Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 42(10): 1229-1241, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35861069

RESUMO

BACKGROUND: Regulation of vascular permeability is critical to maintaining tissue metabolic homeostasis. VEGF (vascular endothelial growth factor) is a key stimulus of vascular permeability in acute and chronic diseases including ischemia reperfusion injury, sepsis, and cancer. Identification of novel regulators of vascular permeability would allow for the development of effective targeted therapeutics for patients with unmet medical need. METHODS: In vitro and in vivo models of VEGFA-induced vascular permeability, pathological permeability, quantitation of intracellular calcium release and cell entry, and phosphatidylinositol 4,5-bisphosphate levels were evaluated with and without modulation of PLC (phospholipase C) ß2. RESULTS: Global knock-out of PLCß2 in mice resulted in blockade of VEGFA-induced vascular permeability in vivo and transendothelial permeability in primary lung endothelial cells. Further work in an immortalized human microvascular cell line modulated with stable knockdown of PLCß2 recapitulated the observations in the mouse model and primary cell assays. Additionally, loss of PLCß2 limited both intracellular release and extracellular entry of calcium following VEGF stimulation as well as reduced basal and VEGFA-stimulated levels of phosphatidylinositol 4,5-bisphosphate compared to control cells. Finally, loss of PLCß2 in both a hyperoxia-induced lung permeability model and a cardiac ischemia:reperfusion model resulted in improved animal outcomes when compared with wild-type controls. CONCLUSIONS: The results implicate PLCß2 as a key positive regulator of VEGF-induced vascular permeability through regulation of both calcium flux and phosphatidylinositol 4,5-bisphosphate levels at the cellular level. Targeting of PLCß2 in a therapeutic setting may provide a novel approach to regulating vascular permeability in patients.


Assuntos
Permeabilidade Capilar , Fosfatidilinositol 4,5-Difosfato , Fosfolipase C beta , Mucosa Respiratória , Fator A de Crescimento do Endotélio Vascular , Animais , Cálcio/metabolismo , Permeabilidade Capilar/genética , Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Humanos , Pulmão/metabolismo , Camundongos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C beta/genética , Fosfolipase C beta/metabolismo , Fosfolipase C beta/fisiologia , Mucosa Respiratória/metabolismo
2.
FASEB J ; 35(3): e21375, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33559200

RESUMO

Host-pathogen interactions play an important role in defining the outcome of a disease. Recent studies have shown that the bacterial quorum sensing molecules (QSM) can interact with host cell membrane proteins, mainly G protein-coupled receptors (GPCRs), and induce innate immune responses. However, few studies have examined QSM-GPCR interactions and their influence on oral innate immune responses. In this study, we examined the role of bitter taste receptor T2R14 in sensing competence stimulating peptides (CSPs) secreted by cariogenic bacterium Streptococcus mutans and in mediating innate immune responses in gingival epithelial cells (GECs). Transcriptomic and western blot analyses identify T2R14 to be highly expressed in GECs. Our data show that only CSP-1 from S. mutans induces robust intracellular calcium mobilization compared to CSP-2 and CSP-3. By using CRISPR-Cas9, we demonstrate that CSP-1 induced calcium signaling and secretion of cytokines CXCL-8/IL-8, TNF-α, and IL-6 is mediated through T2R14 in GECs. Interestingly, the NF-kB signaling activated by CSP-1 in GECs was independent of T2R14. CSP-1-primed GECs attracted differentiated HL-60 immune cells (dHL-60) and this effect was abolished in T2R14 knock down GECs and also in cells primed with T2R14 antagonist 6-Methoxyflavone (6-MF). Our findings identify S. mutans CSP-1 as a peptide ligand for the T2R family. Our study establishes a novel host-pathogen interaction between cariogenic S. mutans CSP-1 and T2R14 in GECs leading to an innate immune response. Collectively, these findings suggest T2Rs as potential therapeutic targets to modulate innate immune responses upon oral bacterial infections.


