Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 54(1): 166-179, 2014 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-24685158

RESUMO

Molecular chaperones triage misfolded proteins via action as substrate selectors for quality control (QC) machines that fold or degrade clients. Herein, the endoplasmic reticulum (ER)-associated Hsp40 JB12 is reported to participate in partitioning mutant conformers of gonadotropin-releasing hormone receptor (GnRHR), a G protein-coupled receptor, between ER-associated degradation (ERAD) and an ERQC autophagy pathway. ERQC autophagy degrades E90K-GnRHR because pools of its partially folded and detergent-soluble degradation intermediates are resistant to ERAD. S168R-GnRHR is globally misfolded and disposed of via ERAD, but inhibition of p97, the protein retrotranslocation motor, shunts S168R-GnRHR from ERAD to ERQC autophagy. Partially folded and grossly misfolded forms of GnRHR associate with JB12 and Hsp70. Elevation of JB12 promotes ERAD of S168R-GnRHR, with E90K-GnRHR being resistant. E90K-GnRHR elicits association of the Vps34 autophagy initiation complex with JB12. Interaction between ER-associated Hsp40s and the Vps34 complex permits the selective degradation of ERAD-resistant membrane proteins via ERQC autophagy.


Assuntos
Autofagia , Degradação Associada com o Retículo Endoplasmático , Dobramento de Proteína , Receptores LHRH/metabolismo , Animais , Autofagia/efeitos dos fármacos , Células COS , Chlorocebus aethiops , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Cinética , Modelos Moleculares , Mutação , Inibidores de Proteassoma/farmacologia , Conformação Proteica , Dobramento de Proteína/efeitos dos fármacos , Transporte Proteico , Proteólise , Interferência de RNA , Receptores LHRH/química , Receptores LHRH/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Transfecção
2.
Biochem J ; 475(18): 2941-2953, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30068530

RESUMO

A series of compounds formerly identified by high-throughput screening was studied for their ability to serve as pharmacoperones for the vasopressin type 2 receptor (V2R) mutant L83Q, which is known to cause nephrogenic diabetes insipidus (NDI). Three compounds were particularly effective in rerouting the mutant receptor in a concentration-dependent manner, were neither agonists nor antagonists, and displayed low cellular toxicity. Compound 1 was most effective and can be used as a molecular probe for future studies of how small molecules may affect NDI caused by mutant V2R. These compounds, however, failed to rescue the V2R Y128S mutant, indicating that the compounds described may not work in the rescue of all known mutants of V2R. Taken collectively, the present studies have now identified a promising lead compound that could function as a pharmacoperone to correct the trafficking defect of the NDI-associated mutant V2R L83Q and thus has the therapeutic potential for the treatment of NDI.


Assuntos
Chaperonas Moleculares/farmacologia , Sondas Moleculares/farmacologia , Mutação de Sentido Incorreto , Receptores de Vasopressinas/metabolismo , Substituição de Aminoácidos , Diabetes Insípido Nefrogênico/tratamento farmacológico , Diabetes Insípido Nefrogênico/genética , Diabetes Insípido Nefrogênico/metabolismo , Células HeLa , Humanos , Chaperonas Moleculares/química , Receptores de Vasopressinas/química , Receptores de Vasopressinas/genética
3.
Proc Natl Acad Sci U S A ; 110(52): 21030-5, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24324164

RESUMO

Mutations in receptors, ion channels, and enzymes are frequently recognized by the cellular quality control system as misfolded and retained in the endoplasmic reticulum (ER) or otherwise misrouted. Retention results in loss of function at the normal site of biological activity and disease. Pharmacoperones are target-specific small molecules that diffuse into cells and serve as folding templates that enable mutant proteins to pass the criteria of the quality control system and route to their physiologic site of action. Pharmacoperones of the gonadotropin releasing hormone receptor (GnRHR) have efficacy in cell culture systems, and their cellular and biochemical mechanisms of action are known. Here, we show the efficacy of a pharmacoperone drug in a small animal model, a knock-in mouse, expressing a mutant GnRHR. This recessive mutation (GnRHR E(90)K) causes hypogonadotropic hypogonadism (failed puberty associated with low or apulsatile luteinizing hormone) in both humans and in the mouse model described. We find that pulsatile pharmacoperone therapy restores E(90)K from ER retention to the plasma membrane, concurrently with responsiveness to the endogenous natural ligand, gonadotropin releasing hormone, and an agonist that is specific for the mutant. Spermatogenesis, proteins associated with steroid transport and steroidogenesis, and androgen levels were restored in mutant male mice following pharmacoperone therapy. These results show the efficacy of pharmacoperone therapy in vivo by using physiological, molecular, genetic, endocrine and biochemical markers and optimization of pulsatile administration. We expect that this newly appreciated approach of protein rescue will benefit other disorders sharing pathologies based on misrouting of misfolded protein mutants.


Assuntos
Hipogonadismo/tratamento farmacológico , Chaperonas Moleculares/farmacologia , Dobramento de Proteína/efeitos dos fármacos , Deficiências na Proteostase/genética , Receptores LHRH/genética , Testículo/fisiologia , Animais , Biomarcadores/metabolismo , Retículo Endoplasmático/metabolismo , Técnicas de Introdução de Genes , Hipogonadismo/genética , Masculino , Camundongos , Chaperonas Moleculares/uso terapêutico , Mutação/genética , Testículo/efeitos dos fármacos
4.
Pharmacol Res ; 83: 38-51, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24373832

RESUMO

A pharmacoperone (from "pharmacological chaperone") is a small molecule that enters cells and serves as molecular scaffolding in order to cause otherwise-misfolded mutant proteins to fold and route correctly within the cell. Pharmacoperones have broad therapeutic applicability since a large number of diseases have their genesis in the misfolding of proteins and resultant misrouting within the cell. Misrouting may result in loss-of-function and, potentially, the accumulation of defective mutants in cellular compartments. Most known pharmacoperones were initially derived from receptor antagonist screens and, for this reason, present a complex pharmacology, although these are highly target specific. In this summary, we describe efforts to produce high throughput screens that identify these molecules from chemical libraries as well as a mouse model which provides proof-of-principle for in vivo protein rescue using existing pharmacoperones.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Proteínas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Transporte Proteico/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química
5.
Subcell Biochem ; 63: 263-89, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23161143

RESUMO

G-protein-coupled receptors (GPCRs) are a large superfamily of plasma membrane proteins that play central roles in transducing endocrine, neural and -sensory signals. In humans, more than 30 disorders are associated with mutations in GPCRs and these proteins are common drug development targets, with 30-50% of drugs targeting them. GPCR mutants are frequently misfolded, recognized as defective by the cellular quality control system, retained in the endoplasmic reticulum and do not traffic to the plasma membrane. The use of small molecules chaperones (pharmacological chaperones or "pharmacoperones") to rescue misfolded GPCRs has provided a new approach for treatment of human diseases caused by misfolding and misrouting. This chapter provides an overview of the molecular basis of this approach using the human gonadotropin-releasing hormone receptor (hGnRHR) as model for treatment of conformational diseases provoked by -misfolded GPCRs.


Assuntos
Preparações Farmacêuticas/metabolismo , Dobramento de Proteína/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/metabolismo , Animais , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 107(9): 4454-8, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20160100

RESUMO

G protein-coupled receptors (GPCRs) play central roles in almost all physiological functions; mutations in GPCRs are responsible for more than 30 disorders. There is a great deal of information about GPCR structure but little information that directly relates structure to protein trafficking or to activation. The gonadotropin releasing hormone receptor, because of its small size among GPCRs, is amenable to preparation of mutants and was used in this study to establish the relation among a salt bridge, protein trafficking, and receptor activation. This bridge, between residues E(90) [located in transmembrane segment (TM) 2] and K(121) (TM3), is associated with correct trafficking to the plasma membrane. Agonists, but not antagonists, interact with residue K(121), and destabilize the TM2-TM3 association of the receptor in the plasma membrane. The hGnRHR mutant E(90)K has a broken salt bridge, which also destabilizes the TM2-TM3 association and is typically retained in the endoplasmic reticulum. We show that this mutant, if rescued to the plasma membrane by either of two different means, has constitutive activity and shows modified ligand specificity, revealing a role for the salt bridge in receptor activation, ligand specificity, trafficking, and structure. The data indicate that destabilizing the TM2-TM3 relation for receptor activation, while requiring an intact salt bridge for correct trafficking, provides a mechanism that protects the cell from plasma membrane expression of constitutive activity.


Assuntos
Transporte Proteico , Receptores Acoplados a Proteínas G/metabolismo , Sais/metabolismo , Humanos , Receptores Acoplados a Proteínas G/química
7.
J Pharmacol Exp Ther ; 338(2): 430-42, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21527534

RESUMO

G protein-coupled receptors (GPCRs) play central roles in most physiological functions, and mutations in them cause heritable diseases. Whereas crystal structures provide details about the structure of GPCRs, there is little information that identifies structural features that permit receptors to pass the cellular quality control system or are involved in transition from the ground state to the ligand-activated state. The gonadotropin-releasing hormone receptor (GnRHR), because of its small size among GPCRs, is amenable to molecular biological approaches and to computer modeling. These techniques and interspecies comparisons are used to identify structural features that are important for both intracellular trafficking and GnRHR activation yet distinguish between these processes. Our model features two salt (Arg(38)-Asp(98) and Glu(90)-Lys(121)) and two disulfide (Cys(14)-Cys(200) and Cys(114)-Cys(196)) bridges, all of which are required for the human GnRHR to traffic to the plasma membrane. This study reveals that both constitutive and ligand-induced activation are associated with a "coincidence detector" that occurs when an agonist binds. The observed constitutive activation of receptors lacking Glu(90)-Lys(121), but not Arg(38)-Asp(98) ionic bridge, suggests that the role of the former connection is holding the receptor in the inactive conformation. Both the aromatic ring and hydroxyl group of Tyr(284) and the hydrogen bonding of Ser(217) are important for efficient receptor activation. Our modeling results, supported by the observed influence of Lys(191) from extracellular loop 2 (EL2) and a four-residue motif surrounding this loop on ligand binding and receptor activation, suggest that the positioning of EL2 within the seven-α-helical bundle regulates receptor stability, proper trafficking, and function.


Assuntos
Aminoácidos Básicos/química , Aminoácidos Básicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/química , Receptores LHRH/fisiologia , Animais , Sítios de Ligação/fisiologia , Células COS , Bovinos , Chlorocebus aethiops , Cristalografia por Raios X , Líquido Extracelular/metabolismo , Humanos , Camundongos , Mutação/fisiologia , Ligação Proteica/fisiologia , Estabilidade Proteica , Transporte Proteico , Ratos , Receptores Acoplados a Proteínas G/química , Sais/química , Sais/metabolismo
8.
Trends Pharmacol Sci ; 30(5): 228-33, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19307028

RESUMO

Proteins serve in cellular roles that necessitate structural precision, a requirement overseen by the cellular quality control system (QCS). By rejecting misfolded proteins, the QCS protects against aberrant activity. Misfolding and subsequent retention by the QCS results in proteins that might maintain function but become misrouted and cause disease. Correcting the misrouting of misfolded mutant proteins often restores activity and addresses the underlying disease. Because of its small size, the gonadotropin-releasing hormone receptor has been an excellent model for G-protein-coupled receptor trafficking and has recently enabled elucidation of both the requirements to pass the QCS and the biochemical mechanism of rescue by pharmacological chaperones; this information will now enable rational design of these therapeutic agents. Here, we summarize what is known about the relation between receptor structure and interactions with the QCS with a view toward therapeutic development based on the rescue of misfolded and, consequently, misrouted mutants with drugs.


Assuntos
Retículo Endoplasmático/metabolismo , Chaperonas Moleculares , Dobramento de Proteína/efeitos dos fármacos , Receptores LHRH/química , Animais , Fenômenos Fisiológicos Celulares , Cisteína/química , Descoberta de Drogas , Humanos , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/farmacologia , Proteínas Mutantes/efeitos dos fármacos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo
9.
Mol Endocrinol ; 23(2): 157-68, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19095769

RESUMO

The human GnRH receptor (hGnRHR), a G protein-coupled receptor, is a useful model for studying pharmacological chaperones (pharmacoperones), drugs that rescue misfolded and misrouted protein mutants and restore them to function. This technique forms the basis of a therapeutic approach of rescuing mutants associated with human disease and restoring them to function. The present study relies on computational modeling, followed by site-directed mutagenesis, assessment of ligand binding, effector activation, and confocal microscopy. Our results show that two different chemical classes of pharmacoperones act to stabilize hGnRHR mutants by bridging residues D(98) and K(121). This ligand-mediated bridge serves as a surrogate for a naturally occurring and highly conserved salt bridge (E(90)-K(121)) that stabilizes the relation between transmembranes 2 and 3, which is required for passage of the receptor through the cellular quality control system and to the plasma membrane. Our model was used to reveal important pharmacophoric features, and then identify a novel chemical ligand, which was able to rescue a D(98) mutant of the hGnRHR that could not be rescued as effectively by previously known pharmacoperones.


Assuntos
Modelos Moleculares , Chaperonas Moleculares/metabolismo , Receptores Acoplados a Proteínas G , Receptores LHRH , Animais , Bovinos , Membrana Celular/metabolismo , Simulação por Computador , Humanos , Ligantes , Estrutura Molecular , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Conformação Proteica , Transporte Proteico/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo
10.
Sci Rep ; 10(1): 10579, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601341

RESUMO

Molecules that correct the folding of protein mutants, restoring their functional trafficking, are called pharmacoperones. Most are clinically irrelevant and possess intrinsic antagonist or agonist activity. Here, we identify compounds capable of rescuing the activity of mutant gonadotropin-releasing hormone receptor or GnRHR which, is sequestered within the cell and if dysfunctional leads to Hypogonadotropic Hypogonadism. To do this we screened the E90K GnRHR mutant vs. a library of 645,000 compounds using a cell-based calcium detection system. Ultimately, we identified 399 compounds with EC50 ≤ 5 µM with no effect in counterscreen assays. Medicinal chemistry efforts confirmed activity of 70 pure samples and mode of action studies, including radioligand binding, inositol phosphate, and toxicity assays, proved that we have a series of tractable compounds that can be categorized into structural clusters. These early lead molecules rescue mutant GnRHR function and are neither agonist nor antagonists of the GnRHR cognate receptor, a feature required for potential clinical utility.


Assuntos
Receptores LHRH/agonistas , Receptores LHRH/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Cálcio/metabolismo , Avaliação Pré-Clínica de Medicamentos , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/metabolismo , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Fosfatos de Inositol/metabolismo , Mutação , Dobramento de Proteína , Transporte Proteico , Receptores LHRH/genética
11.
Mol Cell Endocrinol ; 299(2): 137-45, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19059461

RESUMO

In order to serve as enzymes, receptors and ion channels, proteins require structural precision. This is monitored by a cellular quality control system (QCS) that rejects misfolded proteins and thereby protects the cell against aberrant activity. Misfolding can result in protein molecules that retain intrinsic function, yet become misrouted within the cell; these cease to perform normally and result in disease. A therapeutic opportunity exists to correct misrouting and rescue mutants using "pharmacoperones" (small molecular folding templates, often peptidomimetics, which promote correct folding and rescue) thereby restoring function and potentially curing the underlying disease. Because of its small size, the GnRH (gonadotropin-releasing hormone) receptor (GnRHR) is an excellent model for GPCR (G protein-coupled receptor) and has allowed elucidation of the precise biochemical mechanism of pharmacoperone action necessary for rational design of new therapeutic agents. This review summarizes what has been learned about the structural requirements of the GnRHR that govern its interaction with the QCS and now presents the potential for the rational design of pharmacoperones. Because of the role of protein processing, this approach is likely to be applicable to other GCPCs and other proteins in general.


Assuntos
Doença , Saúde , Receptores LHRH/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Humanos , Dobramento de Proteína , Transporte Proteico , Receptores LHRH/química
12.
Mol Cell Endocrinol ; 298(1-2): 84-8, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-18848862

RESUMO

A thienopyr(im)idine (Org41841) activates the luteinizing hormone (LH) receptor but does not compete with the natural ligand binding site and does not show agonistic action on the follicle-stimulating hormone receptor (hFSHR) at sub-millimolar concentrations. When this drug is preincubated at sub-micromolar concentrations with host cells expressing the hFSHR, and then washed out, binding analysis and assessment of receptor-effector coupling show that it increases plasma membrane expression of the hFSHR. Real-time PCR shows that this effect did not result from increased hFSHR mRNA accumulation. It is possible that Org41841 behaves as a pharmacoperone, a drug which increases the percentage of newly synthesized receptor routing to the membrane. Like pharmacoperones for other receptors, this drug was able to rescue a particular mutant hFSHR (A(189)V) associated with misrouting and endoplasmic reticulum retention, although other mutants could not be rescued. This is potentially the first member of the pharmacoperone drug class which binds at a site that is distinctive from the ligand binding site.


Assuntos
Membrana Celular/efeitos dos fármacos , Piridinas/farmacologia , Receptores do FSH/metabolismo , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Humanos , Modelos Biológicos , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Piridinas/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacologia , Receptores do FSH/genética , Receptores do LH/agonistas , Tiofenos/metabolismo , Tiofenos/farmacologia , Transfecção
14.
Emerg Top Life Sci ; 3(1): 39-52, 2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33523195

RESUMO

Proteostasis refers to the process whereby the cell maintains in equilibrium the protein content of different compartments. This system consists of a highly interconnected network intended to efficiently regulate the synthesis, folding, trafficking, and degradation of newly synthesized proteins. Molecular chaperones are key players of the proteostasis network. These proteins assist in the assembly and folding processes of newly synthesized proteins in a concerted manner to achieve a three-dimensional structure compatible with export from the endoplasmic reticulum to other cell compartments. Pharmacologic interventions intended to modulate the proteostasis network and tackle the devastating effects of conformational diseases caused by protein misfolding are under development. These include small molecules called pharmacoperones, which are highly specific toward the target protein serving as a molecular framework to cause misfolded mutant proteins to fold and adopt a stable conformation suitable for passing the scrutiny of the quality control system and reach its correct location within the cell. Here, we review the main components of the proteostasis network and how pharmacoperones may be employed to correct misfolding of two G protein-coupled receptors, the vasopressin 2 receptor and the gonadotropin-releasing hormone receptor, whose mutations lead to X-linked nephrogenic diabetes insipidus and congenital hypogonadotropic hypogonadism in humans respectively.

15.
FASEB J ; 21(2): 384-92, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17172315

RESUMO

The primate GnRH receptor (GnRHR) is a GPCR (G-protein-coupled receptor) that transduces both amplitude- and frequency-modulated signals; each modality conveys information that regulates primate reproduction. Slower GnRH pulses favor release (and higher circulating levels) of pituitary FSH, while faster pulses favor LH release. We used radioligand binding and inositol phosphate production (a measure of G-protein coupling) in association with mutational analysis to identify the impact of evolved sequence specializations that regulate receptor concentration at the plasma membrane and Kd in primate GnRHRs. Our results show that mutations appear to provide a mechanism that allows independent adjustment of response sensitivity and squelching (suppression) of low-level signals (noise), both desirable features for recognition of frequency-modulated signals. We identify specific amino acid residues that appear to be involved in these processes. This investigation occurred in light of recent observations that restriction of GnRHR plasma membrane expression developed under strong convergent pressure and concurrently with the complex pattern of cyclicity associated with primate reproduction. The findings present an evolved means for increased effectiveness of detection of a frequency-modulated signal and provide a strategy to identify similar mechanisms in other receptors.


Assuntos
Proteínas Mutantes/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/metabolismo , Sequência de Aminoácidos , Animais , Busserrelina/farmacologia , Células COS , Chlorocebus aethiops , Sequência Conservada , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Cães , Humanos , Fosfatos de Inositol/metabolismo , Lisina/química , Lisina/genética , Lisina/metabolismo , Camundongos , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Proteínas Mutantes/química , Proteínas Mutantes/genética , Primatas , Ligação Proteica/efeitos dos fármacos , Ensaio Radioligante , Ratos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores LHRH/química , Receptores LHRH/genética , Especificidade da Espécie , Transfecção
16.
J Med Chem ; 50(9): 2067-77, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17402723

RESUMO

A series of acyline analogues incorporating l- and d-isomers of S-arylated/alkylated norcysteines [Ncy(R), where R is 2-naphthyl, methyl, and isopropyl] at positions 1, 4, 7, and 10 were synthesized. Some of these analogues were mono- and dioxidized to sulfoxides and sulfones. All of the analogues of acyline were screened for the antagonism of the GnRH-induced response in a reporter gene assay in HEK-293 cells expressing the human GnRH receptor. Nine of the analogues (9, 11, 15, 16, 17, 19, 20, 21, and 22) had antagonistic potency (IC50 < 2 nM) similar to that of acyline (IC50 = 0.52 nM) in this assay. Selected analogues (9, 11, 15, 16, 19, and 21) were tested in vitro for their antagonism at the rat GnRH-R in a reporter gene assay as well as in an in vivo intact male rat assay. Analogues 9 and 15 were the most potent in suppressing testosterone levels.


Assuntos
Cisteína/análogos & derivados , Cisteína/síntese química , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Oligopeptídeos/síntese química , Animais , Butiratos/síntese química , Butiratos/farmacologia , Linhagem Celular , Chlorocebus aethiops , Cisteína/farmacologia , Genes Reporter , Hormônio Liberador de Gonadotropina/genética , Humanos , Leucina/análogos & derivados , Leucina/síntese química , Leucina/farmacologia , Masculino , Naftalenos/síntese química , Naftalenos/farmacologia , Oligopeptídeos/farmacologia , Oxirredução , Ratos , Ratos Sprague-Dawley , Solubilidade , Estereoisomerismo , Sulfonas/síntese química , Sulfonas/farmacologia , Sulfóxidos/síntese química , Sulfóxidos/farmacologia , Testosterona/antagonistas & inibidores , Testosterona/sangue
17.
Mol Cell Endocrinol ; 272(1-2): 77-85, 2007 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-17555869

RESUMO

All reported GnRH receptor mutants (causing human hypogonadotropic hypogonadism) are misfolded proteins that cannot traffic to the plasma membrane. Pharmacoperones correct misfolding and rescue mutants, routing them to the plasma membrane where they regain function. Because pharmacoperones are often peptidomimetic antagonists, these must be removed for receptor function after rescue; in vivo this necessitates pulsatile pharmacoperone administration. As an antecedent to in vivo studies, we determined whether pharmacoperones need to be present at the time of synthesis or whether previously misfolded proteins could be refolded and rescued. Accordingly, we blocked either protein synthesis or intra-cellular transport. Biochemical and morphological studies using 12 mutants and 10 pharmacoperones representing three different chemical classes show that previously synthesized mutant proteins, retained by the quality control system (QCS), are rescued by pharmacoperones, showing that pharmacoperone administration in vivo likely need not consider whether the target protein is being synthesized at the time of drug administration.


Assuntos
Chaperonas Moleculares/farmacologia , Dobramento de Proteína , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/metabolismo , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Células COS , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Células HeLa , Humanos , Indóis/farmacologia , Inositol/farmacologia , Mimetismo Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transporte Proteico/efeitos dos fármacos , Piridinas/farmacologia , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores LHRH/química , Receptores LHRH/genética , Transfecção
18.
Mol Endocrinol ; 20(12): 3035-41, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16556733

RESUMO

Recent studies reveal that a number of G protein-coupled receptors (GPCRs) and other proteins are expressed inefficiently at the site normally associated with their biological action. In the case of some GPCRs, large amounts of receptor (perhaps more than half) may be destroyed without ever binding ligand or even arriving at the plasma membrane. For the human GnRH receptor (GnRHR), this apparent inefficiency has evolved under strong and convergent evolutionary pressure. The result is a human GnRHR molecule that is delicately balanced between either expression at the plasma membrane (PM) or retention/degradation in the endoplasmic reticulum, an effect mediated by engagement with the cellular quality control system. This balance appears to be the reason that the human receptor, but not the rat or mouse counterpart (which are more robustly routed to the PM), is highly susceptible to single-point mutations that result in disease. A single change in net charge is sufficient to tip the balance in favor of the endoplasmic reticulum and diminish GnRHR available at the PM. The apparent paradox that results from observing convergent pressure for evolution of a receptor that is both inefficiently produced and highly susceptible to mutational disease suggests that this approach must offer a strong advantage. This review focuses on the evolved mechanisms and considers that this is an underappreciated mechanism by which the cell controls functional levels of receptors and other proteins at the posttranslational level.


Assuntos
Membrana Celular/metabolismo , Evolução Molecular , Receptores Acoplados a Proteínas G/metabolismo , Animais , Humanos , Camundongos , Óperon , Biossíntese de Proteínas , Dobramento de Proteína , Ratos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética
19.
PLoS One ; 12(8): e0181830, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28767678

RESUMO

Pharmacoperones are small molecules that diffuse into cells and rescue misfolded, mistrafficked protein mutants, restoring their function. These substances act with high target specificity, serving as templates to fold (or refold) receptors, enzymes, ion channels or other proteins and enable them to pass the scrutiny of the cellular quality control system ("rescue"). In the present study we demonstrate that a rescued mutant (L83Q) of the vasopressin type 2 receptor (V2R), shows a strong bias for Gs coupling unlike the WT V2 receptor, which couples to both Gs and Gq/11. Failure of the mutant to couple to Gq/11 was not due to a limiting quantity of G-proteins since other Gq/11-coupled receptors (WT V2R, histamine receptor and muscarinic receptor) responded appropriately to their ligands. Transfection with DNA encoding Gq enabled the V2 receptor mutant to couple to this G protein, but only modestly compared with the WT receptor. Fourteen V2R mutant pharmacoperones, of multiple chemical classes, obtained from a high throughput screen of a 660,000 structure library, and one V2R peptidomimetic antagonist rescues L83Q. The rescued mutant shows similar bias with all pharmacoperones identified, suggesting that the bias is intrinsic to the mutant protein's structure, rather than due to the chemical class of the pharmacoperone. In the case of V2R mutant Y128S, rescue with a pharmacoperone revealed constitutive activity, also with bias for Gs, although both IP and cAMP were produced in response to agonist. These results suggest that particular rescued receptor mutants show functional characteristics that differ from the WT receptor; a finding that may be important to consider as pharmacoperones are developed as therapeutic agents.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Morfolinas/farmacologia , Mutação , Receptores de Vasopressinas/genética , Compostos de Espiro/farmacologia , Antagonistas dos Receptores de Hormônios Antidiuréticos/química , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , AMP Cíclico/metabolismo , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Modelos Moleculares , Morfolinas/química , Receptores de Vasopressinas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Compostos de Espiro/química
20.
SLAS Discov ; 22(7): 887-896, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28346094

RESUMO

Primary hyperoxaluria is the underlying cause of oxalosis and is a life-threatening autosomal recessive disease, for which treatment may require dialysis or dual liver-kidney transplantation. The most common primary hyperoxaluria type 1 (PH1) is caused by genetic mutations of a liver-specific enzyme alanine:glyoxylate aminotransferase (AGT), which results in the misrouting of AGT from the peroxisomes to the mitochondria. Pharmacoperones are small molecules with the ability to modify misfolded proteins and route them correctly within the cells, which may present an effective strategy to treat AGT misrouting in PH1 disorders. We miniaturized a cell-based high-content assay into 1536-well plate format and screened ~4200 pharmacologically relevant compounds including Food and Drug Administration, European Union, and Japanese-approved drugs. This assay employs CHO cells stably expressing AGT-170, a mutant that predominantly resides in the mitochondria, where we monitor for its relocation to the peroxisomes through automated image acquisition and analysis. The miniaturized 1536-well assay yielded a Z' averaging 0.70 ± 0.07. Three drugs were identified as potential pharmacoperones from this pilot screen, demonstrating the applicability of this assay for large-scale high-throughput screening.


Assuntos
Hiperoxalúria/tratamento farmacológico , Ionóforos/farmacologia , Nefropatias/tratamento farmacológico , Animais , Células CHO , Cricetulus , Avaliação Pré-Clínica de Medicamentos/métodos , Hiperoxalúria/genética , Hiperoxalúria/metabolismo , Hiperoxalúria Primária/tratamento farmacológico , Hiperoxalúria Primária/genética , Hiperoxalúria Primária/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Transplante de Rim/métodos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mutação/genética , Peroxissomos/efeitos dos fármacos , Peroxissomos/genética , Peroxissomos/metabolismo , Diálise Renal/métodos , Transaminases/genética , Transaminases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA