Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell Biochem ; 479(3): 567-577, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37131040

RESUMO

Quinine, a bitter compound, can act as an agonist to activate the family of bitter taste G protein-coupled receptor family of proteins. Previous work from our laboratory has demonstrated that quinine causes activation of RalA, a Ras p21-related small G protein. Ral proteins can be activated directly or indirectly through an alternative pathway that requires Ras p21 activation resulting in the recruitment of RalGDS, a guanine nucleotide exchange factor for Ral. Using normal mammary epithelial (MCF-10A) and non-invasive mammary epithelial (MCF-7) cell lines, we investigated the effect of quinine in regulating Ras p21 and RalA activity. Results showed that in the presence of quinine, Ras p21 is activated in both MCF-10A and MCF-7 cells; however, RalA was inhibited in MCF-10A cells, and no effect was observed in the case of MCF-7 cells. MAP kinase, a downstream effector for Ras p21, was activated in both MCF-10A and MCF-7 cells. Western blot analysis confirmed the expression of RalGDS in MCF-10A cells and MCF-7 cells. The expression of RalGDS was higher in MCF-10A cells in comparison to the MCF-7 cells. Although RalGDS was detected in MCF-10A and MCF-7 cells, it did not result in RalA activation upon Ras p21 activation with quinine suggesting that the Ras p21-RalGDS-RalA pathway is not active in the MCF-10A cells. The inhibition of RalA activity in MCF-10A cells due to quinine could be as a result of a direct effect of this bitter compound on RalA. Protein modeling and ligand docking analysis demonstrated that quinine can interact with RalA through the R79 amino acid, which is located in the switch II region loop of the RalA protein. It is possible that quinine causes a conformational change that results in the inhibition of RalA activation even though RalGDS is present in the cell. More studies are needed to elucidate the mechanism(s) that regulate Ral activity in mammary epithelial cells.


Assuntos
Quinina , Fator ral de Troca do Nucleotídeo Guanina , Fator ral de Troca do Nucleotídeo Guanina/metabolismo , Quinina/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Epiteliais/metabolismo
2.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-34576085

RESUMO

Bitter-taste receptors (T2Rs) have emerged as key players in host-pathogen interactions and important modulators of oral innate immunity. Previously, we reported that T2R14 is expressed in gingival epithelial cells (GECs) and interacts with competence stimulating peptides (CSPs) secreted by the cariogenic Streptococcus mutans. The underlying mechanisms of the innate immune responses and physiological effects of T2R14 on Gram-positive bacteria are not well characterized. In this study, we examined the role of T2R14 in internalization and growth inhibitory effects on Gram-positive bacteria, namely Staphylococcus aureus and S. mutans. We utilized CRISPR-Cas9 T2R14 knockdown (KD) GECs as the study model to address these key physiological mechanisms. Our data reveal that the internalization of S. aureus is significantly decreased, while the internalization of S. mutans remains unaffected upon knockdown of T2R14 in GECs. Surprisingly, GECs primed with S. mutans CSP-1 resulted in an inhibition of growth for S. aureus, but not for S. mutans. The GECs infected with S. aureus induced T2R14-dependent human ß-defensin-2 (hBD-2) secretion; however, S. mutans-infected GECs did not induce hBD-2 secretion, but induced T2R14 dependent IL-8 secretion. Interestingly, our results show that T2R14 KD affects the cytoskeletal reorganization in GECs, thereby inhibiting S. aureus internalization. Our study highlights the distinct mechanisms and a direct role of T2R14 in influencing physiological responses to Gram-positive bacteria in the oral cavity.


Assuntos
Endocitose , Células Epiteliais/metabolismo , Gengiva/citologia , Bactérias Gram-Positivas/metabolismo , Viabilidade Microbiana , Receptores Acoplados a Proteínas G/metabolismo , Paladar , Actinas/metabolismo , Linhagem Celular , Células Epiteliais/ultraestrutura , Humanos , Interleucina-8/metabolismo , Modelos Biológicos , Nitratos/metabolismo , Nitritos/metabolismo , Staphylococcus aureus/metabolismo , Streptococcus mutans/metabolismo , beta-Defensinas/metabolismo , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
Mol Cell Biochem ; 426(1-2): 129-136, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27873143