Assuntos
Proteínas de Bactérias/fisiologia , Gengiva/imunologia , Interações Hospedeiro-Patógeno , Percepção de Quorum/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Streptococcus mutans/fisiologia , Cálcio/metabolismo , Linhagem Celular , Movimento Celular , Citocinas/biossíntese , Células Epiteliais/imunologia , Gengiva/citologia , Humanos , Imunidade Inata , NF-kappa B/fisiologia , Fosfolipase C beta/fisiologia
3.
Nat Immunol ; 10(9): 936-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19692992

RESUMO

Like every metazoan species hosting a gut flora, drosophila tolerate commensal microbiota yet remain able to mount an efficient immune response to food-borne pathogens. New findings explain how the quantity of reactive oxygen species in the gut is 'tuned' to microbial burden and how intestinal immune homeostasis is thereby maintained


Assuntos
Drosophila melanogaster/imunologia , Imunidade Inata , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Proteínas de Drosophila/fisiologia , Fosfatase 6 de Especificidade Dupla/fisiologia , Homeostase , Intestinos/imunologia , Intestinos/microbiologia , NADPH Oxidases/fisiologia , Fosfolipase C beta/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
4.
Nat Immunol ; 10(9): 949-57, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19668222

RESUMO

All metazoan guts are in permanent contact with the microbial realm. However, understanding of the exact mechanisms by which the strength of gut immune responses is regulated to achieve gut-microbe mutualism is far from complete. Here we identify a signaling network composed of complex positive and negative mechanisms that controlled the expression and activity of dual oxidase (DUOX), which 'fine tuned' the production of microbicidal reactive oxygen species depending on whether the gut encountered infectious or commensal microbes. Genetic analyses demonstrated that negative and positive regulation of DUOX was required for normal host survival in response to colonization with commensal and infectious microbes, respectively. Thus, the coordinated regulation of DUOX enables the host to achieve gut-microbe homeostasis by efficiently combating infection while tolerating commensal microbes.


Assuntos
Drosophila/imunologia , NADPH Oxidases/fisiologia , Fator 2 Ativador da Transcrição/fisiologia , Animais , Células CACO-2 , Calcineurina/fisiologia , Proteínas de Transporte/fisiologia , Regulação Enzimológica da Expressão Gênica , Humanos , Intestinos/imunologia , Intestinos/microbiologia , MAP Quinase Quinase 3/fisiologia , MAP Quinase Quinase Quinase 1/fisiologia , NADPH Oxidases/genética , Fosfolipase C beta/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
5.
J Neurosci ; 35(6): 2530-46, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25673847

RESUMO

Drosophila phototransduction is a model system for the ubiquitous phosphoinositide signaling. In complete darkness, spontaneous unitary current events (dark bumps) are produced by spontaneous single Gqα activation, while single-photon responses (quantum bumps) arise from synchronous activation of several Gqα molecules. We have recently shown that most of the spontaneous single Gqα activations do not produce dark bumps, because of a critical phospholipase Cß (PLCß) activity level required for bump generation. Surpassing the threshold of channel activation depends on both PLCß activity and cellular [Ca(2+)], which participates in light excitation via a still unclear mechanism. We show here that in IP3 receptor (IP3R)-deficient photoreceptors, both light-activated Ca(2+) release from internal stores and light sensitivity were strongly attenuated. This was further verified by Ca(2+) store depletion, linking Ca(2+) release to light excitation. In IP3R-deficient photoreceptors, dark bumps were virtually absent and the quantum-bump rate was reduced, indicating that Ca(2+) release from internal stores is necessary to reach the critical level of PLCß catalytic activity and the cellular [Ca(2+)] required for excitation. Combination of IP3R knockdown with reduced PLCß catalytic activity resulted in highly suppressed light responses that were partially rescued by cellular Ca(2+) elevation, showing a functional cooperation between IP3R and PLCß via released Ca(2+). These findings suggest that in contrast to the current dogma that Ca(2+) release via IP3R does not participate in light excitation, we show that released Ca(2+) plays a critical role in light excitation. The positive feedback between PLCß and IP3R found here may represent a common feature of the inositol-lipid signaling.


Assuntos
Drosophila/fisiologia , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Fosfolipase C beta/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Animais , Animais Geneticamente Modificados , Sinalização do Cálcio/fisiologia , Eletrorretinografia , Hipóxia/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Luz , Masculino , Técnicas de Patch-Clamp , Células Fotorreceptoras de Invertebrados/efeitos da radiação , Interferência de RNA
6.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L747-58, 2016 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-26773068

RESUMO

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.


