effect on glucose uptake in low (<25 mM) glucose concentrations but increased uptake at greater concentrations (>25 mM). CONCLUSIONS: Role of artificial sweeteners on glucose uptake appears to act in part by effects on the enterocyte itself.


Asunto(s)
Enterocitos/efectos de los fármacos , Glucosa/farmacocinética , Absorción Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Edulcorantes/farmacología , Tiazinas/farmacología , Animales , Biomarcadores/metabolismo , Células CACO-2 , Línea Celular , Enterocitos/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Humanos , Absorción Intestinal/fisiología , Intestino Delgado/citología , Intestino Delgado/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/metabolismo , Ratas , Edulcorantes/metabolismo , Edulcorantes/farmacocinética , Tiazinas/metabolismo , Factores de Tiempo
15.
Cell Calcium ; 52(6): 445-56, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22938798

RESUMEN

In skeletal muscles from patient suffering of Duchenne Muscular Dystrophy and from mdx mice, the absence of the cytoskeleton protein dystrophin has been shown to be essential for maintaining a normal calcium influx. We showed that a TRPC store-dependent cation influx is increased by loss of dystrophin or a scaffolding protein α1-syntrophin, however the mechanisms of this calcium mishandling are incompletely understood. First of all, we confirmed that TRPC1 but also STIM1 and Orai1 are supporting the store-operated cation entry which is enhanced in dystrophin-deficient myotubes. Next, we demonstrated that inhibition of PLC or PKC in dystrophin-deficient myotubes restores elevated cation entry to normal levels similarly to enforced minidystrophin expression. In addition, silencing α1-syntrophin also increased cation influx in a PLC/PKC dependent pathway. We also showed that α1-syntrophin and PLCß are part of a same protein complex reinforcing the idea of their inter-relation in calcium influx regulation. This elevated cation entry was decreased to normal levels by chelating intracellular free calcium with BAPTA-AM. Double treatments with BAPTA-AM and PLC or PKC inhibitors suggested that the elevation of cation influx by PLC/PKC pathway is dependent on cytosolic calcium. All these results demonstrate an involvement in dystrophin-deficient myotubes of a specific calcium/PKC/PLC pathway in elevation of store-operated cation influx supported by the STIM1/Orai1/TRPC1 proteins, which is normally regulated by the α1-syntrophin/dystrophin scaffold.


Asunto(s)
Calcio/metabolismo , Distrofina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fosfolipasa C beta/metabolismo , Proteína Quinasa C/metabolismo , Animales , Canales de Calcio/química , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Quelantes/farmacología , Distrofina/genética , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Inhibidores Enzimáticos/farmacología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteína ORAI1 , Fosfolipasa C beta/antagonistas & inhibidores , Proteína Quinasa C/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
16.
Brain Res ; 1472: 38-44, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22771399

RESUMEN

Morphine is a mainstay for chronic pain treatment, but its efficacy has been hampered by physical tolerance. The underlying mechanism for chronic morphine induced tolerance is complicated and not well understood. PLC(ß3) is regarded as an important factor in the morphine tolerance signal pathway. In this study, we determined intrathecal (i.t.) administration of an antisense oligodeoxynucleotide (ODN) of PLC(ß3) could quicken the on-set antinociceptive efficacy of acute morphine treatment and prolong the maximum effect up to 4h. The antisense could also attenuate the development of morphine-induced tolerance and left shift the ED50 after 7 day of coadministration with morphine. These results probably were contributed by the PLC(ß3) antisense ODN as they successfully knocked down protein expression levels and reduced activity of PLC(ß3) in spinal cord in rats. The mismatch group had no such effects. The results confirmed the important involvement of PLC(ß3) in both acute morphine efficacy and chronic morphine tolerance at spinal level in rats. This study may provide an idea for producing a novel adjuvant for morphine treatment.


Asunto(s)
Analgésicos Opioides/farmacología , Tolerancia a Medicamentos , Morfina/farmacología , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Fosfolipasa C beta/antagonistas & inhibidores , Animales , Inyecciones Espinales , Masculino , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos
17.
Blood ; 120(11): 2167-73, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22674805

RESUMEN

VEGF induces vascular permeability (VP) in ischemic diseases and cancer, leading to many pathophysiological consequences. The molecular mechanisms by which VEGF acts to induce hyperpermeability are poorly understood and in vivo models that easily facilitate real-time, genetic studies of VP do not exist. In the present study, we report a heat-inducible VEGF transgenic zebrafish (Danio rerio) model through which VP can be monitored in real time. Using this approach with morpholino-mediated gene knock-down and knockout mice, we describe a novel role of phospholipase Cß3 as a negative regulator of VEGF-mediated VP by regulating intracellular Ca2+ release. Our results suggest an important effect of PLCß3 on VP and provide a new model with which to identify genetic regulators of VP crucial to several disease processes.


Asunto(s)
Permeabilidad Capilar , Endotelio Vascular/metabolismo , Fosfolipasa C beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Animales Modificados Genéticamente , Señalización del Calcio/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Embrión no Mamífero , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Respuesta al Choque Térmico , Ensayos Analíticos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Noqueados , Morfolinos/farmacología , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
18.
Mol Cell Endocrinol ; 351(2): 176-83, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22209746

RESUMEN

The physiological signal activating cytoplasmic accumulation of nuclear receptor interacting protein 140 (RIP140) in adipocytes was unclear. We uncover that endothelin-1 (ET-1) promotes cytoplasmic accumulation of RIP140 in 3T3-L1 adipocytes. We determine ET-1's signal transduction pathway in adipocytes, which is by activating ET(A) receptor-PLCß-nuclear PKCε. Blocking this pathway in 3T3-L1 adipocyte cultures, by treating cells with an ET(A) antagonist, inhibiting PLCß, or silencing PKCε, reduces ET-1-stimulated cytoplasmic accumulation of RIP140. In a HFD-fed obese mouse model, administration of a selective ET(A) antagonist, ambrisentan, effectively dampens cytoplasmic accumulation of RIP140 in the epididymal adipose tissue and reduces HFD-caused adipocyte dysfunctions. Importantly, ambrisentan improves blood glucose control and reduces the severity of hepatic steatosis in HFD-fed mice. This study reports a physiological signal that stimulates nuclear export of RIP140 in adipocytes and provides evidence for a strategy using selective ET(A) antagonist to treat obesity-induced insulin resistance and, possibly, other metabolic disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antagonistas de los Receptores de la Endotelina A , Endotelina-1/antagonistas & inhibidores , Endotelina-1/metabolismo , Proteínas Nucleares/metabolismo , Fenilpropionatos/farmacología , Piridazinas/farmacología , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Glucemia/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa , Endotelina-1/farmacología , Hígado Graso , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteína de Interacción con Receptores Nucleares 1 , Obesidad , Fosfoinositido Fosfolipasa C/genética , Fosfoinositido Fosfolipasa C/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/metabolismo , Interferencia de ARN , ARN Citoplasmático Pequeño , Receptor de Endotelina A/metabolismo
19.
Pflugers Arch ; 463(1): 89-102, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22083644

RESUMEN

Modulation of the standing outward current (I (SO)) by muscarinic acetylcholine (ACh) receptor (MAChR) stimulation is fundamental for the state-dependent change in activity mode of thalamocortical relay (TC) neurons. Here, we probe the contribution of MAChR subtypes, G proteins, phospholipase C (PLC), and two pore domain K(+) (K(2P)) channels to this signaling cascade. By the use of spadin and A293 as specific blockers, we identify TWIK-related K(+) (TREK)-1 channel as new targets and confirm TWIK-related acid-sensitve K(+) (TASK)-1 channels as known effectors of muscarinic signaling in TC neurons. These findings were confirmed using a high affinity blocker of TASK-3 and TREK-1, namely, tetrahexylammonium chloride. It was found that the effect of muscarinic stimulation was inhibited by M(1)AChR-(pirenzepine, MT-7) and M(3)AChR-specific (4-DAMP) antagonists, phosphoinositide-specific PLCß (PI-PLC) inhibitors (U73122, ET-18-OCH(3)), but not the phosphatidylcholine-specific PLC (PC-PLC) blocker D609. By comparison, depleting guanosine-5'-triphosphate (GTP) in the intracellular milieu nearly completely abolished the effect of MAChR stimulation. The block of TASK and TREK channels was accompanied by a reduction of the muscarinic effect on I (SO). Current-clamp recordings revealed a membrane depolarization following MAChR stimulation, which was sufficient to switch TC neurons from burst to tonic firing under control conditions but not during block of M(1)AChR/M(3)AChR and in the absence of intracellular GTP. These findings point to a critical role of G proteins and PLC as well as TASK and TREK channels in the muscarinic modulation of thalamic activity modes.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas Colinérgicas/fisiología , Transducción de Señal/fisiología , Sueño/fisiología , Tálamo/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Neuronas Colinérgicas/efectos de los fármacos , Fenómenos Electrofisiológicos/efectos de los fármacos , Fenómenos Electrofisiológicos/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Expresión Génica/genética , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Guanosina Trifosfato/antagonistas & inhibidores , Guanosina Trifosfato/metabolismo , Concentración de Iones de Hidrógeno , Núcleos Talámicos Laterales/citología , Núcleos Talámicos Laterales/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Muscarina/farmacología , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Proteínas del Tejido Nervioso , Oxotremorina/análogos & derivados , Oxotremorina/farmacología , Técnicas de Placa-Clamp , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Ratas , Ratas Long-Evans , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inhibidores , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/antagonistas & inhibidores , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Transducción de Señal/efectos de los fármacos , Tálamo/citología , Tionucleótidos/farmacología
20.
Placenta ; 32(12): 1033-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21993263

RESUMEN

Serotonin 5-HT(2A) receptor activation improves viability, increases DNA synthesis and activates JAK2-STAT3 and MEK1/2-ERK1/2 signalling pathways in JEG-3 human trophoblast choriocarcinoma cells. The goal of this study was to characterize the signal transduction cascade involved in 5-HT(2A) receptor-induced growth of JEG-3 cells. Selective 5-HT(2A) receptor agonist, DOI, induced JEG-3 cell growth was inhibited by the inhibitor of JAK2 (AG490), MEK1/2 (U0126), phospholipase C-ß (PLC-ß; U73122) and protein kinase C-ß (PKC-ß; Gö6976)), whereas the selective phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) had no effect. Specific inhibitors of PLC-ß, PKC-ß and Ras (farnesylthiosalicylic acid) inhibit activation of ERK1/2, whereas the PKC-ζ inhibitor GF109203X had no effect. Interestingly, inhibition of JAK2 prevented DOI-induced phosphorylation of ERK1/2 whereas inhibition of ERK1/2 pathway had no effect on DOI-induced activation of STAT3. Taken together, our results demonstrate that both the JAK2-STAT3 and PLC-ß-PKC-ß-Ras-ERK1/2 signalling pathways are involved in the stimulation of JEG-3 cell growth mediated by DOI. Moreover, this study shows that activation of JAK2 by the 5-HT(2A) receptor is essential to activate both STAT3 and ERK1/2 signalling pathways as well as to increase JEG-3 choriocarcinoma cell growth and survival.


Asunto(s)
Janus Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Receptor de Serotonina 5-HT2A/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Anfetaminas/antagonistas & inhibidores , Butadienos/farmacología , Carbazoles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Coriocarcinoma/metabolismo , Estrenos/farmacología , Femenino , Humanos , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasa/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosforilación , Embarazo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C beta , Pirrolidinonas/farmacología , Transducción de Señal/fisiología , Tirfostinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...