RESUMO

Rac1 belongs to the Rho family of small GTPases and regulates actin cytoskeleton reorganization. T2R4 is a bitter taste receptor belonging to the G protein-coupled receptor family of proteins. In addition to mediating bitter taste perception from the tongue, T2R4s are found in extra-oral tissues, e.g., nasal epithelium, airways, brain, testis suggesting a much broader physiological function for these receptors. Anti-malarial drug and a bitter tasting compound, quinine, is a known agonist for T2R4, whereas BCML (Nα,Nα-Bis(carboxymethyl)-L-lysine) acts as an inverse agonist. Using western blot and Ca++ mobilization assays, the effects of quinine on Rac1 activity in HEK293T cells stably expressing T2R4/Gα16/44, T2R4, or Gα16/44 and transiently transfected with HA-Rac1 were investigated. Quinine treatment caused a significant reduction in the amount of active Rac1, whereas in the presence of BCML, quinine failed to cause any significant change in active Rac1. No significant change in Rac1 activity was observed in BAPTA-AM plus quinine-treated Gα16/44 cells, suggesting possibility of a pathway in addition to the canonical Ca++-dependent pathway. A noticeable role for Gα16/44 independent of T2R4 is observed in quinine-mediated Rac1 inactivation. Further, a significant difference in quinine-induced Ca++ response in T2R4/Gα16/44 or T2R4 cells was observed validating the partial role of calcium and importance of Gα16/44. This study is the first to show an inhibitory downstream action of a T2R4 agonist on Rac1 function. Further investigation will help in better understanding the downstream signal transduction network of T2R4 and its extra-oral physiological roles.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Quinina/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Proteínas rac1 de Ligação ao GTP/genética
4.
Mol Cell Biochem ; 426(1-2): 137-147, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28012014

RESUMO

The 25 bitter taste receptors (T2Rs) in humans perform a chemosensory function. However, very little is known about the level of expression of these receptors in different tissues. In this study, using nCounter gene expression we analyzed the expression patterns of human TAS2R transcripts in cystic fibrosis bronchial epithelial (CuFi-1), normal bronchial epithelial (NuLi-1), airway smooth muscle (ASM), pulmonary artery smooth muscle (PASM), mammary epithelial, and breast cancer cells. Our results suggest a specific pattern of TAS2R expression with TAS2R3, 4, 5, 10, 13, 19, and 50 transcripts expressed at moderate levels and TAS2R14 and TAS2R20 (or TASR49) at high levels in the various tissues analyzed. This pattern of expression is mostly independent of tissue origin and the pathological state, except in cancer cells. To elucidate the expression at the protein level, we pursued flow cytometry analysis of select T2Rs from CuFi-1 and NuLi-1 cells. The expression levels observed at the gene level by nCounter analysis correlate with the protein levels for the T2Rs analyzed. Next, to assess the functionality of the expressed T2Rs in these cells, we pursued functional assays measuring intracellular calcium mobilization after stimulation with the bitter compound quinine. Using PLC inhibitor, U-73122, we show that the calcium mobilized in these cells predominantly takes place through the Quinine-T2R-Gαßγ-PLC pathway. This report will accelerate studies aimed at analyzing the pathophysiological function of T2Rs in different extraoral tissues.


Assuntos
Brônquios/metabolismo , Regulação da Expressão Gênica/fisiologia , Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Mucosa Respiratória/metabolismo , Sinalização do Cálcio/fisiologia , Humanos , Especificidade de Órgãos/fisiologia
5.
Biochim Biophys Acta ; 1858(9): 2081-2087, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27288892

RESUMO

Bitter taste perception in humans is believed to act as a defense mechanism against ingestion of potential toxic substances. Bitter taste is perceived by 25 distinct bitter taste receptors (T2Rs) which belong to the family of G protein-coupled receptors (GPCRs). In the overall context of the role of membrane lipids in GPCR function, we show here that T2R4, a representative member of the bitter taste receptor family, displays cholesterol sensitivity in its signaling function. In order to gain further insight into cholesterol sensitivity of T2R4, we mutated two residues Tyr114(3.59) and Lys117(3.62) present in the cholesterol recognition amino acid consensus (CRAC) motif in T2R4 with alanines. We carried out functional characterization of the mutants by calcium mobilization, followed by cholesterol depletion and replenishment. CRAC motifs in GPCRs have previously been implicated in preferential cholesterol association. Our analysis shows that the CRAC motif represents an intrinsic feature of bitter taste receptors and is conserved in 22 out of 25 human T2Rs. We further demonstrate that Lys117, an important CRAC residue, is crucial in the reported cholesterol sensitivity of T2R4. Interestingly, cholesterol sensitivity of T2R4 was observed at quinine concentrations in the lower mM range. To the best of our knowledge, our results represent the first report addressing the molecular basis of cholesterol sensitivity in the function of taste receptors.


Assuntos
Colesterol/metabolismo , Quinina/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Células CHO , Colesterol/genética , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Mutação de Sentido Incorreto , Receptores Acoplados a Proteínas G/genética
6.
PLoS One ; 11(5): e0156347, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27223611

RESUMO

Bitter taste is one of the five basic taste sensations which is mediated by 25 bitter taste receptors (T2Rs) in humans. The mechanism of bitter taste signal transduction is not yet elucidated. The cellular processes underlying T2R desensitization including receptor internalization, trafficking and degradation are yet to be studied. Here, using a combination of molecular and pharmacological techniques we show that T2R4 is not internalized upon agonist treatment. Pretreatment with bitter agonist quinine led to a reduction in subsequent quinine-mediated calcium responses to 35 ± 5% compared to the control untreated cells. Interestingly, treatment with different bitter agonists did not cause internalization of T2R4. Instead, quinine treatment led to a 2-fold increase in T2R4 cell surface expression which was sensitive to Brefeldin A, suggesting a novel pharmacochaperone activity of quinine. This phenomenon of chaperone activity of quinine was also observed for T2R7, T2R10, T2R39 and T2R46. Our results suggest that the observed action of quinine for these T2Rs is independent of its agonist activity. This study provides novel insights into the pharmacochaperone activity of quinine and possible mechanism of T2R desensitization, which is of fundamental importance in understanding the mechanism of bitter taste signal transduction.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Quinina/farmacologia , Receptores Acoplados a Proteínas G/genética , Brefeldina A/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Moleculares , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Paladar
7.
Int J Biochem Cell Biol ; 77(Pt B): 197-204, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27032752

RESUMO

Over the past decade tremendous progress has been made in understanding the functional role of bitter taste receptors (T2Rs) and bitter taste perception. This review will cover the recent advances made in identifying the role of T2Rs in pathophysiological states. T2Rs are widely expressed in various parts of human anatomy and have been shown to be involved in physiology of respiratory system, gastrointestinal tract and endocrine system. Empirical evidence has shown T2Rs to be an integral component of antimicrobial immune responses in upper respiratory tract infections. The studies on human airway smooth muscle cells have shown that a potent bitter tastant induced bronchodilatory effects mediated by bitter taste receptors. Clinical data suggests a role for T2R38 polymorphism in predisposition of individuals to chronic rhinosinusitis. The role of genetic variation in T2Rs and its impact on disease susceptibility have been investigated in various other disease risk factors such as alcohol dependence, head and neck cancers. Preliminary reports have demonstrated differential expression of functional T2Rs in breast cancer cell lines. Studies on the role of T2Rs in pathophysiology of diseases including chronic rhinosinusitis, asthma, cystic fibrosis, and cancer have been promising. However, research in this field is in its nascent stages, and more confirmatory studies on animal models and in clinical settings are required.


Assuntos
Doença , Receptores Acoplados a Proteínas G/metabolismo , Animais , Humanos , Receptores Acoplados a Proteínas G/química
8.
Int J Biochem Cell Biol ; 77(Pt B): 184-96, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26995065

RESUMO

Bitter taste receptors (T2Rs) belong to the super family of G protein-coupled receptors (GPCRs). There are 25 T2Rs expressed in humans, and these interact with a large and diverse group of bitter ligands. T2Rs are expressed in many extra-oral tissues and can perform diverse physiological roles. Structure-function studies led to the identification of similarities and dissimilarities between T2Rs and Class A GPCRs including amino acid conservation and novel motifs. However, the efficacy of most of the T2R ligands is not yet elucidated and the biochemical pharmacology of T2Rs is poorly understood. Recent studies on T2Rs characterized novel ligands including blockers for these receptors that include inverse agonist and antagonists. In this review we discuss the techniques used for elucidating bitter blockers, concept of ligand bias, generic amino acid numbering, the role of cholesterol, and conserved water molecules in the biochemistry and pharmacology of T2Rs.