Assuntos
Flavonoides/farmacologia , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Fosfolipase C beta/antagonistas & inibidores , Animais , Aorta/efeitos dos fármacos , Aorta/fisiopatologia , Asma/tratamento farmacológico , Broncoconstrição/efeitos dos fármacos , Sinalização do Cálcio , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/química , Avaliação Pré-Clínica de Medicamentos , Flavonoides/química , Flavonóis , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Camundongos , Contração Muscular , Músculo Liso/fisiologia , Músculo Liso/fisiopatologia , Inibidores da Fosfodiesterase 4/química , Inibidores da Fosfodiesterase 4/farmacologia , Fosfolipase C beta/fisiologia
7.
BMC Cancer ; 15: 775, 2015 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-26497576

RESUMO

BACKGROUND: G proteins are known to modulate various growth signals and are implicated in the regulation of tumorigenesis. The tumor suppressor Fhit is a newly identified interaction partner of Gq proteins that typically stimulate the phospholipase C pathway. Activated Gαq subunits have been shown to interact directly with Fhit, up-regulate Fhit expression and enhance its suppressive effect on cell growth and migration. Other signaling molecules may be involved in modulating Gαq/Fhit interaction. METHODS: To test the relationship of PLCß with the interaction between Gαq and Fhit, co-immunoprecipication assay was performed on HEK293 cells co-transfected with different combinations of Flag-Fhit, Gα16, Gα16QL, pcDNA3 vector, and PLCß isoforms. Possible associations of Fhit with other effectors of Gαq were also demonstrated by co-immunoprecipitation. The regions of Gαq for Fhit interaction and PLCß stimulation were further evaluated by inositol phosphates accumulation assay using a series of Gα16/z chimeras with discrete regions of Gα16 replaced by those of Gαz. RESULTS: PLCß1, 2 and 3 interacted with Fhit regardless of the expression of Gαq. Expression of PLCß increased the affinities of Fhit for both wild-type and activated Gαq. Swapping of the Fhit-interacting α2-ß4 region of Gαq with Gαi eliminated the association of Gαq with Fhit without affecting the ability of the mutant to stimulate PLCß. Other effectors of Gαq including RGS2 and p63RhoGEF were unable to interact with Fhit. CONCLUSIONS: PLCß may participate in the regulation of Fhit by Gq in a unique way. PLCß interacts with Fhit and increases the interaction between Gαq and Fhit. The Gαq/PLCß/Fhit complex formation points to a novel signaling pathway that may negatively regulate tumor cell growth.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfolipase C beta/metabolismo , Western Blotting , Células HEK293 , Humanos , Imunoprecipitação , Fosfolipase C beta/fisiologia , Ligação Proteica , Transdução de Sinais/fisiologia , Regulação para Cima
8.
Mol Pharmacol ; 84(4): 488-500, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23880553

RESUMO

Phospholipase C (PLC) enzymes convert phosphatidylinositol-4,5-bisphosphate into the second messengers diacylglycerol and inositol-1,4,5-triphosphate. The production of these molecules promotes the release of intracellular calcium and activation of protein kinase C, which results in profound cellular changes. The PLCß subfamily is of particular interest given its prominent role in cardiovascular and neuronal signaling and its regulation by G protein-coupled receptors, as PLCß is the canonical downstream target of the heterotrimeric G protein Gαq. However, this is not the only mechanism regulating PLCß activity. Extensive structural and biochemical evidence has revealed regulatory roles for autoinhibitory elements within PLCß, Gßγ, small molecular weight G proteins, and the lipid membrane itself. Such complex regulation highlights the central role that this enzyme plays in cell signaling. A better understanding of the molecular mechanisms underlying the control of its activity will greatly facilitate the search for selective small molecule modulators of PLCß.


