Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
J Biol Chem ; 299(11): 105293, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37774973

RESUMO

ß-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether ß-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., ß-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate ß-arrestins, thereby limiting the ability to distinguish G protein from ß-arrestin-mediated signaling events. We used ß2-adrenergic receptor (ß2AR) and its ß2AR-C tail mutant expressed in human embryonic kidney 293 cells wildtype or CRISPR-Cas9 gene edited for Gαs, ß-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and ß-arrestins in controlling gene expression. We found that Gαs is not required for ß2AR and ß-arrestin conformational changes, ß-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in ß-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of ß2AR in wildtype and ß-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing ß-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of ß2AR. Overall, our results support that Gs is essential for ß2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas ß-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.


Assuntos
Proteínas de Ligação ao GTP , Regulação da Expressão Gênica , Receptores Adrenérgicos beta 2 , beta-Arrestinas , Humanos , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo , beta-Arrestinas/genética , beta-Arrestinas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Ligação ao GTP/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Células HEK293 , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Estrutura Terciária de Proteína , Isoformas de Proteínas , Ativação Enzimática/genética
2.
Sci Signal ; 16(797): eadf2173, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37552769

RESUMO

G protein-coupled receptors engage both G proteins and ß-arrestins, and their coupling can be biased by ligands and mutations. Here, to resolve structural elements and mechanisms underlying effector coupling to the angiotensin II (AngII) type 1 receptor (AT1R), we combined alanine scanning mutagenesis of the entire sequence of the receptor with pharmacological profiling of Gαq and ß-arrestin engagement to mutant receptors and molecular dynamics simulations. We showed that Gαq coupling to AT1R involved a large number of residues spread across the receptor, whereas fewer structural regions of the receptor contributed to ß-arrestin coupling regulation. Residue stretches in transmembrane domain 4 conferred ß-arrestin bias and represented an important structural element in AT1R for functional selectivity. Furthermore, we identified allosteric small-molecule binding sites that were enclosed by communities of residues that produced biased signaling when mutated. Last, we showed that allosteric communication within AT1R emanating from the Gαq coupling site spread beyond the orthosteric AngII-binding site and across different regions of the receptor, including currently unresolved structural regions. Our findings reveal structural elements and mechanisms within AT1R that bias Gαq and ß-arrestin coupling and that could be harnessed to design biased receptors for research purposes and to develop allosteric modulators.


Assuntos
Receptor Tipo 1 de Angiotensina , Transdução de Sinais , beta-Arrestinas/genética , beta-Arrestinas/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , beta-Arrestina 1/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Angiotensina II/metabolismo
3.
Basic Clin Pharmacol Toxicol ; 133(4): 331-341, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37056198

RESUMO

GPR56/ADGRG1 is an adhesion G protein-coupled receptor (GPCR) and mutations on this receptor cause cortical malformation due to the over-migration of neural progenitor cells on brain surface. At pial surface, GPR56 interacts with collagen III, induces Rho-dependent activation through Gα12/13 and inhibits the neuronal migration. In human glioma cells, GPR56 inhibits cell migration through Gαq/11 -dependent Rho pathway. GPR56-tetraspanin complex is known to couple Gαq/11 . GPR56 is an aGPCR that couples with various G proteins and signals through different downstream pathways. In this study, bilateral frontoparietal polymicrogyria (BFPP) mutants disrupting GPR56 function but remaining to be expressed on plasma membrane were used to study receptor signalling through Gα12 , Gα13 and Gα11 with BRET biosensors. GPR56 showed coupling with all three G proteins and activated heterotrimeric G protein signalling upon stimulation with Stachel peptide. However, BFPP mutants showed different signalling defects for each G protein indicative of distinct activation and signalling properties of GPR56 for Gα12 , Gα13 or Gα11 . ß-arrestin recruitment was also investigated following the activation of GPR56 with Stachel peptide using BRET biosensors. N-terminally truncated GPR56 showed enhanced ß-arrestin recruitment; however, neither wild-type receptor nor BFPP mutants gave any measurable recruitment upon Stachel stimulation, pointing different activation mechanisms for ß-arrestin involvement.


Assuntos
Proteínas de Ligação ao GTP , Receptores Acoplados a Proteínas G , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Mutação , Proteínas de Ligação ao GTP/metabolismo , Peptídeos , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
4.
JCI Insight ; 8(5)2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36883568

RESUMO

WHIM syndrome is an inherited immune disorder caused by an autosomal dominant heterozygous mutation in CXCR4. The disease is characterized by neutropenia/leukopenia (secondary to retention of mature neutrophils in bone marrow), recurrent bacterial infections, treatment-refractory warts, and hypogammaglobulinemia. All mutations reported in WHIM patients lead to the truncations in the C-terminal domain of CXCR4, R334X being the most frequent. This defect prevents receptor internalization and enhances both calcium mobilization and ERK phosphorylation, resulting in increased chemotaxis in response to the unique ligand CXCL12. Here, we describe 3 patients presenting neutropenia and myelokathexis, but normal lymphocyte count and immunoglobulin levels, carrying what we believe to be a novel Leu317fsX3 mutation in CXCR4, leading to a complete truncation of its intracellular tail. The analysis of the L317fsX3 mutation in cells derived from patients and in vitro cellular models reveals unique signaling features in comparison with R334X mutation. The L317fsX3 mutation impairs CXCR4 downregulation and ß-arrestin recruitment in response to CXCL12 and reduces other signaling events - including ERK1/2 phosphorylation, calcium mobilization, and chemotaxis - all processes that are typically enhanced in cells carrying the R334X mutation. Our findings suggest that, overall, the L317fsX3 mutation may be causative of a form of WHIM syndrome not associated with an augmented CXCR4 response to CXCL12.


Assuntos
Proteínas de Ligação ao GTP , Doenças da Imunodeficiência Primária , beta-Arrestinas , Humanos , beta-Arrestina 1/genética , beta-Arrestina 1/imunologia , beta-Arrestinas/genética , beta-Arrestinas/imunologia , Cálcio/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/imunologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Mutação , Neutropenia/genética , Neutropenia/imunologia , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/imunologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Verrugas/genética , Verrugas/imunologia
5.
J Biol Chem ; 298(9): 102351, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35940305

RESUMO

G protein-coupled receptor (GPCR) kinases (GRKs) and arrestins mediate GPCR desensitization, internalization, and signaling. The spatial pattern of GPCR phosphorylation is predicted to trigger these discrete GRK and arrestin-mediated functions. Here, we provide evidence that distal carboxyl-terminal tail (C-tail), but not proximal, phosphorylation of the chemokine receptor CXCR4 specifies ßarrestin1 (ßarr1)-dependent signaling. We demonstrate by pharmacologic inhibition of GRK2/3-mediated phosphorylation of the chemokine receptor CXCR4 coupled with site-directed mutagenesis and bioluminescence resonance energy transfer approaches that distal, not proximal, C-tail phosphorylation sites are required for recruitment of the adaptor protein STAM1 (signal-transducing adaptor molecule) to ßarr1 and focal adhesion kinase phosphorylation but not extracellular signal-regulated kinase 1/2 phosphorylation. In addition, we show that GPCRs that have similarly positioned C-tail phosphoresidues are also able to recruit STAM1 to ßarr1. However, although necessary for some GPCRs, we found that distal C-tail sites might not be sufficient to specify recruitment of STAM1 to ßarr1 for other GPCRs. In conclusion, this study provides evidence that distal C-tail phosphorylation sites specify GRK-ßarrestin-mediated signaling by CXCR4 and other GPCRs.


Assuntos
Arrestina , Quinases de Receptores Acoplados a Proteína G , Arrestina/metabolismo , Arrestinas/metabolismo , Quinases de Receptores Acoplados a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/metabolismo , Fosforilação/fisiologia , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
7.
J Biol Chem ; 298(6): 102057, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35605660

RESUMO

The melanocortin receptor accessory protein 2 (MRAP2) is essential for several physiological functions of the ghrelin receptor growth hormone secretagogue receptor 1a (GHSR1a), including increasing appetite and suppressing insulin secretion. In the absence of MRAP2, GHSR1a displays high constitutive activity and a weak G-protein-mediated response to ghrelin and readily recruits ß-arrestin. In the presence of MRAP2, however, G-protein-mediated signaling via GHSR1a is strongly dependent on ghrelin stimulation and the recruitment of ß-arrestin is significantly diminished. To better understand how MRAP2 modifies GHSR1a signaling, here we investigated the role of several phosphorylation sites within the C-terminal tail and third intracellular loop of GHSR1a, as well as the mechanism behind MRAP2-mediated inhibition of ß-arrestin recruitment. We show that Ser252 and Thr261 in the third intracellular loop of GHSR1a contribute to ß-arrestin recruitment, whereas the C-terminal region is not essential for ß-arrestin interaction. Additionally, we found that MRAP2 inhibits GHSR1a phosphorylation by blocking the interaction of GRK2 and PKC with the receptor. Taken together, these data suggest that MRAP2 alters GHSR1a signaling by directly impacting the phosphorylation state of the receptor and that the C-terminal tail of GHSR1a prevents rather than contribute to ß-arrestin recruitment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Grelina , Receptores de Grelina , beta-Arrestinas , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Grelina/metabolismo , Melanocortinas , Fosforilação , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
8.
Mol Pharmacol ; 101(2): 87-94, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34853152

RESUMO

G protein-coupled receptors (GPCRs) transduce a diverse variety of extracellular stimuli into intracellular signaling. These receptors are the most clinically productive drug targets at present. Despite decades of research on the signaling consequences of molecule-receptor interactions, conformational components of receptor-effector interactions remain incompletely described. The ß 2-adrenergic receptor (ß 2AR) is a prototypical and extensively studied GPCR that can provide insight into this aspect of GPCR signaling thanks to robust structural data and rich pharmacopeia. Using bioluminescence resonance energy transfer -based biosensors, second messenger assays, and biochemical techniques, we characterize the properties of ß 2AR-F193A. This single point mutation in extracellular loop 2 of the ß 2AR is sufficient to intrinsically bias the ß 2AR away from ß-arrestin interaction and demonstrates altered regulatory outcomes downstream of this functional selectivity. This study highlights the importance of extracellular control of intracellular response to stimuli and suggests a previously undescribed role for the extracellular loops of the receptor and the extracellular pocket formed by transmembrane domains 2, 3, and 7 in GPCR regulation that may contribute to biased signaling at GPCRs. SIGNIFICANCE STATEMENT: The role of extracellular G protein-coupled receptor (GPCR) domains in mediating intracellular interactions is poorly understood. We characterized the effects of extracellular loop mutations on agonist-promoted interactions of GPCRs with G protein and ß-arrestin. Our studies reveal that F193 in extracellular loop 2 in the ß2-adrenergic receptor mediates interactions with G protein and ß-arrestin with a biased loss of ß-arrestin binding. These results provide new insights on the role of the extracellular domain in differentially modulating intracellular interactions with GPCRs.


Assuntos
Líquido Extracelular/metabolismo , Fenilalanina/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , beta-Arrestinas/metabolismo , Sequência de Aminoácidos , Relação Dose-Resposta a Droga , Técnicas de Inativação de Genes/métodos , Células HEK293 , Humanos , Fenilalanina/química , Fenilalanina/genética , Estrutura Secundária de Proteína , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , beta-Arrestinas/química , beta-Arrestinas/genética
9.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34916293

RESUMO

G protein-coupled receptors (GPCRs) are the largest family of human proteins. They have a common structure and, signaling through a much smaller set of G proteins, arrestins, and effectors, activate downstream pathways that often modulate hallmark mechanisms of cancer. Because there are many more GPCRs than effectors, mutations in different receptors could perturb signaling similarly so as to favor a tumor. We hypothesized that somatic mutations in tumor samples may not be enriched within a single gene but rather that cognate mutations with similar effects on GPCR function are distributed across many receptors. To test this possibility, we systematically aggregated somatic cancer mutations across class A GPCRs and found a nonrandom distribution of positions with variant amino acid residues. Individual cancer types were enriched for highly impactful, recurrent mutations at selected cognate positions of known functional motifs. We also discovered that no single receptor drives this pattern, but rather multiple receptors contain amino acid substitutions at a few cognate positions. Phenotypic characterization suggests these mutations induce perturbation of G protein activation and/or ß-arrestin recruitment. These data suggest that recurrent impactful oncogenic mutations perturb different GPCRs to subvert signaling and promote tumor growth or survival. The possibility that multiple different GPCRs could moonlight as drivers or enablers of a given cancer through mutations located at cognate positions across GPCR paralogs opens a window into cancer mechanisms and potential approaches to therapeutics.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Cálcio , Linhagem Celular Tumoral , Simulação por Computador , Ensaio de Imunoadsorção Enzimática , Humanos , Mutação , Neoplasias/genética , Conformação Proteica , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética
10.
Int J Mol Sci ; 22(19)2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34638633

RESUMO

Opioid addiction is a complex phenomenon with genetic, social, and other components. Due to such complexity, it is difficult to interpret the outcome of clinical studies, and thus, mutations found in individuals with these addictions are still not indisputably classified as opioid addiction-causing variants. Here, we computationally investigated two such mutations, A6V and N40D, found in the mu opioid receptor gene OPRM1. The mutations are located in the extracellular domain of the corresponding protein, which is important to the hetero-dimerization of OPRM1 with the delta opioid receptor protein (OPRD1). The hetero-dimerization of OPRD1-OPRM1 affects the signaling pathways activated by opioids and natural peptides and, thus, could be considered a factor contributing to addiction. In this study, we built four 3D structures of molecular pathways, including the G-protein signaling pathway and the ß-arrestin signaling pathway of the heterodimer of OPRD1-OPRM1. We also analyzed the effect of mutations of A6V and N40D on the stability of individual OPRM1/OPRD1 molecules and the OPRD1-OPRM1 heterodimer with the goal of inferring their plausible linkage with opioid addiction. It was found that both mutations slightly destabilize OPRM1/OPRD1 monomers and weaken their association. Since hetero-dimerization is a key step for signaling processes, it is anticipated that both mutations may be causing increased addiction risk.


Assuntos
Transtornos Relacionados ao Uso de Opioides/genética , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Receptores Opioides/genética , Transdução de Sinais/genética , Dimerização , Humanos , Mutação/genética , beta-Arrestinas/genética
11.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34599099

RESUMO

Alternative splicing of G protein-coupled receptors has been observed, but their functions are largely unknown. Here, we report that a splice variant (SV1) of the human growth hormone-releasing hormone receptor (GHRHR) is capable of transducing biased signal. Differing only at the receptor N terminus, GHRHR predominantly activates Gs while SV1 selectively couples to ß-arrestins. Based on the cryogenic electron microscopy structures of SV1 in the apo state or GHRH-bound state in complex with the Gs protein, molecular dynamics simulations reveal that the N termini of GHRHR and SV1 differentiate the downstream signaling pathways, Gs versus ß-arrestins. As suggested by mutagenesis and functional studies, it appears that GHRH-elicited signal bias toward ß-arrestin recruitment is constitutively mediated by SV1. The level of SV1 expression in prostate cancer cells is also positively correlated with ERK1/2 phosphorylation but negatively correlated with cAMP response. Our findings imply that constitutive signal bias may be a mechanism that ensures cancer cell proliferation.


Assuntos
Processamento Alternativo/genética , Variação Genética/genética , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Células Cultivadas , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/genética , Células PC-3 , Células Sf9 , Transdução de Sinais/genética , beta-Arrestinas/genética
12.
Front Endocrinol (Lausanne) ; 12: 714561, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484125

RESUMO

ß-arrestins are partners of the G protein-coupled receptors (GPCRs), regulating their intracellular trafficking and signaling. Development of biased GPCR agonists, selectively targeting either G protein or ß-arrestin pathways, are in the focus of interest due to their therapeutic potential in different pathological conditions. The CB2 cannabinoid receptor (CB2R) is a GPCR involved in various functions in the periphery and the central nervous system. Two common occurring variants of CB2R, harboring Q63R or L133I missense mutations, have been implicated in the development of a diverse set of disorders. To evaluate the effect of these mutations, we characterized the binding profile of these mutant CB2 receptors to G proteins and ß-arrestin2. Although their ability to inhibit cAMP signaling was similar, the Q63R mutant had increased, whereas the L133I mutant receptor had decreased ß-arrestin2 binding. In line with these observations, the variants also had altered intracellular trafficking. Our results show that two common variants of the CB2 receptor have biased signaling properties, which may contribute to the pathogenesis of the associated disorders and may offer CB2R as a target for further development of biased receptor activation strategies.


Assuntos
Mutação de Sentido Incorreto , Receptor CB2 de Canabinoide/metabolismo , beta-Arrestinas/metabolismo , Células HEK293 , Humanos , Ligação Proteica , Transporte Proteico , Receptor CB2 de Canabinoide/química , Receptor CB2 de Canabinoide/genética , beta-Arrestinas/genética
13.
Mol Cell ; 81(20): 4165-4175.e6, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34433090

RESUMO

GPCR functional selectivity opens new opportunities for the design of safer drugs. Ligands orchestrate GPCR signaling cascades by modulating the receptor conformational landscape. Our study provides insights into the dynamic mechanism enabling opioid ligands to preferentially activate the G protein over the ß-arrestin pathways through the µ-opioid receptor (µOR). We combine functional assays in living cells, solution NMR spectroscopy, and enhanced-sampling molecular dynamic simulations to identify the specific µOR conformations induced by G protein-biased agonists. In particular, we describe the dynamic and allosteric communications between the ligand-binding pocket and the receptor intracellular domains, through conserved motifs in class A GPCRs. Most strikingly, the biased agonists trigger µOR conformational changes in the intracellular loop 1 and helix 8 domains, which may impair ß-arrestin binding or signaling. The findings may apply to other GPCR families and provide key molecular information that could facilitate the design of biased ligands.


Assuntos
Analgésicos Opioides/farmacologia , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/química , Animais , Sítios de Ligação , Desenho Assistido por Computador , Desenho de Fármacos , Agonismo Parcial de Drogas , Células HEK293 , Humanos , Ligantes , Camundongos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Células Sf9 , Relação Estrutura-Atividade , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
14.
BMC Biotechnol ; 21(1): 41, 2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225700

RESUMO

BACKGROUND: CCR6 chemokine receptor is an important target in inflammatory diseases. Th17 cells express CCR6 and a number of inflammatory cytokines, including IL-17 and IL-22, which are involved in the propagation of inflammatory immune responses. CCR6 antagonist would be a potential treatment for inflammatory diseases such as psoriasis or rheumatoid arthritis. The aim of this study is to develop an antagonistic monoclonal antibody (mAb) against human CCR6 receptor (hCCR6). RESULTS: We generate monoclonal antibodies against hCCR6 immunizing Balb/c mice with hCCR6 overexpressing cells. The antibodies were tested by flow cytometry for specific binding to hCCR6, cloned by limiting dilution and resulted in the isolation and purification monoclonal antibody 1C6. By ELISA and flow cytometry, was determined that the antibody obtained binds to hCCR6 N-terminal domain. The ability of 1C6 to neutralize hCCR6 signaling was tested and we determined that 1C6 antibody were able to block response in ß-arrestin recruitment assay with IC50 10.23 nM, but did not inhibit calcium mobilization. In addition, we found in a chemotaxis assay that 1C6 reduces the migration of hCCR6 cells to their ligand CCL20. Finally, we determined by RT-qPCR that the expression of IL-17A in Th17 cells treated with 1C6 was inhibited. CONCLUSIONS: In the present study, we applied whole cell immunization for successfully obtain an antibody that is capable to neutralize hCCR6 signaling and to reduce hCCR6 cells migration and IL-17 expression. These results provide an efficient approach to obtain therapeutic potential antibodies in the treatment of CCR6-mediated inflammatory diseases.


Assuntos
Anticorpos Monoclonais/imunologia , Quimiocina CCL20/imunologia , Interleucina-17/imunologia , Receptores CCR6/química , Receptores CCR6/imunologia , beta-Arrestinas/imunologia , Animais , Quimiocina CCL20/genética , Feminino , Humanos , Inflamação/genética , Inflamação/imunologia , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos BALB C , Domínios Proteicos , Receptores CCR6/genética , Transdução de Sinais , beta-Arrestinas/genética
15.
FEBS J ; 288(8): 2502-2512, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33738925

RESUMO

G protein-coupled receptors (GPCRs) are membrane-bound proteins that are ubiquitously expressed in many cell types and take part in mediating multiple signaling pathways. GPCRs are dynamic proteins and exist in an equilibrium between an ensemble of conformational states such as inactive and fully active states. This dynamic nature of GPCRs is one of the factors that confers their basal activity even in the absence of any ligand-mediated activation. Ligands selectively bind and stabilize a subset of the conformations from the ensemble leading to a shift in the equilibrium toward the inactive or the active state depending on the nature of the ligand. This ligand-selective effect is achieved through allosteric communication between the ligand binding site and G protein or ß-arrestin coupling site. Similarly, the G protein coupling to the receptor exerts the allosteric effect on the ligand binding region leading to increased binding affinity for agonists and decreased affinity for antagonists or inverse agonists. In this review, we enumerate the current state of our understanding of the mechanism of allosteric communication in GPCRs with a specific focus on the critical role of computational methods in delineating the residues involved in allosteric communication. Analyzing allosteric communication mechanism using molecular dynamics simulations has revealed (a) a structurally conserved mechanism of allosteric communication that regulates the G protein coupling, (b) a rational structure-based approach to designing selective ligands, and (c) an approach to designing allosteric GPCR mutants that are either ligand and G protein or ß-arrestin selective.


Assuntos
Proteínas de Ligação ao GTP/genética , Receptores Adrenérgicos beta 2/genética , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética , Regulação Alostérica/genética , Sítio Alostérico/genética , Humanos , Ligantes , Ligação Proteica/genética , Conformação Proteica , Transdução de Sinais/genética
16.
FEBS J ; 288(8): 2529-2549, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33690974

RESUMO

Arrestins (arr) are multifunctional cytosolic adaptors that bind to active and phosphorylated G protein-coupled receptors (GPCRs) via a highly versatile interface. Arrestins stop G protein signaling and trigger other signaling pathways. Recently, 3D structures of arr-GPCR complexes have been solved, which provide a bulk of structural information for understanding the mechanism of arr recruitment and activation. However, many questions about the functional consequences of structural details and the dynamics of the arr-GPCR interaction remain open. A wealth of information about key determinants for the arr-GPCR interaction and their functional relevance, and dynamic insights into the process of arr binding and the functional outcomes of different binding modes have been provided by a series of biochemical methods which we review here. Importantly, most of these methods provide information from the live cell, which is a necessary validation and complement for structural data. With the main focus on the most recent research, we will highlight major findings about arr structure, function, and dynamics derived from mutagenesis studies, cross-linking studies, conformational probes, and sensors, and we summarize available systems to detect arr recruitment. Furthermore, we discuss recent findings and directions of in silico investigations in arr-GPCR complexes.


Assuntos
Proteínas de Ligação ao GTP/genética , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética , Humanos , Fosforilação/genética , Transdução de Sinais/genética , Relação Estrutura-Atividade
17.
Med Oncol ; 38(4): 38, 2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-33721131

RESUMO

Non-visual arrestins (ß-arrestins) are endocytic proteins that mediate agonist-activated GPCRs internalization and signaling pathways in an independent manner. The involvement of ß-arrestins in cancer invasion and metastasis is increasingly reported. So, it is hypothesized that inhibition of ß-arrestins may diminish the survival chances of cancer cells. This study aimed to evaluate the in vitro impact of inhibiting ß-arrestins on the autophagic and/or apoptotic responsiveness of breast cancer cells. We used Barbadin to selectively inhibit ß-Arr/AP2 interaction in AVP-stimulated V2R receptor of triple-negative breast cancer cells (MDA MB-231). Autophagy was assessed by the microtubule-associated protein 1 light chain 3-II (LC3II), apoptosis was measured by Annexin-V/PI staining and cell cycle distribution was investigated based upon the DNA content using flow cytometry. Barbadin reduced cell viability to 69.1% and increased the autophagy marker LC3II and its autophagic effect disappeared in cells transiently starved in Earle's balanced salt solution (EBSS). Also, Barbadin mildly enhanced the expression of P62 mRNA and arrested 63.7% of cells in G0/G1 phase. In parallel, the drug-induced apoptosis in 29.9% of cells (by AV/PI) and 27.8% of cells were trapped in sub-G1 phase. The apoptotic effect of Barbadin was enhanced when autophagy was inhibited by the PI3K inhibitor (Wortmannin). Conclusively, the data demonstrate the dual autophagic and apoptotic effects of ß-ßArr/AP2 inhibition in triple-negative breast cancer cells. These observations nominate ß-Arrs as selective targets in breast cancer treatment.


Assuntos
Apoptose , Autofagia , Pontos de Checagem da Fase G1 do Ciclo Celular , Receptores de Vasopressinas/metabolismo , beta-Arrestinas/antagonistas & inibidores , Complexo 2 de Proteínas Adaptadoras/metabolismo , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Pirimidinas/farmacologia , Receptores de Vasopressinas/agonistas , Transdução de Sinais/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
18.
FEBS J ; 288(8): 2513-2528, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33621418

RESUMO

G protein-coupled receptors (GPCRs) are essential cell membrane signaling molecules and represent the most important class of drug targets. Some signaling pathways downstream of a GPCR may be responsible for drug adverse effects, while others mediate therapeutic efficacy. Biased ligands preferentially activate only a subset of all GPCR signaling pathways. They hold great potential to become next-generation GPCR drugs with less side effects due to their potential to exclusively activate desired signaling pathways. However, the molecular basis of biased agonism is poorly understood. GPCR activation occurs through allosteric coupling, the propagation of conformational changes from the extracellular ligand-binding pocket to the intracellular G protein-binding interface. Comparison of GPCR structures in complex with G proteins or ß-arrestin reveals that intracellular transducer coupling results in closure of the ligand-binding pocket trapping the agonist inside its binding site. Allosteric coupling appears to be transducer-specific offering the possibility of harnessing this mechanism for the design of biased ligands. Here, we review the biochemical, pharmacological, structural, and biophysical evidence for allosteric coupling and delineate that biased agonism should be a consequence of preferential allosteric coupling from the ligand-binding pocket to one transducer-binding site. As transducer binding leads to large structural rearrangements in the extracellular ligand-binding pocket, we survey biased ligands with an extended binding mode that interact with extracellular receptor domains. We propose that biased ligands use ligand-specific triggers inside the binding pocket that are relayed through preferential allosteric coupling to a specific transducer, eventually leading to biased signaling.


Assuntos
Proteínas de Ligação ao GTP/genética , Conformação Proteica , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética , Regulação Alostérica/genética , Sítios de Ligação/genética , Humanos , Ligantes , Ligação Proteica/genética , Domínios Proteicos/genética , Transdução de Sinais/genética
19.
FEBS J ; 288(8): 2562-2569, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33605032

RESUMO

G protein-coupled receptors (GPCRs) are a large class of cell-surface receptor involved in cellular signaling that are currently the target of over one third of all clinically approved therapeutics. Classically, an agonist-bound, active GPCR couples to and activates G proteins through the receptor intracellular core. To attenuate G protein signaling, the GPCR is phosphorylated at its C-terminal tail and/or relevant intracellular loops, allowing for the recruitment of ß-arrestins (ßarrs). ßarrs then couple to the receptor intracellular core in order to mediate receptor desensitization and internalization. However, our laboratory and others have observed that some GPCRs are capable of continuously signaling through G protein even after internalization. This mode of sustained signaling stands in contrast with our previous understanding of GPCR signaling, and its molecular mechanism is still not well understood. Recently, we have solved the structure of a GPCR-G protein-ßarr megacomplex by cryo-electron microscopy. This 'megaplex' structure illustrates the independent and simultaneous coupling of a G protein to the receptor intracellular core, and binding of a ßarr to a phosphorylated receptor C-terminal tail, with all three components maintaining their respective canonically active conformations. The structure provides evidence for the ability of a GPCR to activate G protein even while being bound to and internalized by ßarr. It also reveals that the binding of G protein and ßarr to the same GPCR is not mutually exclusive, and raises a number of future questions to be answered regarding the mechanism of sustained signaling.


Assuntos
Endossomos/genética , Proteínas de Ligação ao GTP/genética , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética , Arrestinas/genética , Arrestinas/ultraestrutura , Microscopia Crioeletrônica , Endocitose/genética , Endossomos/ultraestrutura , Proteínas de Ligação ao GTP/ultraestrutura , Humanos , Conformação Molecular , Fosforilação , Ligação Proteica/genética , Receptores Acoplados a Proteínas G/ultraestrutura , Transdução de Sinais/genética
20.
FEBS J ; 288(8): 2550-2561, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33539669

RESUMO

G protein-coupled receptors (GPCRs) canonically couple to specific Gα protein subtypes and ß-arrestin adaptor proteins to initiate cellular signaling events. G protein-mediated signaling and ß-arrestin-mediated signaling have broadly been considered separable. However, noncanonical interactions between G proteins and GPCRs are now appreciated that do not result in nucleotide exchange and classical G protein signaling. New findings also demonstrate direct interactions between G proteins and ß-arrestins that are required for certain signaling and physiological events. Further adding to the intrigue of these newly appreciated G protein:ß-arrestin complexes, only the Gαi subtype family members, and not Gαs, Gαq/11, or Gα12/13 subtypes, appear to form direct interactions with ß-arrestin. Here, we review the recent discovery and initial characterization of G protein:ß-arrestin complexes and describe how these complexes provide mechanistic insight into seemingly disparate observations. G protein:ß-arrestin complexes build upon other observations of noncanonical G protein and ß-arrestin signaling events to add an additional dimension to our understanding of GPCR signaling.


Assuntos
Proteínas de Ligação ao GTP/genética , Conformação Proteica , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética , Células HEK293 , Humanos , Fosforilação/genética , Ligação Proteica/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...