Assuntos
Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Percepção Gustatória/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Humanos , Ligantes , Receptores Acoplados a Proteínas G/genética
9.
Methods Enzymol ; 556: 267-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25857786

RESUMO

G protein-coupled receptors (GPCRs) are cell surface proteins and play crucial role in mediating effective communication between extracellular and intracellular milieu of the cell. To understand the structure and function of these membrane proteins, it is imperative to express the proteins in a functional form and in sufficient quantities. However, heterologous expression of GPCRs in sufficient amounts for structural studies is a daunting task and over the years researchers have tried various expression systems to achieve this goal. Here, we describe a method developed in the laboratory of Dr. Har Gobind Khorana and successfully used over the past decade to express GPCRs and other membrane proteins in mammalian cells. Improvements to this method include use of genes codon optimized for expression in mammalian cells, use of expression tags at N-terminus, and affinity tags at the N-terminus and C-terminus of GPCRs. It also provides details for the process of selection for stably transfected clones, and to identify clones that are effectively expressing the GPCR of interest on the cell surface using flow cytometry. Purification of the overexpressed GPCR involves use of affinity chromatography. The method discussed here can also be effectively used to express cytotoxic proteins such as some constitutively active GPCRs. The method is robust and reliable to express GPCRs.


Assuntos
Clonagem Molecular/métodos , Receptores Acoplados a Proteínas G/genética , Animais , Linhagem Celular , Códon , Expressão Gênica , Genes Sintéticos , Células HEK293 , Humanos , Receptores Acoplados a Proteínas G/isolamento & purificação , Transfecção/métodos
10.
Biochim Biophys Acta ; 1848(7): 1502-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25858111

RESUMO

Bitter taste, in humans, is sensed by 25 G protein-coupled receptors, referred to as bitter taste receptors (T2Rs). The diverse roles of T2Rs in various extraoral tissues have implicated them as a potential target for therapeutic intervention. Structure-function studies have provided insights into the role of transmembrane and loop regions in the activation mechanism of T2Rs. However, studies aimed at deciphering the role of their carboxyl-terminus (C-terminus) are limited. In this study, we identified a KLK/R motif in the C-terminus that is conserved in 19 of the 25 T2Rs. Using site-directed mutagenesis we studied the role of 16 residues in the C-terminus of T2R4. The C-terminus of T2R4 is polybasic with 6 of the 16 residues consisting of lysines, constituting two separate KK motifs. We analyzed the effect of the C-terminus mutations on plasma membrane trafficking, and characterized their function in response to the T2R4 agonist quinine. The majority of the mutants showed defective receptor trafficking with ≤50% expression on the cell surface. Interestingly, mutation of the distal Lys296 of the KLK motif in T2R4 resulted in constitutive activity. The K296A mutant displayed five-fold basal activity over wild type T2R4, while the conservative substitution K296R showed wild type characteristics. The Lys294, Leu295 and Lys296 of the KLK motif in T2R4 were found to perform crucial roles, both, in receptor trafficking and function. Results from this study provide unique mechanistic insights into the structure-function role of the C-terminus in T2R signaling.


Assuntos
Motivos de Aminoácidos/genética , Mutação , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Sequência de Aminoácidos , Citometria de Fluxo , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
11.
Biochemistry ; 54(16): 2622-31, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25844797

RESUMO

Bitter taste receptors (T2Rs) belong to the G protein-coupled receptor superfamily. In humans, 25 T2Rs mediate bitter taste sensation. In addition to the oral cavity, T2Rs are expressed in many extraoral tissues, including the central nervous system, respiratory system, and reproductive system. To understand the mechanistic roles of the T2Rs in oral and extraoral tissues, novel blockers or antagonists are urgently needed. Recently, we elucidated the binding pocket of T2R4 for its agonist quinine, and an antagonist and inhibitory neurotransmitter, γ-aminobutyric acid. This structure-function information about T2R4 led us to screen the plant hormone abscisic acid (ABA), its precursor (xanthoxin), and catabolite phaseic acid for their ability to bind and activate or inhibit T2R4. Molecular docking studies followed by functional assays involving calcium imaging confirmed that ABA is an antagonist with an IC50 value of 34.4 ± 1.1 µM. However, ABA precursor xanthoxin acts as an agonist on T2R4. Interestingly, molecular model-guided site-directed mutagenesis suggests that the T2R4 residues involved in quinine binding are also predominantly involved in binding to the novel antagonist, ABA. The antagonist ability of ABA was tested using another T2R4 agonist, yohimbine. Our results suggest that ABA does not inhibit yohimbine-induced T2R4 activity. The discovery of natural bitter blockers has immense nutraceutical and physiological significance and will help in dissecting the T2R molecular pathways in various tissues.


Assuntos
Ácido Abscísico/química , Modelos Moleculares , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/química , Sítios de Ligação , Carotenoides/química , Humanos , Mutagênese Sítio-Dirigida , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sesquiterpenos/química , Relação Estrutura-Atividade , Percepção Gustatória/fisiologia , Ioimbina/química
12.
PLoS One ; 9(10): e110373, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25340739

RESUMO

Activation of bitter taste receptors (T2Rs) in human airway smooth muscle cells leads to muscle relaxation and bronchodilation. This finding led to our hypothesis that T2Rs are expressed in human pulmonary artery smooth muscle cells and might be involved in regulating the vascular tone. RT-PCR was performed to reveal the expression of T2Rs in human pulmonary artery smooth muscle cells. Of the 25 T2Rs, 21 were expressed in these cells. Functional characterization was done by calcium imaging after stimulating the cells with different bitter agonists. Increased calcium responses were observed with most of the agonists, the largest increase seen for dextromethorphan. Previously in site-directed mutational studies, we have characterized the response of T2R1 to dextromethorphan, therefore, T2R1 was selected for further analysis in this study. Knockdown with T2R1 specific shRNA decreased mRNA levels, protein levels and dextromethorphan-induced calcium responses in pulmonary artery smooth muscle cells by up to 50%. To analyze if T2Rs are involved in regulating the pulmonary vascular tone, ex vivo studies using pulmonary arterial and airway rings were pursued. Myographic studies using porcine pulmonary arterial and airway rings showed that stimulation with dextromethorphan led to contraction of the pulmonary arterial and relaxation of the airway rings. This study shows that dextromethorphan, acting through T2R1, causes vasoconstrictor responses in the pulmonary circuit and relaxation in the airways.


Assuntos
Dextrometorfano/farmacologia , Pulmão/irrigação sanguínea , Artéria Pulmonar/patologia , Receptores Acoplados a Proteínas G/metabolismo , Paladar/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Animais , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Técnicas In Vitro , Inositol 1,4,5-Trifosfato/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miografia , Cadeias Leves de Miosina/metabolismo , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxidos/metabolismo , Sus scrofa
13.
J Biol Chem ; 289(36): 25054-66, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25059668

RESUMO

In humans, the 25 bitter taste receptors (T2Rs) are activated by hundreds of structurally diverse bitter compounds. However, only five antagonists or bitter blockers are known. In this study, using molecular modeling guided site-directed mutagenesis, we elucidated the ligand-binding pocket of T2R4. We found seven amino acids located in the extracellular side of transmembrane 3 (TM3), TM4, extracellular loop 2 (ECL2), and ECL3 to be involved in T2R4 binding to its agonist quinine. ECL2 residues Asn-173 and Thr-174 are essential for quinine binding. Guided by a molecular model of T2R4, a number of amino acid derivatives were screened for their ability to bind to T2R4. These predictions were tested by calcium imaging assays that led to identification of γ-aminobutryic acid (GABA) and Nα,Nα-bis(carboxymethyl)-L-lysine (BCML) as competitive inhibitors of quinine-activated T2R4 with an IC50 of 3.2 ± 0.3 µM and 59 ± 18 nM, respectively. Interestingly, pharmacological characterization using a constitutively active mutant of T2R4 reveals that GABA acts as an antagonist, whereas BCML acts as an inverse agonist on T2R4. Site-directed mutagenesis confirms that the two novel bitter blockers share the same orthosteric site as the agonist quinine. The signature residues Ala-90 and Lys-270 play important roles in interacting with BCML and GABA, respectively. This is the first report to characterize a T2R endogenous antagonist and an inverse agonist. The novel bitter blockers will facilitate physiological studies focused on understanding the roles of T2Rs in extraoral tissues.


Assuntos
Aminoácidos/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Paladar , Alanina/química , Alanina/genética , Alanina/metabolismo , Aminoácidos/química , Aminoácidos/genética , Asparagina/química , Asparagina/genética , Asparagina/metabolismo , Sítios de Ligação/genética , GABAérgicos/química , GABAérgicos/farmacologia , Células HEK293 , Humanos , Lisina/química , Lisina/genética , Lisina/farmacologia , Modelos Moleculares , Estrutura Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Quinina/química , Quinina/farmacologia , Receptores Acoplados a Proteínas G/química , Treonina/química , Treonina/genética , Treonina/metabolismo , Ácido gama-Aminobutírico/química , Ácido gama-Aminobutírico/farmacologia
14.
Adv Pharmacol ; 70: 303-26, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24931200

RESUMO

G protein-coupled receptors (GPCRs) play a vital role in transmitting an extracellular stimuli or signal into an intracellular response in various cells. In some scenarios, GPCRs or their mutants can also signal in the absence of an agonist or an external stimulus, referred to as basal or constitutive activity, and those mutants are termed constitutively active mutants (CAMs). Bitter taste is one of the five basic tastes and is mediated by bitter taste receptors (T2Rs), which belong to the GPCR superfamily. The 25 T2Rs present in humans do not belong to any of the major GPCR classes, and their classification is ambiguous. The characterization of T2Rs in many extraoral tissues including the airways and upper respiratory tract, where they were shown to cause bronchodilation and influence host susceptibility to infection, underscores the therapeutic relevance of these receptors. Recent structure-function and pharmacological studies on T2Rs led to the identification of CAMs. In this review, we summarize the major findings on constitutive activity of T2Rs and their diverse roles. We discuss the usefulness of the T2R CAMs in terms of the discovery of bitter taste blockers, elucidating the mechanisms of T2R activation and dissecting the physiological pathways.


Assuntos
Mutação/genética , Receptores Acoplados a Proteínas G/genética , Paladar , Sequência de Aminoácidos , Bioensaio , Sequência Conservada , Humanos , Dados de Sequência Molecular , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo
15.
PLoS One ; 9(1): e85937, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465800

RESUMO

G protein-coupled receptors (GPCRs) show some level of basal activity even in the absence of an agonist, a phenomenon referred to as constitutive activity. Such constitutive activity in GPCRs is known to have important pathophysiological roles in human disease. The thromboxane A2 receptor (TP) is a GPCR that promotes thrombosis in response to binding of the prostanoid, thromboxane A2. TP dysfunction is widely implicated in pathophysiological conditions such as bleeding disorders, hypertension and cardiovascular disease. Recently, we reported the characterization of a few constitutively active mutants (CAMs) in TP, including a genetic variant A160T. Using these CAMs as reporters, we now test the inverse agonist properties of known antagonists of TP, SQ 29,548, Ramatroban, L-670596 and Diclofenac, in HEK293T cells. Interestingly, SQ 29,548 reduced the basal activity of both, WT-TP and the CAMs while Ramatroban was able to reduce the basal activity of only the CAMs. Diclofenac and L-670596 showed no statistically significant reduction in basal activity of WT-TP or CAMs. To investigate the role of these compounds on human platelet function, we tested their effects on human megakaryocyte based system for platelet activation. Both SQ 29,548 and Ramatroban reduced the platelet hyperactivity of the A160T genetic variant. Taken together, our results suggest that SQ 29,548 and Ramatroban are inverse agonists for TP, whereas, L-670596 and Diclofenac are neutral antagonists. Our findings have important therapeutic applications in the treatment of TP mediated pathophysiological conditions.


Assuntos
Carbazóis/farmacologia , Hidrazinas/farmacologia , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Sulfonamidas/farmacologia , Substituição de Aminoácidos , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes , Sinalização do Cálcio/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Ácidos Graxos Insaturados , Células HEK293 , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Mutagênese Sítio-Dirigida , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo
16.
PLoS One ; 8(9): e76481, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086743

RESUMO

G protein-coupled receptors (GPCRs) exhibit some level of basal signaling even in the absence of a bound agonist. This basal or constitutive signaling can have important pathophysiological roles. In the past few years, a number of high resolution crystal structures of GPCRs have been reported, including two crystal structures of constitutively active mutants (CAM) of the dim-light receptor, rhodopsin. The structural characterizations of CAMs are impeded by the lack of proper expression systems. The thromboxane A2 receptor (TP) is a GPCR that mediates vasoconstriction and promotes thrombosis in response to the binding of thromboxane. Here, we report on the expression and purification of a genetic variant and CAM in TP, namely A160T, using tetracycline-inducible HEK293S-TetR and HEK293S (GnTI¯)-TetR cell lines. Expression of the TP and the A160T genes in these mammalian cell lines resulted in a 4-fold increase in expression to a level of 15.8 ±0.3 pmol of receptor/mg of membrane protein. The receptors expressed in the HEK293S (GnTI(-))-TetR cell line showed homogeneous glycosylation. The functional yield of the receptors using a single step affinity purification was 45 µg/106 cells. Temperature- dependent secondary structure changes of the purified TP and A160T receptors were characterized using circular dichroism (CD) spectropolarimetry. The CD spectra shows that the loss of activity or thermal sensitivity that was previously observed for the A160T mutant, is not owing to large unfolding of the protein but rather to a more subtle effect. This is the first study to report on the successful high-level expression, purification, and biophysical characterization of a naturally occurring, diffusible ligand activated GPCR CAM.


Assuntos
Engenharia Genética/métodos , Polimorfismo Genético , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Sequência de Aminoácidos , Detergentes/farmacologia , Expressão Gênica , Células HEK293 , Humanos , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Receptores de Tromboxano A2 e Prostaglandina H2/química , Receptores de Tromboxano A2 e Prostaglandina H2/isolamento & purificação , Tetraciclina/farmacologia
17.
PLoS One ; 7(8): e42975, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22905193

RESUMO

Rac1, a member of the Rho family of small GTPases, has been shown to promote formation of lamellipodia at the leading edge of motile cells and affect cell migration. We previously demonstrated that calmodulin can bind to a region in the C-terminal of Rac1 and that this interaction is important in the activation of platelet Rac1. Now, we have analyzed amino acid residue(s) in the Rac1-calmodulin binding domain that are essential for the interaction and assessed their functional contribution in Rac1 activation. The results demonstrated that region 151-164 in Rac1 is essential for calmodulin binding. Within the 151-164 region, positively-charged amino acids K153 and R163 were mutated to alanine to study impact on calmodulin binding. Mutant form of Rac1 (K153A) demonstrated significantly reduced binding to calmodulin while the double mutant K153A/R163A demonstrated complete lack of binding to calmodulin. Thrombin or EGF resulted in activation of Rac1 in CHRF-288-11 or HeLa cells respectively and W7 inhibited this activation. Immunoprecipitation studies demonstrated that higher amount of CaM was associated with Rac1 during EGF dependent activation. In cells expressing mutant forms of Rac1 (K153A or K153A/R163A), activation induced by EGF was significantly decreased in comparison to wild type or the R163A forms of Rac1. The lack of Rac1 activation in mutant forms was not due to an inability of GDP-GTP exchange or a change in subcelllular distribution. Moreover, Rac1 activation was decreased in cells where endogenous level of calmodulin was reduced using shRNA knockdown and increased in cells where calmodulin was overexpressed. Docking analysis and modeling demonstrated that K153 in Rac1 interacts with Q41 in calmodulin. These results suggest an important role for calmodulin in the activation of Rac1 and thus, in cytoskeleton reorganization and cell migration.


Assuntos
Calmodulina/química , Proteínas rac1 de Ligação ao GTP/química , Animais , Calmodulina/metabolismo , Bovinos , Movimento Celular , Citoesqueleto/metabolismo , Ativação Enzimática , Fator de Crescimento Epidérmico/metabolismo , Escherichia coli/metabolismo , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Mutagênese , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Trombina/química , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
Mol Cell Biochem ; 353(1-2): 73-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21404019

RESUMO

Rac1 and Cdc42 are members of the Rho family of small GTPases and have been shown to promote the formation of lamellipodia and filopodia at the leading edge of motile cells and affect cell migration. In this study the authors have investigated the activation of Rac1 and Cdc42 by thrombin or collagen using the megakaryocytic cell line, CHRF-288-11. Maximal activation of Rac1 by thrombin or collagen was observed at 3 and 1 min, respectively. Similar results were obtained for the activation of Cdc42 by thrombin or collagen. The activation of Rac1 by thrombin and collagen was inhibited by the calmodulin-specific inhibitor, W7. However, W7 had no effect on the activation of Cdc42 by thrombin or collagen. The less potent calmodulin inhibitor, W5, did not have any effect on Rac1 or Cdc42 activation by thrombin or collagen. Transient over-expression of calmodulin in CHRF cells increased the basal and thrombin-mediated activation of Rac1 when compared to control but had no effect on the basal and thrombin-mediated activation of Cdc42 when compared to control. The results suggest that calmodulin regulates the activation of Rac1 in CHRF-288-11 cells in a manner similar to that in the human platelets.


Assuntos
Colágeno/farmacologia , Trombina/farmacologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Calmodulina/genética , Calmodulina/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Hemostáticos/farmacologia , Humanos , Megacariócitos/metabolismo , Megacariócitos/patologia , Sulfonamidas/farmacologia , Fatores de Tempo , Transfecção
19.
Biochem Biophys Res Commun ; 383(4): 401-5, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19379713

RESUMO

Previously, we have shown that RalA, a calmodulin (CaM)-binding protein, binds to the C2 region in the C-terminal of PLC-delta1, and increases its enzymatic activity. Since PLC-delta1 contains a CaM-like region in its N-terminus, we have investigated if RalA can also bind to the N-terminus of PLC-delta1. Therefore, we created a GST-PLC-delta1 construct consisting of the first 294 amino acids of PLC-delta1 (GST-PLC-delta1(1-294)). In vitro binding experiments confirmed that PLC-delta1(1-294) was capable of binding directly to RalA. W-7 coupled to polyacrylamide beads bound pure PLC-delta1, demonstrating that PLC-delta1 contains a CaM-like region. Competition assays with W-7, peptides representing RalA and the newly identified RalB CaM-binding regions, or the IQ peptide from PLC-delta1 were able to inhibit RalA binding to PLC-delta1(1-294). This study demonstrates that there are two binding sites for RalA in PLC-delta1 and provides further insight into the role of Ral GTPase in the regulation of PLC-delta1 function.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Fosfolipase C delta/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Calmodulina/metabolismo , Proteínas de Ligação a Calmodulina/genética , Bovinos , Células HeLa , Humanos , Dados de Sequência Molecular , Fosfolipase C delta/genética , Estrutura Terciária de Proteína , Ratos , Proteínas ral de Ligação ao GTP/genética
20.
Biochim Biophys Acta ; 1783(5): 770-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18328269

RESUMO

Rac1 and Cdc42 are members of the Rho family of small GTPases and have been shown to induce lamellipodia and filopodia formation, respectively. This leads to changes in cytoskeleton organization and as a consequence affects cell migration. In the present work we demonstrate that endogenous Rac1 and Cdc42 interact with calmodulin (CaM) in a Ca(2+)-dependent fashion. The interaction of Rac1 and Cdc42 with CaM was shown to be direct. This novel interaction was further confirmed in platelets using co-immunoprecipitation studies. Using CaM database analysis and in vitro peptide competition assays we have identified a 14 amino acid region in Rac1 that is essential for CaM binding. The scrambled form of the peptide did not bind CaM demonstrating specificity of the predicted CaM binding region in Rac1. A similar region capable of binding CaM exists in Cdc42. Furthermore, using the optimal activation time-point for each GTPase, the role of CaM in the function of Rac1 and Cdc42 was examined. Results demonstrate that in human platelets, thrombin caused maximal activation of Rac1 and Cdc42 at ~60 s and ~25 s respectively. The potent CaM antagonist W7 abolished thrombin-mediated activation of Rac1. However, addition of W7 resulted in the activation of Cdc42 over basal and W7 did not inhibit thrombin-mediated activation of Cdc42. The less potent CaM inhibitor, W5, did not have any effect on Rac1 and Cdc42 activation. The results demonstrate that in platelets, binding of CaM to Rac1 increases its activation while its binding to Cdc42 reduces the activation of this GTPase. This suggests an important role for CaM in coordinating Rac1 and Cdc42 activation and in the regulation of cytoskeleton remodeling.


Assuntos
Plaquetas/enzimologia , Calmodulina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Calmodulina/fisiologia , Bovinos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Peptídeos/metabolismo , Sulfonamidas/farmacologia , Proteínas rac1 de Ligação ao GTP/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...