Assuntos
Fosfolipase C beta/química , Fosfolipase C beta/fisiologia , Animais , Humanos , Isoenzimas/química , Isoenzimas/fisiologia , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia
9.
Curr Top Microbiol Immunol ; 362: 235-45, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23086421

RESUMO

Myelodysplastic syndromes (MDS), clonal hematopoietic stem-cell disorders mainly affecting older adult patients, show ineffective hematopoiesis in one or more of the lineages of the bone marrow. A number of MDS progresses to acute myeloid leukemia (AML) with the involvement of genetic and epigenetic mechanisms affecting PI-PLC ß1. The molecular mechanisms underlying the MDS evolution to AML are still unclear, even though it is now clear that the nuclear signaling elicited by PI-PLC ß1, Cyclin D3, and Akt plays an important role in the control of the balance between cell cycle progression and apoptosis in both normal and pathologic conditions. Moreover, a correlation between other PI-PLCs, such as PI-PLC ß3, kinases and phosphatases has been postulated in MDS pathogenesis. Here, we review the findings hinting at the role of nuclear lipid signaling pathways in MDS, which could become promising therapeutic targets.


Assuntos
Núcleo Celular/enzimologia , Síndromes Mielodisplásicas/etiologia , Fosfatidilinositóis/metabolismo , Fosfolipase C beta/fisiologia , Epigenômica , Humanos , Transdução de Sinais/fisiologia
10.
J Immunol ; 187(3): 1440-7, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21705626

RESUMO

The CXCR4 chemokine receptor is a G protein-coupled receptor that signals in T lymphocytes by forming a heterodimer with the TCR. CXCR4 and TCR functions are consequently highly cross regulated, affecting T cell immune activation, cytokine secretion, and T cell migration. The CXCR4-TCR heterodimer stimulates T cell migration and activation of the ERK MAPK and downstream AP-1-dependent cytokine transcription in response to stromal cell-derived factor-1 (SDF-1), the sole chemokine ligand of CXCR4. These responses require Gi-type G proteins as well as TCR ITAM domains and the ZAP70 tyrosine kinase, thus indicating that the CXCR4-TCR heterodimer signals to integrate G protein-coupled receptor-associated and TCR-associated signaling molecules in response to SDF-1. Yet, the phospholipase C (PLC) isozymes responsible for coupling the CXCR4-TCR heterodimer to distinct downstream cellular responses are incompletely characterized. In this study, we demonstrate that PLC activity is required for SDF-1 to induce ERK activation, migration, and CXCR4 endocytosis in human T cells. SDF-1 signaling via the CXCR4-TCR heterodimer uses PLC-ß3 to activate the Ras-ERK pathway and increase intracellular calcium ion concentrations, whereas PLC-γ1 is dispensable for these outcomes. In contrast, PLC-γ1, but not PLC-ß3, is required for SDF-1-mediated migration via a mechanism independent of LAT. These results increase understanding of the signaling mechanisms employed by the CXCR4-TCR heterodimer, characterize new roles for PLC-ß3 and PLC-γ1 in T cells, and suggest that multiple PLCs may also be activated downstream of other chemokine receptors to distinctly regulate migration versus other signaling functions.


Assuntos
Quimiocina CXCL12/fisiologia , Fosfolipase C beta/fisiologia , Fosfolipase C gama/fisiologia , Multimerização Proteica/imunologia , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores CXCR4/fisiologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Sinalização do Cálcio/imunologia , Movimento Celular/imunologia , Endocitose/imunologia , Humanos , Líquido Intracelular/enzimologia , Líquido Intracelular/imunologia , Isoenzimas/fisiologia , Células Jurkat , Sistema de Sinalização das MAP Quinases/imunologia , Receptores CXCR4/metabolismo , Subpopulações de Linfócitos T/enzimologia
11.
Blood ; 116(26): 6003-13, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20858858

RESUMO

Hyperactivation of the transcription factor Stat5 leads to various leukemias. Stat5 activity is regulated by the protein phosphatase SHP-1 in a phospholipase C (PLC)-ß3-dependent manner. Thus, PLC-ß3-deficient mice develop myeloproliferative neoplasm, like Lyn (Src family kinase)- deficient mice. Here we show that Lyn/PLC-ß3 doubly deficient lyn(-/-);PLC-ß3(-/-) mice develop a Stat5-dependent, fatal myelodysplastic/myeloproliferative neoplasm, similar to human chronic myelomonocytic leukemia (CMML). In hematopoietic stem cells of lyn(-/-);PLC-ß3(-/-) mice that cause the CMML-like disease, phosphorylation of SHP-1 at Tyr(536) and Tyr(564) is abrogated, resulting in reduced phosphatase activity and constitutive activation of Stat5. Furthermore, SHP-1 phosphorylation at Tyr(564) by Lyn is indispensable for maximal phosphatase activity and for suppression of the CMML-like disease in these mice. On the other hand, Tyr(536) in SHP-1 can be phosphorylated by Lyn and another kinase(s) and is necessary for efficient interaction with Stat5. Therefore, we identify a novel Lyn/PLC-ß3-mediated regulatory mechanism of SHP-1 and Stat5 activities.


Assuntos
Leucemia Mielomonocítica Crônica/metabolismo , Transtornos Mieloproliferativos/metabolismo , Fosfolipase C beta/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Fator de Transcrição STAT5/metabolismo , Quinases da Família src/fisiologia , Animais , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Inflamação/etiologia , Inflamação/patologia , Janus Quinase 2/metabolismo , Leucemia Mielomonocítica Crônica/etiologia , Leucemia Mielomonocítica Crônica/patologia , Pneumopatias/etiologia , Pneumopatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/patologia , Fenótipo , Fosforilação , Transdução de Sinais , Tirosina/metabolismo , Quinases da Família src/metabolismo
12.
Proc Natl Acad Sci U S A ; 106(51): 21912-7, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19955421

RESUMO

Absence seizures are characterized by cortical spike-wave discharges (SWDs) on electroencephalography, often accompanied by a shift in the firing pattern of thalamocortical (TC) neurons from tonic to burst firing driven by T-type Ca(2+) currents. We recently demonstrated that the phospholipase C beta4 (PLCbeta4) pathway tunes the firing mode of TC neurons via the simultaneous regulation of T- and L-type Ca(2+) currents, which prompted us to investigate the contribution of TC firing modes to absence seizures. PLCbeta4-deficient TC neurons were readily shifted to the oscillatory burst firing mode after a slight hyperpolarization of membrane potential. TC-limited knockdown as well as whole-animal knockout of PLCbeta4 induced spontaneous SWDs with simultaneous behavioral arrests and increased the susceptibility to drug-induced SWDs, indicating that the deletion of thalamic PLCbeta4 leads to the genesis of absence seizures. The SWDs were effectively suppressed by thalamic infusion of a T-type, but not an L-type, Ca(2+) channel blocker. These results reveal a primary role of TC neurons in the genesis of absence seizures and provide strong evidence that an alteration of the firing property of TC neurons is sufficient to generate absence seizures. Our study presents PLCbeta4-deficient mice as a potential animal model for absence seizures.


Assuntos
Epilepsia Tipo Ausência/enzimologia , Fosfolipase C beta/fisiologia , Tálamo/fisiopatologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Eletroencefalografia , Ativadores de Enzimas/farmacologia , Epilepsia Tipo Ausência/fisiopatologia , Agonistas GABAérgicos/farmacologia , Inativação Gênica , Potenciais da Membrana , Camundongos , Camundongos Knockout , Fosfolipase C beta/genética , Tálamo/enzimologia
13.
J Neurosci ; 30(43): 14361-5, 2010 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-20980592

RESUMO

Many neurons in the nervous systems express afterhyperpolarizations that are mediated by a slow calcium-activated potassium current. This current shapes neuronal firing and is inhibited by neuromodulators, suggesting an important role in the regulation of neuronal function. Surprisingly, very little is currently known about the molecular basis for this current or how it is gated by calcium. Recently, the neuronal calcium sensor protein hippocalcin was identified as a calcium sensor for the slow afterhyperpolarizing current in the hippocampus. However, while hippocalcin is very strongly expressed in the hippocampus, this protein shows a relatively restricted distribution in the brain. Furthermore, the genetic deletion of this protein only partly reduces the slow hyperpolarizing current in hippocampus. These considerations question whether hippocalcin can be the sole calcium sensor for the slow afterhyperpolarizing current. Here we use loss of function and overexpression strategies to show that hippocalcin functions as a calcium sensor for the slow afterhyperpolarizing current in the cerebral cortex, an area where hippocalcin is expressed at much lower levels than in hippocampus. In addition we show that neurocalcin δ, but not VILIP-2, can also act as a calcium sensor for the slow afterhyperpolarizing current. Finally we show that hippocalcin and neurocalcin δ both increase the calcium sensitivity of the afterhyperpolarizing current but do not alter its sensitivity to inhibition by carbachol acting through the Gαq-11-PLCß signaling cascade. These results point to a general role for a subgroup of visinin-like neuronal calcium sensor proteins in the activation of the slow calcium-activated afterhyperpolarizing current.


Assuntos
Sinalização do Cálcio/fisiologia , Córtex Cerebral/fisiologia , Hipocalcina/biossíntese , Neurocalcina/fisiologia , Animais , Carbacol/farmacologia , Eletrofisiologia , Eletroporação , Hipocalcina/genética , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Agonistas Muscarínicos/farmacologia , Fosfolipase C beta/fisiologia , Células Piramidais/fisiologia , Ratos , Transdução de Sinais/fisiologia
14.
Oncogene ; 40(4): 806-820, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33262460

RESUMO

Uveal melanoma (UM) is a currently untreatable form of melanoma with a 50% mortality rate. Characterization of the essential signaling pathways driving this cancer is critical to develop target therapies. Activating mutations in the Gαq signaling pathway at the level of GNAQ, GNA11, or rarely CYSLTR2 or PLCß4 are considered alterations driving proliferation in UM and several other neoplastic disorders. Here, we systematically examined the oncogenic signaling output of various mutations recurrently identified in human tumors. We demonstrate that CYSLTR2 → GNAQ/11 → PLCß act in a linear signaling cascade that, via protein kinase C (PKC), activates in parallel the MAP-kinase and FAK/Yes-associated protein pathways. Using genetic ablation and pharmacological inhibition, we show that the PKC/RasGRP3/MAPK signaling branch is the essential component that drives the proliferation of UM. Only inhibition of the MAPK branch but not the FAK branch synergizes with inhibition of the proximal cascade, providing a blueprint for combination therapy. All oncogenic signaling could be extinguished by the novel GNAQ/11 inhibitor YM-254890, in all UM cells with driver mutation in the Gαq subunit or the upstream receptor. Our findings highlight the GNAQ/11 → PLCß â†’ PKC → MAPK pathway as the central signaling axis to be suppressed pharmacologically to treat for neoplastic disorders with Gαq pathway mutations.


Assuntos
Melanoma/genética , Oncogenes/fisiologia , Neoplasias Uveais/genética , Animais , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases , Melanoma/patologia , Camundongos , Mutação , Fosfolipase C beta/fisiologia , Proteína Quinase C/fisiologia , Receptores de Leucotrienos/fisiologia , Transdução de Sinais/fisiologia , Neoplasias Uveais/patologia
15.
Chem Senses ; 35(5): 363-73, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20233741

RESUMO

To clarify the role of phospholipase C (PLC) in insect olfactory transduction, we have undertaken its molecular identification in the moth Spodoptera littoralis. From the analysis of a male antennal expressed sequence tag library, we succeeded in cloning a full-length cDNA encoding a PLC that belongs to the cluster of PLC-beta subtypes. In adult males, the PLC-beta transcript was located predominantly in brain and antennae where its presence was detected in the olfactory sensilla trichodea. Moreover, PLC-beta was expressed in antennae at the beginning of the pupal stage, then reached a maximum at the end of this stage and was maintained at this level during the adult period. Taken together, these results provided molecular evidence for the putative participation of a PLC-beta in signaling pathways responsible for the establishment and the functioning of insect olfactory system.


Assuntos
Encéfalo/metabolismo , Condutos Olfatórios/metabolismo , Neurônios Receptores Olfatórios/fisiologia , Fosfolipase C beta/fisiologia , Spodoptera/enzimologia , Animais , DNA Complementar/genética , Masculino , Mariposas , Fosfolipase C beta/genética , Proteínas Recombinantes/farmacologia , Spodoptera/genética
16.
Can J Physiol Pharmacol ; 88(3): 331-40, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20393598

RESUMO

We have recently shown that A10 vascular smooth muscle cells (VSMCs) exposed to high glucose exhibited enhanced expression of G(alpha)q and PLCbeta proteins. Since high glucose has been reported to increase the levels of vasoactive peptides and oxidative stress, the present study was undertaken to investigate the implication of angiotensin II (Ang II), endothelin (ET)-1, and oxidative stress in the high glucose-induced enhanced expression of G(alpha)q/11 and PLCbeta proteins and associated signaling in A10 VSMCs. The levels of G(alpha)q, G(alpha)11, PLCbeta-1, and PLCbeta-2 proteins, as determined by Western blotting, were significantly higher in A10 VSMCs exposed to high glucose than in control cells. The elevated levels were restored to control values by the antioxidant diphenyleneiodonium (DPI), as well as by the antagonist of Ang II AT1 receptor losartan and the antagonists of ETA and ETB receptors BQ123 and BQ788, respectively. In addition, ET-1-stimulated production of inositol trisphosphate (IP3), which was enhanced by high glucose, was also restored toward control levels by DPI. Furthermore, the enhanced production of superoxide anion (O2-), increased NADPH oxidase activity, and enhanced expression of p22phox and p47phox proteins induced by high glucose were restored to control levels by losartan, BQ123, and BQ788. These results suggest that through increased oxidative stress, high glucose-induced enhanced levels of endogenous Ang II and ET-1 may contribute to the increased levels of G(alpha)q/11 and mediated signaling in A10 VSMCs.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Glucose/administração & dosagem , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/fisiologia , Angiotensina II/fisiologia , Animais , Linhagem Celular , Endotelina-1/biossíntese , Endotelina-1/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/biossíntese , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Fosfolipase C beta/biossíntese , Fosfolipase C beta/fisiologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
17.
Sci Rep ; 10(1): 8813, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483199

RESUMO

Sleep abnormality often accompanies the impairment of cognitive function. Both rapid eye movement (REM) and non-REM (NREM) sleep have associated with improved memory performance. However, the role of composition in NREM sleep, consisting of light and deep NREM, for memory formation is not fully understood. We investigated how the dynamics of NREM sleep states influence memory consolidation. Thalamocortical (TC) neuron-specific phospholipase C ß4 (PLCß4) knockout (KO) increased the total duration of NREM sleep, consisting of destabilized light NREM and stabilized deep NREM. Surprisingly, the longer NREM sleep did not improve memory consolidation but rather impaired it in TC-specific PLCß4 KO mice. Memory function was positively correlated with the stability of light NREM and spindle activity occurring in maintained light NREM period. Our study suggests that a single molecule, PLCß4, in TC neurons is critical for tuning the NREM sleep states and thus affects sleep-dependent memory formation.


Assuntos
Consolidação da Memória/fisiologia , Transtornos da Memória/enzimologia , Proteínas do Tecido Nervoso/fisiologia , Fosfolipase C beta/fisiologia , Fases do Sono/fisiologia , Tálamo/enzimologia , Animais , Córtex Cerebral/enzimologia , Condicionamento Clássico/fisiologia , Ritmo Delta/fisiologia , Eletroencefalografia , Eletromiografia , Éxons/genética , Comportamento Exploratório , Medo/fisiologia , Masculino , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Neurônios/enzimologia , Fosfolipase C beta/deficiência , Reconhecimento Psicológico , Deleção de Sequência , Sono de Ondas Lentas/fisiologia , Fatores de Tempo
18.
Life Sci ; 257: 118046, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32622948

RESUMO

Orexin-A is an endogenous peptide with receptors throughout the brain. According to some recent research, learning and memory are affected by the central administration of orexin; however, no study so far has investigated the long-term inhibition of the orexinergic system. The present study has evaluated the effect of pretraining administration of orexin 1 receptor (OXR1) antagonist, SB-334867, on the acquisition of memory. The Morris water maze (MWM) task was used for training and trial purposes in all groups. Memory performance was analyzed by measuring escape latency, traveled distance, and time spent in the target quadrant. Moreover, the effect of SB-334867 on phospholipase Cß3 (PLCß3) levels in the CA1 region of hippocampus slices was examined. Hippocampus slices were prepared using an immunohistochemistry (IHC) approach. SB-334867 (20 mg/kg) increased escape latency in SB-treated rats compared to SB-vehicle group (P < 0.01). SB-treated rats spent less time in the target quadrant compared to the SB-vehicle group (P < 0.001). Distance traveled in the target quadrant was significantly more in SB-treated rats compared to the SB-vehicle group (P < 0.001). Furthermore, SB-334867 decreased PLCß3 levels in the CA1 of the hippocampus (P < 0.01 and P < 0.05, respectively). Put together, our results suggest that the long-term inhibition of OXR1 plays a prominent role in spatial learning and memory, probably by attenuating PLCß3 in CA1 neurons.


Assuntos
Memória/efeitos dos fármacos , Memória/fisiologia , Fosfolipase C beta/metabolismo , Animais , Benzoxazóis/farmacologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Naftiridinas/farmacologia , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Fosfolipase C beta/fisiologia , Ratos , Ratos Wistar , Ureia/análogos & derivados , Ureia/farmacologia
19.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118649, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31954103

RESUMO

GSK-3 and PLCbeta enzymes are responsible for the regulation of several signalling pathways related to many cellular functions. In hematopoietic cells, GSK-3 deficiency is correlated with an MDS-like phenotype and with leukemogenesis, showing a prognostic potential in AML cells. GSK-3 interacts with Wnt or MAPK signalling, but it is also linked to PI3K/Akt/mTOR pathways to regulate cell proliferation and apoptosis of hematopoietic stem cell progenitors. PLCbeta enzymes are involved in cell cycle progression of hematopoietic, MDS/AML and immune cells, through activation of PKC or calcium signalling. Of note, a PLCbeta1/PKCalpha pathway is modulated during MDS pathogenesis, with a specific involvement of the inositides localized in the nucleus. Here we focus on GSK-3 and PLCbeta signalling, describing the many evidences that underline the pivotal role of both GSK-3 and PLCbeta-dependent pathways in MDS/AML, their association with therapy and their possible interactions.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Leucemia Mieloide Aguda/enzimologia , Síndromes Mielodisplásicas/enzimologia , Fosfolipase C beta/metabolismo , Transdução de Sinais , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/fisiologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Fosfolipase C beta/fisiologia
20.
J Mol Cell Cardiol ; 47(5): 676-83, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19729020

RESUMO

Atrial fibrillation (AF) is commonly associated with chronic dilatation of the left atrium, both in human disease and animal models. The immediate signaling enzyme phospholipase C (PLC) is activated by mechanical stretch to generate the Ca2+-releasing messenger inositol(1,4,5)trisphosphate (Ins(1,4,5)P3) and sn-1,2-diacylglycerol (DAG), an activator of protein kinase C subtypes. There is also evidence that heightened activity of PLC, caused by the receptor coupling protein Gq, can contribute to atrial remodelling. We examined PLC activation in right and left atrial appendage from patients with mitral valve disease (VHD) and in a mouse model of dilated cardiomyopathy caused by transgenic overexpression of the stress-activated protein kinase, mammalian sterile 20 like kinase 1 (Mst1) (Mst1-TG). PLC activation was heightened 6- to 10-fold in atria from VHD patients compared with right atrial tissue from patients undergoing coronary artery bypass surgery (CABG) and was also heightened in the dilated atria from Mst1-TG. PLC activation in human left atrial appendage and in mouse left atria correlated with left atrial size, implying a relationship between PLC activation and chronic dilatation. Dilated atria from human and mouse showed heightened expression of PLCbeta1b, but not of other PLC subtypes. PLCbeta1b, but not PLCbeta1a, caused apoptosis when overexpressed in neonatal rat cardiomyocytes, suggesting that PLCbeta1b may contribute to chamber dilatation. The activation of PLCbeta1b is a possible therapeutic target to limit atrial remodelling in VHD patients.


Assuntos
Cardiomiopatia Dilatada/enzimologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Fosfolipase C beta/fisiologia , Animais , Animais Recém-Nascidos , Apêndice Atrial/metabolismo , Apêndice Atrial/patologia , Fibrilação Atrial/enzimologia , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Células Cultivadas , Modelos Animais de Doenças , Átrios do Coração , Humanos , Técnicas In Vitro , Camundongos , Insuficiência da Valva Mitral/enzimologia , Insuficiência da Valva Mitral/patologia , Miócitos Cardíacos/metabolismo , Fosfolipase C beta/genética , Fosfolipase C beta/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA