Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Exp Dermatol ; 32(7): 999-1006, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37009806

RESUMO

Thermoregulation and heat dissipation by sweat production and evaporation are vital for human survival. However, hyperhidrosis or excessive perspiration might affect people's quality of life by causing discomfort and stress. The prolonged use of classical antiperspirants, anticholinergic medications or botulinum toxin injections for persistent hyperhidrosis might produce diverse side effects that limit their clinical use. Inspired by botox molecular mode of action, we used an in silico molecular modelling approach to design novel peptides to target neuronal acetylcholine exocytosis by interfering with the Snapin-SNARE complex formation. Our exhaustive design rendered the selection of 11 peptides that decreased calcium-dependent vesicle exocytosis in rat DRG neurons, reducing αCGRP release and TRPV1 inflammatory sensitization. The most potent peptides were palmitoylated peptides SPSR38-4.1 and SPSR98-9.1 that significantly suppressed acetylcholine release in vitro in human LAN-2 neuroblastoma cells. Noteworthy, local acute and chronic administration of SPSR38-4.1 peptide significantly decreased, in a dose-dependent manner, pilocarpine-induced sweating in an in vivo mouse model. Taken together, our in silico approach lead to the identification of active peptides able to attenuate excessive sweating by modulating neuronal acetylcholine exocytosis, and identified peptide SPSR38-4.1 as a promising new antihyperhidrosis candidate for clinical development.


Assuntos
Antiperspirantes , Hiperidrose , Humanos , Ratos , Camundongos , Animais , Antiperspirantes/farmacologia , Qualidade de Vida , Acetilcolina/farmacologia , Acetilcolina/uso terapêutico , Hiperidrose/tratamento farmacológico , Hiperidrose/etiologia , Peptídeos/química , Exocitose/fisiologia , Neurônios/fisiologia
2.
Pharmacol Ther ; 240: 108297, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36202261

RESUMO

Chronic pain is a major burden for the society and remains more prevalent and severe in females. The presence of chronic pain is linked to persistent alterations in the peripheral and the central nervous system. One of the main types of peripheral pain transducers are the transient receptor potential channels (TRP), also known as thermoTRP channels, which intervene in the perception of hot and cold external stimuli. These channels, and especially TRPV1, TRPA1 and TRPM8, have been subjected to profound investigation because of their role as thermosensors and also because of their implication in acute and chronic pain. Surprisingly, their sensitivity to endogenous signaling has been far less studied. Cumulative evidence suggests that the function of these channels may be differently modulated in males and females, in part through sexual hormones, and this could constitute a significant contributor to the sex differences in chronic pain. Here, we review the exciting advances in thermoTRP pharmacology for males and females in two paradigmatic types of chronic pain with a strong peripheral component: chronic migraine and chemotherapy-induced peripheral neuropathy (CIPN). The possibilities of peripheral druggability offered by these channels and the differential exploitation for men and women represent a development opportunity that will lead to a significant increment of the armamentarium of analgesic medicines for personalized chronic pain treatment.


Assuntos
Dor Crônica , Transtornos de Enxaqueca , Doenças do Sistema Nervoso Periférico , Termorreceptores , Canais de Potencial de Receptor Transitório , Feminino , Humanos , Masculino , Analgésicos/uso terapêutico , Dor Crônica/tratamento farmacológico , Transtornos de Enxaqueca/tratamento farmacológico , Caracteres Sexuais , Canais de Potencial de Receptor Transitório/metabolismo , Antineoplásicos/efeitos adversos , Termorreceptores/metabolismo
3.
Nat Struct Mol Biol ; 29(8): 781-790, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35948766

RESUMO

Many disordered proteins conserve essential functions in the face of extensive sequence variation, making it challenging to identify the mechanisms responsible for functional selection. Here we identify the molecular mechanism of functional selection for the disordered adenovirus early gene 1A (E1A) protein. E1A competes with host factors to bind the retinoblastoma (Rb) protein, subverting cell cycle regulation. We show that two binding motifs tethered by a hypervariable disordered linker drive picomolar affinity Rb binding and host factor displacement. Compensatory changes in amino acid sequence composition and sequence length lead to conservation of optimal tethering across a large family of E1A linkers. We refer to this compensatory mechanism as conformational buffering. We also detect coevolution of the motifs and linker, which can preserve or eliminate the tethering mechanism. Conformational buffering and motif-linker coevolution explain robust functional encoding within hypervariable disordered linkers and could underlie functional selection of many disordered protein regions.


Assuntos
Proteínas Intrinsicamente Desordenadas , Proteínas E1A de Adenovirus/química , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Sequência de Aminoácidos , Proteínas Intrinsicamente Desordenadas/química , Ligação Proteica , Domínios Proteicos , Proteína do Retinoblastoma/metabolismo
4.
Int J Mol Sci ; 22(5)2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33673444

RESUMO

Transient receptor potential cation channel subfamily M member 8 (TRPM8) is a Ca2+ non-selective ion channel implicated in a variety of pathological conditions, including cancer, inflammatory and neuropathic pain. In previous works we identified a family of chiral, highly hydrophobic ß-lactam derivatives, and began to intuit a possible effect of the stereogenic centers on the antagonist activity. To investigate the influence of configuration on the TRPM8 antagonist properties, here we prepare and characterize four possible diastereoisomeric derivatives of 4-benzyl-1-[(3'-phenyl-2'-dibenzylamino)prop-1'-yl]-4-benzyloxycarbonyl-3-methyl-2-oxoazetidine. In microfluorography assays, all isomers were able to reduce the menthol-induced cell Ca2+ entry to larger or lesser extent. Potency follows the order 3R,4R,2'R > 3S,4S,2'R ≅ 3R,4R,2'S > 3S,4S,2'S, with the most potent diastereoisomer showing a half inhibitory concentration (IC50) in the low nanomolar range, confirmed by Patch-Clamp electrophysiology experiments. All four compounds display high receptor selectivity against other members of the TRP family. Furthermore, in primary cultures of rat dorsal root ganglion (DRG) neurons, the most potent diastereoisomers do not produce any alteration in neuronal excitability, indicating their high specificity for TRPM8 channels. Docking studies positioned these ß-lactams at different subsites by the pore zone, suggesting a different mechanism than the known N-(3-aminopropyl)-2-[(3-methylphenyl)methoxy]-N-(2-thienylmethyl)-benzamide (AMTB) antagonist.


Assuntos
Neurônios/metabolismo , Fenilalanina/farmacologia , Canais de Cátion TRPM/antagonistas & inibidores , beta-Lactamas/farmacologia , Animais , Células Cultivadas , Gânglios Espinais/metabolismo , Simulação de Acoplamento Molecular , Neurônios/efeitos dos fármacos , Fenilalanina/análogos & derivados , Fenilalanina/química , Ratos , Relação Estrutura-Atividade , beta-Lactamas/química
5.
Expert Opin Investig Drugs ; 29(11): 1209-1222, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32941080

RESUMO

INTRODUCTION: Thermo transient receptor potential (thermoTRP) channels are some of the most intensely pursued therapeutic targets of the past decade. They are considered promising targets of numerous diseases including chronic pain and cancer. Modulators of these proteins, in particular TRPV1-4, TRPM8 and TRPA1, have reached clinical development, but none has been approved for clinical practice yet. AREAS COVERED: The therapeutic potential of targeting thermoTRP channels is discussed. The discussion is centered on our experience and on available data found in SciFinder, PubMed, and ClinicalTrials.gov database from the past decade. This review focuses on the therapeutic progress concerning this family of channels, including strategies to improve their therapeutic index for overcoming adverse effects. EXPERT OPINION: Although thermoTRPs are pivotal drug targets, translation to the clinic has faced two key problems, (i) unforeseen side effects in Phase I trials and, (ii) poor clinical efficacy in Phase II trials. Thus, there is a need for (i) an enhanced understanding of the physiological role of these channels in tissues and organs and (ii) the development of human-based pre-clinical models with higher clinical translation. Furthermore, progress in nanotechnology-based delivery strategies will positively impact thermoTRP human pharmacology.


Assuntos
Desenvolvimento de Medicamentos , Drogas em Investigação/farmacologia , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Animais , Dor Crônica/tratamento farmacológico , Dor Crônica/patologia , Sistemas de Liberação de Medicamentos , Drogas em Investigação/efeitos adversos , Humanos , Nanotecnologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Canais de Potencial de Receptor Transitório/metabolismo
6.
Sci Rep ; 10(1): 14154, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843690

RESUMO

The cool sensor transient receptor potential melastatin channel 8 (TRPM8) is highly expressed in trigeminal and dorsal root ganglia, playing a key role in cold hypersensitivity associated to different peripheral neuropathies. Moreover, these channels are aberrantly expressed in different cancers, and seem to participate in tumor progression, survival and invasion. Accordingly, the search for potent and selective TRPM8 modulators attracted great interest in recent years. We describe new heterocyclic TRPM8 antagonist chemotypes derived from N-cloroalkyl phenylalaninol-Phe conjugates. The cyclization of these conjugates afforded highly substituted ß-lactams and/or 2-ketopiperazine (KP) derivatives, with regioselectivity depending on the N-chloroalkyl group and the configuration. These derivatives behave as TRPM8 antagonists in the Ca2+ microfluorometry assay, and confirmed electrophysiologically for the best enantiopure ß-lactams 24a and 29a (IC50, 1.4 and 0.8 µM). Two putative binding sites by the pore zone, different from those found for typical agonists and antagonists, were identified by in silico studies for both ß-lactams and KPs. ß-Lactams 24a and 29a display antitumor activity in different human tumor cell lines (micromolar potencies, A549, HT29, PSN1), but correlation with TRPM8 expression could not be established. Additionally, compound 24a significantly reduced cold allodynia in a mice model of oxaliplatin-induced peripheral neuropathy.


Assuntos
Analgésicos/uso terapêutico , Antineoplásicos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Piperazinas/uso terapêutico , Canais de Cátion TRPM/antagonistas & inibidores , beta-Lactamas/uso terapêutico , Analgésicos/síntese química , Analgésicos/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Temperatura Baixa/efeitos adversos , Simulação por Computador , Citofotometria , Avaliação Pré-Clínica de Medicamentos , Masculino , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Molecular , Oxaliplatina/toxicidade , Técnicas de Patch-Clamp , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Piperazinas/síntese química , Piperazinas/farmacologia , Relação Estrutura-Atividade , beta-Lactamas/síntese química , beta-Lactamas/farmacologia
7.
Methods Mol Biol ; 1987: 187-206, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31028681

RESUMO

The implication of several TRP ion channels (e.g., TRPV1) in diverse physiological and pathological processes has signaled them as pivotal drug targets. Consequently, the identification of selective and potent ligands for these channels is of great interest in pharmacology and biomedicine. However, a major challenge in the design of modulators is ensuring the specificity for their intended targets. In recent years, the emergence of high-resolution structures of ion channels facilitates the computer-assisted drug design at molecular levels. Here we describe some computational methods and general protocols to discover channel modulators, including homology modelling, docking and virtual screening, and structure-based peptide design.


Assuntos
Desenho de Fármacos , Simulação de Acoplamento Molecular/métodos , Simulação de Dinâmica Molecular , Canais de Cátion TRPV/química , Simulação por Computador , Desenho Assistido por Computador , Descoberta de Drogas , Ligantes , Peptídeos/síntese química , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo
8.
Sci Rep ; 7(1): 10766, 2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28883526

RESUMO

The mammalian transient receptor potential melastatin channel 8 (TRPM8), highly expressed in trigeminal and dorsal root ganglia, mediates the cooling sensation and plays an important role in the cold hypersensitivity characteristic of some types of neuropathic pain, as well as in cancer. Consequently, the identification of selective and potent ligands for TRPM8 is of great interest. Here, a series of compounds, having a ß-lactam central scaffold, were prepared to explore the pharmacophore requirements for TRPM8 modulation. Structure-activity studies indicate that the minimal requirements for potent ß-lactam-based TRPM8 blockers are hydrophobic groups (benzyl preferentially or t Bu) on R1, R2, R3 and R5 and a short N-alkyl chain (≤3 carbons). The best compounds in the focused library (41 and 45) showed IC50 values of 46 nM and 83 nM, respectively, in electrophysiology assays. These compounds selectively blocked all modalities of TRPM8 activation, i.e. menthol, voltage, and temperature. Molecular modelling studies using a homology model of TRPM8 identified two putative binding sites, involving networks of hydrophobic interactions, and suggesting a negative allosteric modulation through the stabilization of the closed state. Thus, these ß-lactams provide a novel pharmacophore scaffold to evolve TRPM8 allosteric modulators to treat TRPM8 channel dysfunction.


Assuntos
Canais de Cátion TRPM/antagonistas & inibidores , beta-Lactamas/farmacologia , Linhagem Celular Tumoral , Temperatura Baixa , Estimulação Elétrica , Eletrofisiologia , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Mentol , beta-Lactamas/síntese química , beta-Lactamas/química
9.
Drug Des Devel Ther ; 9: 5877-95, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26604687

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) is a well-characterized member of the PPAR family that is predominantly expressed in adipose tissue and plays a significant role in lipid metabolism, adipogenesis, glucose homeostasis, and insulin sensitization. Full agonists of synthetic thiazolidinediones (TZDs) have been therapeutically used in clinical practice to treat type 2 diabetes for many years. Although it can effectively lower blood glucose levels and improve insulin sensitivity, the administration of TZDs has been associated with severe side effects. Based on recent evidence obtained with plant-derived polyphenols, the present in silico study aimed at finding new selective human PPARγ (hPPARγ) modulators that are able to improve glucose homeostasis with reduced side effects compared with TZDs. Docking experiments have been used to select compounds with strong binding affinity (ΔG values ranging from -10.0±0.9 to -11.4±0.9 kcal/mol) by docking against the binding site of several X-ray structures of hPPARγ. These putative modulators present several molecular interactions with the binding site of the protein. Additionally, most of the selected compounds have favorable druggability and good ADMET properties. These results aim to pave the way for further bench-scale analysis for the discovery of new modulators of hPPARγ that do not induce any side effects.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , PPAR gama/efeitos dos fármacos , Polifenóis/farmacologia , Sítios de Ligação , Glicemia/efeitos dos fármacos , Simulação por Computador , Diabetes Mellitus Tipo 2/fisiopatologia , Desenho de Fármacos , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/isolamento & purificação , Insulina/metabolismo , Resistência à Insulina , Simulação de Acoplamento Molecular , PPAR gama/metabolismo , Plantas Medicinais/química , Polifenóis/efeitos adversos , Polifenóis/isolamento & purificação , Tiazolidinedionas/farmacologia
10.
Nat Commun ; 6: 8095, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26311398

RESUMO

Hyaluronan (HA) is present in the extracellular matrix of all body tissues, including synovial fluid in joints, in which it behaves as a filter that buffers transmission of mechanical forces to nociceptor nerve endings thereby reducing pain. Using recombinant systems, mouse-cultured dorsal root ganglia (DRG) neurons and in vivo experiments, we found that HA also modulates polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channels. HA diminishes heat, pH and capsaicin (CAP) responses, thus reducing the opening probability of the channel by stabilizing its closed state. Accordingly, in DRG neurons, HA decreases TRPV1-mediated impulse firing and channel sensitization by bradykinin. Moreover, subcutaneous HA injection in mice reduces heat and capsaicin nocifensive responses, whereas the intra-articular injection of HA in rats decreases capsaicin joint nociceptor fibres discharge. Collectively, these results indicate that extracellular HA reduces the excitability of the ubiquitous TRPV1 channel, thereby lowering impulse activity in the peripheral nociceptor endings underlying pain.


Assuntos
Adjuvantes Imunológicos/farmacologia , Ácido Hialurônico/farmacologia , Neurônios/efeitos dos fármacos , Dor Nociceptiva , Nociceptores/efeitos dos fármacos , Joelho de Quadrúpedes/efeitos dos fármacos , Canais de Cátion TRPV/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Bradicinina/farmacologia , Células CHO , Cálcio/metabolismo , Capsaicina/farmacologia , Linhagem Celular Tumoral , Cricetulus , Gânglios Espinais/citologia , Células HEK293 , Temperatura Alta , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Modelos Moleculares , Mutagênese Sítio-Dirigida , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Fármacos do Sistema Sensorial/farmacologia , Joelho de Quadrúpedes/inervação , Canal de Cátion TRPA1 , Canais de Cátion TRPM/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/metabolismo , Vasodilatadores/farmacologia
11.
J Gen Physiol ; 143(3): 361-75, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24567510

RESUMO

The transient receptor potential vanilloid receptor subtype I (TRPV1) channel acts as a polymodal sensory receptor gated by chemical and physical stimuli. Like other TRP channels, TRPV1 contains in its C terminus a short, conserved domain called the TRP box, which is necessary for channel gating. Substitution of two TRP box residues-I696 and W697-with Ala markedly affects TRPV1's response to all activating stimuli, which indicates that these two residues play a crucial role in channel gating. We systematically replaced I696 and W697 with 18 native l-amino acids (excluding cysteine) and evaluated the effect on voltage- and capsaicin-dependent gating. Mutation of I696 decreased channel activation by either voltage or capsaicin; furthermore, gating was only observed with substitution of hydrophobic amino acids. Substitution of W697 with any of the 18 amino acids abolished gating in response to depolarization alone, shifting the threshold to unreachable voltages, but not capsaicin-mediated gating. Moreover, vanilloid-activated responses of W697X mutants showed voltage-dependent gating along with a strong voltage-independent component. Analysis of the data using an allosteric model of activation indicates that mutation of I696 and W697 primarily affects the allosteric coupling constants of the ligand and voltage sensors to the channel pore. Together, our findings substantiate the notion that inter- and/or intrasubunit interactions at the level of the TRP box are critical for efficient coupling of stimulus sensing and gate opening. Perturbation of these interactions markedly reduces the efficacy and potency of the activating stimuli. Furthermore, our results identify these interactions as potential sites for pharmacological intervention.


Assuntos
Ativação do Canal Iônico , Mutação de Sentido Incorreto , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Regulação Alostérica , Sítio Alostérico , Animais , Capsaicina/farmacologia , Células HEK293 , Humanos , Potenciais da Membrana , Ratos , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPV/química
12.
Endocr Pract ; 20(5): e75-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24449679

RESUMO

OBJECTIVE: BRAF mutations are the most common genetic alteration found in papillary thyroid carcinoma (PTC). Approximately, 90% correspond to BRAFV600E, although other less common BRAF mutations have been described. The aim of this study was to describe a new mutation on BRAF gene discovered on the previous thyroid cytology of a patient diagnosed with a follicular variant of PTC (FV-PTC). METHODS: The mutation was identified by independent cloning of the 2 alleles and direct sequencing in the previous cytology and tumor tissue samples from a patient diagnosed with FV-PTC. To elucidate the effect of the mutation on the structure and hence on the activating mechanism of the protein, the structures of BRAFI599Ins, BRAFT599Ins, BRAFV599Ins and BRAFV600E were modeled by using the reconstructed wild-type BRAF (BRAFWT) crystal structure. RESULTS: The novel mutation in BRAF consisted in the in-frame insertion of 3 nucleotides (TAA) after nucleotide 1795, resulting in the incorporation of an extra isoleucine residue at position 599 (BRAFI599Ins) of the protein. The structural comparison of BRAFI599Ins, BRAFT599Ins, BRAFV599Ins with BRAFWT, and BRAFV600E models revealed that the overall shape of the kinase was conserved in the protein produced by this novel mutation, except for the displacement of the activation loop (A-loop), as a direct consequence of the increase in loop size, and the exposition of 1 of the 2 residues involved in BRAF activation (T599), probably facilitating its phosphorylation. CONCLUSION: BRAFI599Ins mutation constitutes a new BRAF mutation affecting the length of the A-loop, which most likely facilitates BRAF activation by altering the A-loop conformation.

14.
Expert Opin Ther Pat ; 22(9): 999-1017, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22835143

RESUMO

INTRODUCTION: Thermosensory channels are a subfamily of the transient receptor potential (TRP) channel family that are activated by changes in the environmental temperature. These channels, known as thermoTRPs, cover the entire spectrum of temperatures, from noxious cold (< 15°C) to injurious heat (> 42°C). In addition, dysfunction of these channels contributes to the thermal hypersensitivity that accompanies painful conditions. Moreover, because of their wide tissue and cellular distribution, thermoTRPs are also involved in the pathophysiology of several diseases, from inflammation to cancer. AREAS COVERED: Although the number of thermoTRPs is increasing with the identification of novel members such as TRPM3, we will cover the recent advances in the pharmacology of the classical thermosensory channels, namely TRPV1, TRPV2, TRPV3, TRPV4, TRPM8 and TRPA1. This review will focus on the therapeutic progress carried out for all these channels and will highlight the tenet that TRPV1, TRPM8 and TRPA1 are the most exploited channels, and that the interest on TRPV3 and TRPV4 is growing with the first TRPV3 antagonist that moves into Phase-II clinical trials. In contrast, the pharmacology of TRPV2 is yet in its infancy. EXPERT OPINION: Despite the tremendous academic and industrial investment to develop therapeutic modulators of thermoTRPs, it apparently seems that we are still far from the first successful product, although hope is maintained high for all compounds currently in clinical trials. A major concern has been the appearance of side effects. A better knowledge of the thermosensory protein networks (signal-plexes), along with the application of system biology approaches may provide novel strategies to modulate thermoTRPs activity with improved therapeutic index. A case in point is TRPV1, where acting on interacting proteins is providing new therapeutic opportunities.


Assuntos
Dor/fisiopatologia , Canais de Cátion TRPV/metabolismo , Animais , Temperatura Baixa , Temperatura Alta , Humanos , Inflamação/fisiopatologia , Neoplasias/fisiopatologia , Patentes como Assunto
15.
Biochemistry ; 51(16): 3470-84, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22471585

RESUMO

Snapin is a 15 kDa protein present in neuronal and non-neuronal cells that has been implicated in the regulation of exocytosis and endocytosis. Protein kinase A (PKA) phosphorylates Snapin at Ser-50, modulating its function. Likewise, mutation of Cys-66, which mediates protein dimerization, impairs its cellular activity. Here, we have investigated the impact of mutating these two positions on protein oligomerization, structure, and thermal stability, along with the interaction with SNARE proteins. We found that recombinant purified Snapin in solution appears mainly as dimers in equilibrium with tetramers. The protein exhibits modest secondary structure elements and notable thermal stability. Mutation of Cys-66 to Ser abolished subunit dimerization, but not higher-order oligomers. This mutant augmented the presence of α-helical structure and slightly increased the protein thermal stability. Similarly, the S50A mutant, mimicking the unphosphorylated protein, also exhibited a higher helical secondary structure content than the wild type, along with greater thermal stability. In contrast, replacement of Ser-50 with Asp (S50D), emulating the protein-phosphorylated state, produced a loss of α-helical structure, concomitant with a decrease in protein thermal stability. In vitro, the wild type and mutants weakly interacted with SNAP-25 and the reconstituted SNARE complex, although S50D exhibited the strongest binding to the SNARE complex, consistent with the observed higher cellular activity of PKA-phosphorylated Snapin. Our observations suggest that the stronger binding of S50D to SNAREs might be due to a destabilization of tetrameric assemblies of Snapin that favor the interaction of protein dimers with the SNARE proteins. Therefore, phosphorylation of Ser-50 has an important impact on the protein structure and stability that appears to underlie its functional modulation.


Assuntos
Cisteína/genética , Mutação , Serina/genética , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética , Conformação Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Proteínas SNARE/química , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Proteínas de Transporte Vesicular/metabolismo
16.
J Neurochem ; 117(6): 995-1008, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21480900

RESUMO

Recently, we described estrogen and agonists of the G-protein coupled estrogen receptor GPR30 to induce protein kinase C (PKC)ε-dependent pain sensitization. PKCε phosphorylates the ion channel transient receptor potential, vanilloid subclass I (TRPV1) close to a novel microtubule-TRPV1 binding site. We now modeled the binding of tubulin to the TRPV1 C-terminus. The model suggests PKCε phosphorylation of TRPV1-S800 to abolish the tubulin-TRPV1 interaction. Indeed, in vitro PKCε phosphorylation of TRPV1 hindered tubulin-binding to TRPV1. In vivo, treatment of sensory neurons and F-11 cells with estrogen and the GPR30 agonist, G-1, resulted in microtubule destabilization and retraction of microtubules from filopodial structures. We found estrogen and G-1 to regulate the stability of the microtubular network via PKC phosphorylation of the PKCε-phosphorylation site TRPV1-S800. Microtubule disassembly was not, however, dependent on TRPV1 ion conductivity. TRPV1 knock-down in rats inverted the effect of the microtubule-modulating drugs, Taxol and Nocodazole, on estrogen-induced and PKCε-dependent mechanical pain sensitization. Thus, we suggest the C-terminus of TRPV1 to be a signaling intermediate downstream of estrogen and PKCε, regulating microtubule-stability and microtubule-dependent pain sensitization.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Microtúbulos/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Animais , Ligação Competitiva , Linhagem Celular , Ciclopentanos/farmacologia , Estrogênios/fisiologia , Gânglios Espinais/citologia , Técnicas de Silenciamento de Genes , Ativação do Canal Iônico , Ligantes , Masculino , Microtúbulos/ultraestrutura , Modelos Moleculares , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Dor/fisiopatologia , Fosforilação , Ligação Proteica , Proteína Quinase C-épsilon/fisiologia , Pseudópodes/ultraestrutura , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais , Canais de Cátion TRPV/genética , Tubulina (Proteína)/metabolismo
17.
FASEB J ; 25(5): 1628-40, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21307333

RESUMO

The transient receptor potential vanilloid 1 (TRPV1) channel is a thermosensory receptor implicated in diverse physiological and pathological processes. The TRP domain, a highly conserved region in the C terminus adjacent to the internal channel gate, is critical for subunit tetramerization and channel gating. Here, we show that cell-penetrating, membrane-anchored peptides patterned after this protein domain are moderate and selective TRPV1 antagonists both in vitro and in vivo, blocking receptor activity in intact rat primary sensory neurons and their peripheral axons with mean decline time of 30 min. The most potent lipopeptide, TRP-p5, blocked all modes of TRPV1 gating with micromolar efficacy (IC(50)<10 µM), without significantly affecting other thermoTRP channels. In contrast, its retrosequence or the corresponding sequences of other TRPV channels did not alter TRPV1 channel activity (IC(50)>100 µM). TRP-p5 did not affect the capsaicin sensitivity of the vanilloid receptor. Our data suggest that TRP-p5 interferes with protein-protein interactions at the level of the TRP domain that are essential for the "conformational" change that leads to gate opening. Therefore, these palmitoylated peptides, which we termed TRPducins, are noncompetitive, voltage-independent, sequence-specific TRPV1 blockers. Our findings indicate that TRPducin-like peptides may embody a novel molecular strategy that can be exploited to generate a selective pharmacological arsenal for the TRP superfamily of ion channels.


Assuntos
Peptídeos/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Animais , Animais Recém-Nascidos , Capsaicina/farmacologia , Linhagem Celular , Células Cultivadas , Eletrofisiologia , Células HEK293 , Humanos , Imuno-Histoquímica , Peptídeos/química , Ratos , Canais de Cátion TRPV/química
18.
Pharmaceutics ; 3(4): 932-53, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24309315

RESUMO

Ion channels are involved in a broad range of physiological and pathological processes. The implications of ion channels in a variety of diseases, including diabetes, epilepsy, hypertension, cancer and even chronic pain, have signaled them as pivotal drug targets. Thus far, drugs targeting ion channels were developed without detailed knowledge of the molecular interactions between the lead compounds and the target channels. In recent years, however, the emergence of high-resolution structures for a plethora of ion channels paves the way for computer-assisted drug design. Currently, available functional and structural data provide an attractive platform to generate models that combine substrate-based and protein-based approaches. In silico approaches include homology modeling, quantitative structure-activity relationships, virtual ligand screening, similarity and pharmacophore searching, data mining, and data analysis tools. These strategies have been frequently used in the discovery and optimization of novel molecules with enhanced affinity and specificity for the selected therapeutic targets. In this review we summarize recent applications of in silico methods that are being used for the development of ion channel drugs.

19.
J Mol Biol ; 388(4): 902-16, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19324052

RESUMO

A great challenge in the proteomics and structural genomics era is to discover protein structure and function, including the identification of biological partners. Experimental investigation is costly and time-consuming, making computational methods very attractive for predicting protein function. In this work, we used the existing structural information in the SH3 family to first extract all SH3 structural features important for binding and then used this information to select the right templates to homology model most of the Saccharomyces cerevisiae SH3 domains. Second, we classified, based on ligand orientation with respect to the SH3 domain, all SH3 peptide ligands into 29 conformations, of which 18 correspond to variants of canonical type I and type II conformations and 11 correspond to non-canonical conformations. Available SH3 templates were expanded by chimera construction to cover some sequence variability and loop conformations. Using the 29 ligand conformations and the homology models, we modelled all possible complexes. Using these complexes and in silico mutagenesis scanning, we constructed position-specific ligand binding matrices. Using these matrices, we determined which sequences will be favorable for every SH3 domain and then validated them with available experimental data. Our work also allowed us to identify key residues that determine loop conformation in SH3 domains, which could be used to model human SH3 domains and do target prediction. The success of this methodology opens the way for sequence-based, genome-wide prediction of protein-protein interactions given enough structural coverage.


Assuntos
Ligantes , Conformação Proteica , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Domínios de Homologia de src , Algoritmos , Sequência de Aminoácidos , Simulação por Computador , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Reprodutibilidade dos Testes , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência
20.
J Biol Chem ; 283(26): 18076-85, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18430729

RESUMO

The effects of the inactivating peptide from the eukaryotic Shaker BK(+) channel (the ShB peptide) on the prokaryotic KcsA channel have been studied using patch clamp methods. The data show that the peptide induces rapid, N-type inactivation in KcsA through a process that includes functional uncoupling of channel gating. We have also employed saturation transfer difference (STD) NMR methods to map the molecular interactions between the inactivating peptide and its channel target. The results indicate that binding of the ShB peptide to KcsA involves the ortho and meta protons of Tyr(8), which exhibit the strongest STD effects; the C4H in the imidazole ring of His(16); the methyl protons of Val(4), Leu(7), and Leu(10) and the side chain amine protons of one, if not both, the Lys(18) and Lys(19) residues. When a noninactivating ShB-L7E mutant is used in the studies, binding to KcsA is still observed but involves different amino acids. Thus, the strongest STD effects are now seen on the methyl protons of Val(4) and Leu(10), whereas His(16) seems similarly affected as before. Conversely, STD effects on Tyr(8) are strongly diminished, and those on Lys(18) and/or Lys(19) are abolished. Additionally, Fourier transform infrared spectroscopy of KcsA in presence of (13)C-labeled peptide derivatives suggests that the ShB peptide, but not the ShB-L7E mutant, adopts a beta-hairpin structure when bound to the KcsA channel. Indeed, docking such a beta-hairpin structure into an open pore model for K(+) channels to simulate the inactivating peptide/channel complex predicts interactions well in agreement with the experimental observations.


Assuntos
Proteínas de Bactérias/química , Epitopos/química , Canais de Potássio/química , Sequência de Aminoácidos , Aminoácidos/química , Eletrofisiologia , Proteínas de Escherichia coli/química , Lisina/química , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Mutação , Peptídeos/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Ligação Proteica , Espectroscopia de Infravermelho com Transformada de Fourier
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA