Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-39110164

RESUMEN

Persistent pulmonary hypertension of the newborn (PPHN) is a hypoxic disorder of pulmonary vascular relaxation, mediated in part by adenylyl cyclase (AC). Neonatal pulmonary arteries (PA) express mainly AC6 isoform, followed by AC3, 7 and 9. AC6 expression is upregulated in hypoxia. We reported AC enzyme inhibition due to S-nitrosylation in PPHN PA, and in PA myocytes exposed to hypoxia. We hypothesize that hypoxia promotes cysteine thiol nitrosylation of AC6, impairing cAMP production. HEK293T cells stably expressing AC isoforms (AC3, 5, 6, 7, 9), or cysteine-to-alanine mutants AC6_C1004A, AC6_C1145A or AC6_C447A were cultured in normoxia (21% O2) or hypoxia (10% O2) for 72 hours, or challenged with nitroso donor S-nitrosocysteine (CysNO). AC activity was determined by real-time live-cell cAMP measurement (cADDis assay) or terbium-norfloxacin AC catalytic assay, with or without challenge by allosteric agonist forskolin; protein S-nitrosylation detected by biotin switch method and quantified by affinity precipitation. Only AC6 catalytic activity is inhibited in hypoxia or by S-nitrosylating agent, in presence or absence of forskolin; impaired cAMP production in hypoxia correlates with increased cysteine nitrosylation of AC6. Selective AC6 inhibition in pulmonary artery myocytes extinguishes AC sensitivity to inhibition by hypoxia. Alanine substitution of C1004, but not of other cysteines, decreases S-nitrosylation of AC6. AC activity is diminished in AC6_C1004A compared to AC6 wild type. Substitution of C1004 also extinguishes the inhibition of AC6 by hypoxia. We conclude AC6 is uniquely S-nitrosylated in hypoxia, inhibiting its activity and cAMP generation. We speculate that S-nitrosylation at C1004 may inhibit AC6 interaction with Gαs, playing a role in PPHN pathophysiology.

2.
BMC Oral Health ; 24(1): 1036, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39227897

RESUMEN

BACKGROUND: Silver diamine fluoride (SDF) is an antimicrobial agent and alternative treatment option that can be used to arrest dental decay. While there is optimism with SDF with regard to caries management, there is no true consensus on the number and frequency of applications for children. The purpose of this study was to examine the effectiveness of 38% SDF to arrest early childhood caries (ECC) at three different application regimen intervals. METHODS: Children with teeth that met International Caries Detection and Assessment System codes 5 or 6 criteria were recruited from community dental clinics into an open-label, parallel-group, randomized clinical trial from October 2019 to June 2021. Participants were randomized to one of three groups using sealed envelopes that were prepared with one of three regimens inside: visits one month, four months, or six months apart. Participants received applications of 38% SDF, along with 5% sodium fluoride varnish (NaFV), at the first two visits to treat cavitated carious lesions. Lesions were followed and arrest rates were calculated. Lesions were considered arrested if they were hard on probing and black in colour. Statistics included descriptive and bivariate analyses (Kruskal one-way analysis of variance and Pearson's Chi-squared test). A p-value of ≤ 0.05 was considered significant. RESULTS: Eighty-four children participated in the study (49 males and 35 females, mean age: 44.4 ± 14.2 months). Treatment groups were well matched with 28 participants per group. A total of 374 teeth and 505 lesions were followed. Posterior lesions represented only 40.6% of affected surfaces. Almost all SDF treated lesions were arrested for the one-month (192/196, 98%) and four-month (159/166, 95.8%) interval groups at the final visit. The six-month group experienced the lowest arrest rates; only 72% (103/143) of lesions were arrested (p < 0.001). The duration of application intervals was inversely associated with improvements in arrest rates for all lesions. CONCLUSIONS: Two applications of 38% SDF and 5% NaFV in one-month and four-month intervals were comparable and very effective in arresting ECC. Applications six months apart were less effective and could be considered inferior treatment. TRIAL REGISTRATION: ClinicalTrials.gov NCT04054635 (first registered 13/08/2019).


Asunto(s)
Cariostáticos , Caries Dental , Fluoruros Tópicos , Compuestos de Amonio Cuaternario , Compuestos de Plata , Humanos , Compuestos de Plata/uso terapéutico , Caries Dental/prevención & control , Fluoruros Tópicos/uso terapéutico , Compuestos de Amonio Cuaternario/uso terapéutico , Compuestos de Amonio Cuaternario/administración & dosificación , Femenino , Masculino , Cariostáticos/uso terapéutico , Preescolar , Niño , Fluoruro de Sodio/uso terapéutico
3.
FASEB J ; 35(3): e21375, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33559200

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/fisiología , Encía/inmunología , Interacciones Huésped-Patógeno , Percepción de Quorum/fisiología , Receptores Acoplados a Proteínas G/fisiología , Streptococcus mutans/fisiología , Calcio/metabolismo , Línea Celular , Movimiento Celular , Citocinas/biosíntesis , Células Epiteliales/inmunología , Encía/citología , Humanos , Inmunidad Innata , FN-kappa B/fisiología , Fosfolipasa C beta/fisiología
4.
Handb Exp Pharmacol ; 275: 177-202, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33580389

RESUMEN

Bitter taste receptors (T2Rs) belong to the G protein-coupled receptor superfamily. Humans express 25 T2Rs that are known to detect several bitter compounds including bacterial quorum sensing molecules (QSM). Primarily found to be key receptors for bitter sensation T2Rs are known to play an important role in mediating innate immune responses in oral and extraoral tissues. Several studies have led to identification of Gram-negative and Gram-positive bacterial QSMs as agonists for T2Rs in airway epithelial cells and immune cells. However, the pharmacological characterization for many of the QSM-T2R interactions remains poorly defined. In this chapter, we discuss the extraoral roles including localization of T2Rs in extracellular vesicles, molecular pharmacology of QSM-T2R interactions, role of T2Rs in mediating innate immune responses, and some of the challenges in understanding T2R pharmacology.


Asunto(s)
Interacciones Microbiota-Huesped , Receptores Acoplados a Proteínas G , Células Epiteliales , Humanos , Inmunidad Innata , Sistema Respiratorio , Gusto
5.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36613519

RESUMEN

Polymorphisms in taste receptor genes have been shown to play a role in early childhood caries (ECC), a multifactorial, biofilm-mediated disease. This study aimed to evaluate associations between severe-ECC (S-ECC), the oral microbiome, and variants in genes that encode components of the G protein-coupled receptor (GPCR) signaling cascade involved in taste sensation. A total of 176 children (88 caries-free; 88 with S-ECC) were recruited. Analyses of 16S and ITS1 rRNA microbial genes and seven (GNAQ, GNAS, GNAT3, GNAI2, RAC1, RALB, and PLCB2) human genes were pursued using next-generation sequencing. Regression analyses were performed to evaluate associations between genetic variants, S-ECC, and the supragingival plaque microbiome. Results suggest that PLCB2 rs2305645 (T), rs1869901 (G), and rs2305649 (G) alleles had a protective effect on S-ECC (rs2305645, odds ratio (OR) = 0.27 (95% confidence interval (CI): 0.14-0.51); rs1869901, OR = 0.34 (95% CI: 0.20-0.58); and rs2305649, OR = 0.43 (95% CI: 0.26-0.71)). Variants in GNAQ, GNAS, GNAT3, PLCB2, RALB, and RAC1 were associated with oral fungal and bacterial community composition. This study revealed that three loci at PLCB2 are significantly associated with S-ECC. Variants in multiple genes were associated with the composition of dental biofilm. These findings contribute to the current knowledge about the role of genetics in S-ECC.


Asunto(s)
Caries Dental , Microbiota , Niño , Humanos , Preescolar , Gusto , Microbiota/genética , Bacterias/genética , ARN Ribosómico , Biopelículas , Caries Dental/genética
6.
Curr Issues Mol Biol ; 43(3): 1460-1472, 2021 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-34698096

RESUMEN

The association of taste genetics and the oral microbiome in autoimmune diseases such as rheumatoid arthritis (RA) has not been reported. We explored a novel oral mucosal innate immune pathway involving the bitter taste G protein-coupled receptor T2R38. This case-control study aimed to evaluate whether T2R38 polymorphisms associate with the buccal microbial composition in RA. Genomic DNA was obtained from buccal swabs of 35 RA patients and 64 non-RA controls. TAS2R38 genotypes were determined by Sanger sequencing. The buccal microbiome was assessed by Illumina MiSeq sequencing of the V4-16S rRNA gene. Bacterial community differences were analyzed with alpha and beta diversity measures. Linear discriminant analysis effect size identified taxa discriminating between RA versus non-RA and across TAS2R38 genotypes. TAS2R38 genotype frequency was similar between RA and non-RA controls (PAV/PAV; PAV/AVI; AVI/AVI: RA 42.9%; 45.7%; 11.4% versus controls 32.8%; 48.4%; 18.8%, chi-square (2, N = 99) = 2.1, p = 0.35). The relative abundance of Porphyromonas, among others, differed between RA and non-RA controls. The relative abundance of several bacterial species also differed across TAS2R38 genotypes. These findings suggest an association between T2R38 polymorphisms and RA buccal microbial composition. However, further research is needed to understand the impact of T2R38 in oral health and RA development.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Susceptibilidad a Enfermedades , Microbiota , Mucosa Bucal/microbiología , Polimorfismo de Nucleótido Simple , Receptores Acoplados a Proteínas G/genética , Adulto , Anciano , Alelos , Artritis Reumatoide/patología , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Microbiota/inmunología , Persona de Mediana Edad , ARN Ribosómico 16S , Receptores Acoplados a Proteínas G/metabolismo
7.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34576085

RESUMEN

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.


Asunto(s)
Endocitosis , Células Epiteliales/metabolismo , Encía/citología , Bacterias Grampositivas/metabolismo , Viabilidad Microbiana , Receptores Acoplados a Proteínas G/metabolismo , Gusto , Actinas/metabolismo , Línea Celular , Células Epiteliales/ultraestructura , Humanos , Interleucina-8/metabolismo , Modelos Biológicos , Nitratos/metabolismo , Nitritos/metabolismo , Staphylococcus aureus/metabolismo , Streptococcus mutans/metabolismo , beta-Defensinas/metabolismo , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo
8.
FASEB J ; 33(1): 501-517, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30011231

RESUMEN

Many medications including antibiotics taste bitter. The potency of these antibiotics on the 25 bitter taste receptors (T2Rs) in humans remains poorly understood. Here we characterize by sensory and structure-function analyses how antibiotics frequently used to treat airway infections in cystic fibrosis activate multiple human T2Rs. The potency of the broad-spectrum antibiotics, tobramycin, levofloxacin, and azithromycin on the highly expressed T2Rs in airways, T2R4, T2R14, and T2R20 was pursued. The amino acids and structural features of T2R4, T2R14, and T2R20 important for antibiotic binding were characterized by mutational analysis in heterologous cell-based assays. Strikingly, extracellular loop 2 in T2Rs performs a key function in binding to antibiotics with contribution from residues in transmembrane helices. Our results suggest that different antibiotics activate multiple T2Rs with different potencies. An understanding of the nonantibiotic and physiologic effects mediated through T2Rs on the host cells is much needed.-Jaggupilli, A., Singh, N., De Jesus, V. C., Gounni, M. S., Dhanaraj, P., Chelikani, P. Chemosensory bitter taste receptors (T2Rs) are activated by multiple antibiotics.


Asunto(s)
Antibacterianos/farmacología , Calcio/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Gusto/fisiología , Secuencia de Aminoácidos , Células HEK293 , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Estructura Secundaria de Proteína , Receptores Acoplados a Proteínas G/genética , Gusto/efectos de los fármacos
9.
Mol Cell Biochem ; 463(1-2): 57-66, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31541354

RESUMEN

Membrane lipids regulate the structure and function of G protein-coupled receptors (GPCRs). Previously we have shown that membrane cholesterol regulates the signaling of two human bitter taste receptors (T2Rs), T2R4 and T2R14. Another major plasma membrane lipid known to influence the function of membrane proteins including GPCRs is sphingomyelin. The role of sphingomyelin in T2R function is unexplored thus far. In this work, we examined the significance of sphingomyelin in T2R14 signaling. Results suggest that unavailability of membrane sphingomyelin did not affect the agonist-promoted T2R14 Ca2+ signaling in heterologous expression system and also in primary airway smooth muscle cells (HASM cells). In addition, T2R14 mediated downstream AMPK activation was also unaffected in sphingomyelin-depleted condition; however, cholesterol depletion impaired the T2R14-mediated AMPK activation. Angiotensin II type1A receptor (AT1R) expressed in HASM cells and signals through Ca2+ and AMPK was used as a control. Results suggest that similar to T2R14, membrane sphingomyelin depletion did not affect AT1R signaling. However, membrane cholesterol depletion impaired AT1R mediated Ca2+ signaling and AMPK activation. Interestingly, amino acid sequence analysis revealed the presence of putative sphingolipid binding motif in both T2R14 and AT1R suggesting that the presence of a motif alone might not be suggestive of sphingomyelin sensitivity. In conclusion, these results demonstrate that in contrast to membrane cholesterol, sphingomyelin does not affect the agonist-induced T2R14 signaling, however it may play a role in other aspects of T2R14 function.


Asunto(s)
Señalización del Calcio , Membrana Celular/metabolismo , Colesterol/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Esfingomielinas/metabolismo , Línea Celular , Membrana Celular/genética , Colesterol/genética , Humanos , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Acoplados a Proteínas G/genética , Esfingomielinas/genética
10.
Mol Cell Biochem ; 465(1-2): 199-214, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31894529

RESUMEN

The emerging significance of the bitter taste receptors (T2Rs) role in the extraoral tissues alludes to their potential role in many pathophysiological conditions. The dysregulation of T2R expression and function in disease conditions has now been demonstrated in airways diseases, neurological disorders, and in some cancers. However, the role of T2Rs in the pathophysiology of breast cancer is unexplored thus far. Previously, we demonstrated differential expression of the 25 T2Rs in breast cancer (BC) cells. Based on our previous findings we selected two T2Rs, T2R4 and T2R14 for this work. The objective of the current study is to investigate the expression of T2R4 and T2R14 in BC clinical samples and to examine their physiological role using highly metastatic BC and non-cancerous cell lines. Using approaches, which involve receptor knockdown, pharmacological activation and biochemical assays we report that (i) T2R4 and T2R14 expression patterns are dissimilar, with decreased levels of T2R4 and increased levels of T2R14 in BC clinical samples compared to non-cancerous controls. (ii) Activation of T2Rs with their respective agonist elicited physiological responses in metastatic breast cancer cells, and no responses were seen in non-tumorigenic breast epithelial cells. (iii) Agonist activation of T2Rs (irrespective of T2R subtype) induced anti-proliferative, pro-apoptotic, and anti-migratory responses in highly metastatic breast cancer cells. Taken together, our findings demonstrate that the chemosensory T2R signaling network is involved in evoking physiological responses in the metastatic breast cancer cell line.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Receptores Acoplados a Proteínas G/genética
11.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L45-L57, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30358435

RESUMEN

Bitter taste receptors (T2Rs) are a group of 25 chemosensory receptors expressed at significant levels in the human airways. In human airways, bitter taste receptor 14 (T2R14)-mediated physiological response in ameliorating obstructive airway disorders is an active area of investigation. Therefore, understanding various factors regulating the structure and function of T2R14 will be beneficial. We hypothesize that membrane lipids like cholesterol play a regulatory role in T2R14 signaling in airway cells. We confirmed the expression and signaling of T2R14 in primary human airway smooth muscle (HASM) cells and the human airway epithelial cell line (NuLi-1) using immunoblot analysis and intracellular calcium concentration mobilization experiments, respectively. Next, T2R14 signaling was examined in membrane cholesterol-altered environments by methyl-ß-cyclodextrin or cholesterol oxidase treatments. In the cells analyzed, cholesterol depletion affected the agonist-induced T2R14 signaling, and cholesterol replenishment rescued its efficacy. An alternative approach for cholesterol depletion (with cholesterol oxidase pretreatment) also negatively affected the agonist potency at T2R14 in HASM cells. To understand the molecular mechanism of interaction between cholesterol and T2R14, we used site-directed mutagenesis coupled with functional assays and examined the role of putative cholesterol-binding motifs (CRAC and CARC) in T2R14. Functional characterization of wild-type and mutant T2R14 receptors suggests that amino acid residues K110, F236, and L239 are crucial in T2R14-cholesterol functional interaction. In conclusion, our results show that cholesterol influences the T2R14 signaling efficacy by forming direct interactions with the receptor and consequently plays a regulatory role in T2R14-mediated signaling in human airway cells.


Asunto(s)
Colesterol/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratorio/metabolismo , Transducción de Señal , Secuencias de Aminoácidos , Colesterol/genética , Humanos , Mutagénesis Sitio-Dirigida , Miocitos del Músculo Liso/citología , Receptores Acoplados a Proteínas G/genética , Sistema Respiratorio/citología
12.
Biochim Biophys Acta Biomembr ; 1860(5): 991-999, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29355483

RESUMEN

Bitter taste receptors (T2Rs) are a group of 25 G protein-coupled receptors (GPCRs) in humans. The cognate agonists and the mechanism of ligand binding to the majority of the T2Rs remain unknown. Here we report the first structure-function analysis of T2R7 and study the ability of this receptor to bind to different agonists by site-directed mutagenesis. Screening of ligands for T2R7 in calcium based assays lead to the identification of novel compounds that activate this receptor. Quinine, diphenidol, dextromethorphan and diphenhydramine showed substantial activation of T2R7. Interestingly, these bitter compounds showed different pharmacological characteristics. To investigate the structural features in T2R7 that might contribute to the observed differences in agonist specificities, molecular model guided ligand docking and site-directed mutagenesis was pursued. Amino acids D65, D86, W89, N167, T169, W170, S181, T255 and E271 in the ligand-binding pocket were replaced and the mutants characterized pharmacologically. Our results suggest D86, S181 and W170 present on the extracellular side of transmembrane 3 (TM3), TM5 and in extracellular loop 2 (ECL2) are essential for agonist binding in T2R7. Mutations of these amino acids lead to loss-of-function. We also identified gain-of-function residues that are agonist specific. These results suggest that agonists bind at an extracellular site rather than deep within the TM core involving residues present in both ECL2 and TM helices in T2R7. Similar to majority of the Class A GPCRs, ECL2 in T2R7 plays a significant role in agonist binding and activation.


Asunto(s)
Dominios y Motivos de Interacción de Proteínas , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Sustitución de Aminoácidos , Sitios de Unión/genética , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Estructura Secundaria de Proteína/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Relación Estructura-Actividad , Gusto
13.
Mol Cell Biochem ; 446(1-2): 63-72, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29327289

RESUMEN

Adenylyl cyclases (ACs) are membrane bound enzymes that catalyze the production of cAMP from ATP in response to the activation by G-protein Gαs. Different isoforms of ACs are ubiquitously expressed in different tissues involved in regulatory mechanisms in response to specific stimulants. There are 9 AC isoforms present in humans, with AC5 and AC6 proposed to play a vital role in cardiac functions. The activity of AC6 is sensitive to nitric oxide, such that nitrosylation of the protein might regulate its function. However, the information on structural determinants of nitrosylation in ACs and how they interact with Gαs is limited. Here we used homology modeling to build a molecular model of human AC6 bound to Gαs. Based on this 3D model, we predict the nitrosylation amenable cysteines, and identify potential novel ligands of AC6 using virtual ligand screening. Our model suggests Cys1004 in AC6 (subunit C2) and Cys174 in Gαs present at the AC-Gαs interface as the possible residues that might undergo reversible nitrosylation. Docking analysis predicted novel ligands of AC6 that include forskolin-based compounds and its derivatives. Further work involving site-directed mutagenesis of the predicted residues will allow manipulation of AC activity using novel ligands, and crucial insights on the role of nitrosylation of these proteins in pathophysiological conditions.


Asunto(s)
Adenilil Ciclasas/química , Cromograninas/química , Colforsina , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Simulación del Acoplamiento Molecular , Adenilil Ciclasas/metabolismo , Cromograninas/metabolismo , Colforsina/análogos & derivados , Colforsina/química , Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Ligandos , Estructura Cuaternaria de Proteína
14.
Biochim Biophys Acta ; 1858(9): 2081-2087, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27288892

RESUMEN

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.


Asunto(s)
Colesterol/metabolismo , Quinina/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Células CHO , Colesterol/genética , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Humanos , Mutación Missense , Receptores Acoplados a Proteínas G/genética
15.
Mol Cell Biochem ; 426(1-2): 129-136, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27873143

RESUMEN

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.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Quinina/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Células HEK293 , Humanos , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Proteína de Unión al GTP rac1/genética
16.
Mol Cell Biochem ; 426(1-2): 137-147, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28012014

RESUMEN

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.


Asunto(s)
Bronquios/metabolismo , Regulación de la Expresión Génica/fisiología , Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Mucosa Respiratoria/metabolismo , Señalización del Calcio/fisiología , Humanos , Especificidad de Órganos/fisiología
17.
Biochim Biophys Acta ; 1848(7): 1502-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25858111

RESUMEN

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.


Asunto(s)
Secuencias de Aminoácidos/genética , Mutación , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Secuencia de Aminoácidos , Citometría de Flujo , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
18.
Biochemistry ; 54(16): 2622-31, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25844797

RESUMEN

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.


Asunto(s)
Ácido Abscísico/química , Modelos Moleculares , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/química , Sitios de Unión , Carotenoides/química , Humanos , Mutagénesis Sitio-Dirigida , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sesquiterpenos/química , Relación Estructura-Actividad , Percepción del Gusto/fisiología , Yohimbina/química
19.
J Biol Chem ; 289(36): 25054-66, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25059668

RESUMEN

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.


Asunto(s)
Aminoácidos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Gusto , Alanina/química , Alanina/genética , Alanina/metabolismo , Aminoácidos/química , Aminoácidos/genética , Asparagina/química , Asparagina/genética , Asparagina/metabolismo , Sitios de Unión/genética , GABAérgicos/química , GABAérgicos/farmacología , Células HEK293 , Humanos , Lisina/química , Lisina/genética , Lisina/farmacología , Modelos Moleculares , Estructura Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Quinina/química , Quinina/farmacología , Receptores Acoplados a Proteínas G/química , Treonina/química , Treonina/genética , Treonina/metabolismo , Ácido gamma-Aminobutírico/química , Ácido gamma-Aminobutírico/farmacología
20.
Biochim Biophys Acta ; 1838(1 Pt B): 231-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23994601

RESUMEN

Bitter taste receptors (T2Rs) belong to the superfamily of G protein-coupled receptors (GPCRs). T2Rs are chemosensory receptors with important therapeutic potential. In humans, bitter taste is perceived by 25 T2Rs, which are distinct from the well-studied Class A GPCRs. The activation mechanism of T2Rs is poorly understood and none of the structure-function studies are focused on the role of the important third intracellular loop (ICL3). T2Rs have a unique signature sequence at the cytoplasmic end of fifth transmembrane helix (TM5), a highly conserved LxxSL motif. Here, we pursue an alanine scan mutagenesis of the ICL3 of T2R4 and characterize the functionality of 23 alanine mutants. We identify four mutants, H214A, Q216A, V234A and M237A, that exhibit constitutive activity. To our surprise, the H214A mutant showed very high constitutive activity over wild type T2R4. Interestingly, His214 is highly conserved (96%) in T2Rs and is present two amino acids below the LxxSL motif in TM5. Molecular modeling shows a dynamic network of interactions involving residues in TM5-ICL3-TM6 that restrain the movement of the helices. Changes in this network, as in the case of H214A, Q216A, V234A and M237A mutants, cause the receptor to adopt an active conformation. The conserved LxxSL motif in TM5 performs both structural and functional roles in this process. These results provide insight into the activation mechanism of T2Rs, and emphasize the unique functional role of ICL3 even within the GPCR subfamilies.


Asunto(s)
Alanina/química , Receptores Acoplados a Proteínas G/química , Proteínas Recombinantes de Fusión/química , Papilas Gustativas/química , Alanina/genética , Alanina/metabolismo , Secuencias de Aminoácidos , Calcio/metabolismo , Secuencia Conservada , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Células HEK293 , Histidina/química , Histidina/genética , Histidina/metabolismo , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Quinina/química , Quinina/farmacología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Papilas Gustativas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA