Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Biol Chem ; 297(6): 101355, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34717959

RESUMO

The ion pump Na+,K+-ATPase is a critical determinant of neuronal excitability; however, its role in the etiology of diseases of the central nervous system (CNS) is largely unknown. We describe here the molecular phenotype of a Trp931Arg mutation of the Na+,K+-ATPase catalytic α1 subunit in an infant diagnosed with therapy-resistant lethal epilepsy. In addition to the pathological CNS phenotype, we also detected renal wasting of Mg2+. We found that membrane expression of the mutant α1 protein was low, and ion pumping activity was lost. Arginine insertion into membrane proteins can generate water-filled pores in the plasma membrane, and our molecular dynamic (MD) simulations of the principle states of Na+,K+-ATPase transport demonstrated massive water inflow into mutant α1 and destabilization of the ion-binding sites. MD simulations also indicated that a water pathway was created between the mutant arginine residue and the cytoplasm, and analysis of oocytes expressing mutant α1 detected a nonspecific cation current. Finally, neurons expressing mutant α1 were observed to be depolarized compared with neurons expressing wild-type protein, compatible with a lowered threshold for epileptic seizures. The results imply that Na+,K+-ATPase should be considered a neuronal locus minoris resistentia in diseases associated with epilepsy and with loss of plasma membrane integrity.


Assuntos
Epilepsia/genética , Mutação de Sentido Incorreto , ATPase Trocadora de Sódio-Potássio/genética , Animais , Anticonvulsivantes/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Resistência a Medicamentos , Epilepsia/tratamento farmacológico , Epilepsia/patologia , Humanos , Lactente , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto/efeitos dos fármacos , Subunidades Proteicas/análise , Subunidades Proteicas/genética , ATPase Trocadora de Sódio-Potássio/análise , Xenopus
2.
Neuropharmacology ; 189: 108543, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33794243

RESUMO

Hyperekplexia is a rare sensorimotor syndrome characterized by pathological startle reflex in response to unexpected trivial stimuli for which there is no specific treatment. Neonates suffer from hypertonia and are at high risk of sudden death due to apnea episodes. Mutations in the human SLC6A5 gene encoding the neuronal glycine transporter GlyT2 may disrupt the inhibitory glycinergic neurotransmission and cause a presynaptic form of the disease. The phenotype of missense mutations giving rise to protein misfolding but maintaining residual activity could be rescued by facilitating folding or intracellular trafficking. In this report, we characterized the trafficking properties of two mutants associated with hyperekplexia (A277T and Y707C, rat numbering). Transporter molecules were partially retained in the endoplasmic reticulum showing increased interaction with the endoplasmic reticulum chaperone calnexin. One transporter variant had export difficulties and increased ubiquitination levels, suggestive of enhanced endoplasmic reticulum-associated degradation. However, the two mutant transporters were amenable to correction by calnexin overexpression. Within the search for compounds capable of rescuing mutant phenotypes, we found that the arachidonic acid derivative N-arachidonoyl glycine can rescue the trafficking defects of the two variants in heterologous cells and rat brain cortical neurons. N-arachidonoyl glycine improves the endoplasmic reticulum output by reducing the interaction transporter/calnexin, increasing membrane expression and improving transport activity in a comparable way as the well-established chemical chaperone 4-phenyl-butyrate. This work identifies N-arachidonoyl glycine as a promising compound with potential for hyperekplexia therapy.


Assuntos
Ácidos Araquidônicos/uso terapêutico , Variação Genética/fisiologia , Proteínas da Membrana Plasmática de Transporte de Glicina/genética , Glicina/análogos & derivados , Hiperecplexia/genética , Mutação de Sentido Incorreto/fisiologia , Neurônios/fisiologia , Animais , Ácidos Araquidônicos/farmacologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Feminino , Variação Genética/efeitos dos fármacos , Glicina/farmacologia , Glicina/uso terapêutico , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Hiperecplexia/tratamento farmacológico , Hiperecplexia/metabolismo , Mutação de Sentido Incorreto/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Wistar
3.
Anal Biochem ; 586: 113433, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31521670

RESUMO

This study investigated the association of A419T (rs121909661) and T449I (rs28928870) with infertility among Iranian women and possible treatments by agonizing the mutated receptor. 151 women were genotyped at A419T and T449I sites. Homology modeling, pharmacophore modeling, virtual screening, docking and molecular dynamics (MD) were performed. A419T and T449I indicated a significant and a weak association with infertility among Iranian women (P = 0.005 and P = 0.03, respectively). Significant differences found among three genotypes of A419T with FSH (P = 0.01) and LH (P < 0.0001). G-allele carriers of A419T had susceptibility to display higher FSH and LH serum levels. In silico results revealed the most potent agonists among 3041 similar compounds and MD supported this finding. Altogether, genotyping of A419T and T449I as potential markers might be helpful in prognosis and treatment of infertility. Also, a new series of potent FSHR agonists were identified for future drug development and treatment of infertility related to FSHR dysfunction.


Assuntos
Infertilidade Feminina/tratamento farmacológico , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto/efeitos dos fármacos , Receptores do FSH/agonistas , Receptores do FSH/antagonistas & inibidores , Adulto , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Infertilidade Feminina/genética , Irã (Geográfico) , Ligantes , Masculino , Estrutura Molecular , Gravidez , Receptores do FSH/genética , Adulto Jovem
4.
Arch Virol ; 163(10): 2757-2764, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29961118

RESUMO

Hepatitis C (HCV)-infected patients are treated with direct-acting antiviral agents (DAAs) in highly effective, well-tolerated, all-oral regimens. However, naturally occurring resistance-associated amino acid substitutions (RASs) may be selected during treatment. This study aimed to screen naturally occurring RASs NS3/NS4A inhibitors (PIs). Samples were obtained from DAA naïve patients, living in São Paulo state, Brazil. Screening for RASs in the HCV NS3 region was conducted in 859 samples from HCV-infected patients, of which 425 and 434 samples were subtype 1a and 1b, respectively. HCV-RNA was extracted, amplified, and sequenced. The overall prevalence of RASs to HCV PIs was 9.4%. The following RASs were observed in HCV-1a subtype infected patients: V36L (2.6%), T54S (1.6%), V55I/A (1.2% / 8.9%, respectively), Q80K (2.1%), R155K (0.5%), and D168E (0.2%); and in HCV-1b infected patients: V36L (0.7%), T54A/S (0.2% and 0.5%, respectively), V55A (0.5%), Q80K (0.2%), D168E (1.6%), and M175L (0.5%). HCV 1a infected subjects had higher serum viral load than that seen in patients infected with HCV 1b. There was no difference between the proportions of NS3 RASs with regards to geographic distribution within the investigated areas. These findings should be supported by additional studies in Brazil to help in the formation of local clinical guidelines for managing hepatitis C.


Assuntos
Antivirais/administração & dosagem , Proteínas de Transporte/antagonistas & inibidores , Farmacorresistência Viral , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepatite C/tratamento farmacológico , Inibidores de Proteases/administração & dosagem , Proteínas não Estruturais Virais/antagonistas & inibidores , Adulto , Substituição de Aminoácidos , Brasil/epidemiologia , Proteínas de Transporte/metabolismo , Feminino , Genótipo , Hepacivirus/isolamento & purificação , Hepacivirus/metabolismo , Hepatite C/epidemiologia , Hepatite C/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto/efeitos dos fármacos , Prevalência , Proteínas não Estruturais Virais/metabolismo , Adulto Jovem
5.
Neoplasia ; 20(5): 467-477, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29605720

RESUMO

BRAF is the most frequently mutated gene in melanoma. Constitutive activation of mutant BRAFV600E leads to aberrant Ras-independent MAPK signaling and cell transformation. Inhibition of mutant BRAF is a current frontline therapy for such cases, with improved survival compared with chemotherapy. Unfortunately, reactivation of MAPK signaling by several mechanisms has been shown to cause drug resistance and disease recurrence. In this work, we describe the co-occurrence of an in-frame deletion within an amplified BRAFV600E locus and a missense point mutation of the transcriptional repressor BCORL1 in vemurafenib-resistant A375 melanoma cells. Functional data confirmed that truncated p47BRAFV600E and mutant BCORL1Q1076H both contribute to resistance. Interestingly, either endogenous BCORL1 silencing or ectopic BCORL1Q1076H expression mimicked the effects of a CRISPR/Cas9-edited BCORL1Q1076H locus, suggesting a complex mixture of loss- and gain-of-function effects caused by the mutation. Transcriptomic data confirmed this hypothesis. Finally, we show that the pan-RAF inhibitor sorafenib is not affected by expression of BRAF deletion variant and effectively synergizes with vemurafenib to block resistant cells, suggesting a possible intervention for this class of mutants.


Assuntos
Melanoma/tratamento farmacológico , Melanoma/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Repressoras/genética , Vemurafenib/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/efeitos dos fármacos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Mutação de Sentido Incorreto/efeitos dos fármacos , Mutação de Sentido Incorreto/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Inibidores de Proteínas Quinases/farmacologia
6.
Hum Mol Genet ; 27(6): 943-953, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29325094

RESUMO

Adenosine triphosphate (ATP)-binding cassette subfamily A member 3 (ABCA3), a phospholipid transporter in lung lamellar bodies (LBs), is essential for the assembly of pulmonary surfactant and LB biogenesis. Mutations in the ABCA3 gene are an important genetic cause for respiratory distress syndrome in neonates and interstitial lung disease in children and adults, for which there is currently no cure. The aim of this study was to prove that disease causing misfolding ABCA3 mutations can be corrected in vitro and to investigate available options for correction. We stably expressed hemagglutinin (HA)-tagged wild-type ABCA3 or variants p.Q215K, p.M760R, p.A1046E, p.K1388N or p.G1421R in A549 cells and assessed correction by quantitation of ABCA3 processing products, their intracellular localization, resembling LB morphological integrity and analysis of functional transport activity. We showed that all mutant proteins except for M760R ABCA3 were rescued by the bithiazole correctors C13 and C17. These variants were also corrected by the chemical chaperone trimethylamine N-oxide and by low temperature. The identification of lead molecules C13 and C17 is an important step toward pharmacotherapy of ABCA3 misfolding-induced lung disease.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Doenças Pulmonares Intersticiais/tratamento farmacológico , Doenças Pulmonares Intersticiais/genética , Metilaminas/farmacologia , Mutação de Sentido Incorreto/efeitos dos fármacos , Deficiências na Proteostase/tratamento farmacológico , Deficiências na Proteostase/genética , Células A549 , Transportadores de Cassetes de Ligação de ATP/metabolismo , Humanos , Doenças Pulmonares Intersticiais/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Estudo de Prova de Conceito , Dobramento de Proteína , Deficiências na Proteostase/metabolismo
7.
J Biol Chem ; 293(7): 2247-2259, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29275329

RESUMO

Nucleoside reverse transcriptase (RT) inhibitors (NRTIs) are the backbone of current antiretroviral treatments. However, the emergence of viral resistance against NRTIs is a major threat to their therapeutic effectiveness. In HIV-1, NRTI resistance-associated mutations either reduce RT-mediated incorporation of NRTI triphosphates (discrimination mechanism) or confer an ATP-mediated nucleotide excision activity that removes the inhibitor from the 3' terminus of DNA primers, enabling further primer elongation (excision mechanism). In HIV-2, resistance to zidovudine (3'-azido-3'-deoxythymidine (AZT)) and other NRTIs is conferred by mutations affecting nucleotide discrimination. Mutations of the excision pathway such as M41L, D67N, K70R, or S215Y (known as thymidine-analogue resistance mutations (TAMs)) are rare in the virus from HIV-2-infected individuals. Here, we demonstrate that mutant M41L/D67N/K70R/S215Y HIV-2 RT lacks ATP-dependent excision activity, and recombinant virus containing this RT remains susceptible to AZT inhibition. Mutant HIV-2 RTs were tested for their ability to unblock and extend DNA primers terminated with AZT and other NRTIs, when complexed with RNA or DNA templates. Our results show that Met73 and, to a lesser extent, Ile75 suppress excision activity when TAMs are present in the HIV-2 RT. Interestingly, recombinant HIV-2 carrying a mutant D67N/K70R/M73K RT showed 10-fold decreased AZT susceptibility and increased rescue efficiency on AZT- or tenofovir-terminated primers, as compared with the double-mutant D67N/K70R. Molecular dynamics simulations reveal that Met73influences ß3-ß4 hairpin loop conformation, whereas its substitution affects hydrogen bond interactions at position 70, required for NRTI excision. Our work highlights critical HIV-2 RT residues impeding the development of excision-mediated NRTI resistance.


Assuntos
Farmacorresistência Viral , Infecções por HIV/virologia , Transcriptase Reversa do HIV/química , Transcriptase Reversa do HIV/metabolismo , HIV-2/enzimologia , Nucleosídeos/farmacologia , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Fármacos Anti-HIV/farmacologia , Reparo do DNA/efeitos dos fármacos , Transcriptase Reversa do HIV/genética , HIV-2/química , HIV-2/efeitos dos fármacos , HIV-2/genética , Humanos , Mutação de Sentido Incorreto/efeitos dos fármacos , Inibidores da Transcriptase Reversa/farmacologia
8.
J Antimicrob Chemother ; 72(11): 3043-3046, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28981647

RESUMO

OBJECTIVES: To assess the prevalence of cryptic silver (Ag+) resistance amongst clinical isolates of Gram-negative bacteria, and to examine how overt Ag+ resistance becomes activated in such strains. METHODS: Established methods were used to determine the susceptibility of 444 recent clinical isolates to Ag+, and to evaluate the potential for overt Ag+ resistance to emerge in susceptible isolates by spontaneous mutation. The genetic basis for Ag+ resistance was investigated using PCR amplification and DNA sequencing. RESULTS: None of the isolates tested displayed overt Ag+ resistance. However, upon silver challenge, high-level Ag+ resistance (silver nitrate MIC >128 mg/L) was selected at high frequency (10-7 to 10-8) in 76% of isolates of Enterobacter spp., ∼58% of isolates of Klebsiella spp. and ∼0.7% of isolates of Escherichia coli. All strains in which Ag+ resistance could be selected harboured the sil operon, with resistance apparently resulting from activation of this system as a consequence of single missense mutations in silS. By contrast, Ag+ resistance was not selected in isolates lacking sil, which included all tested representatives of Pseudomonas aeruginosa, Acinetobacter spp., Citrobacter spp. and Proteus spp. CONCLUSIONS: Whilst overt Ag+ resistance in Gram-negative pathogens is uncommon, cryptic Ag+ resistance pertaining to the sil operon is prevalent and readily activated in particular genera (Enterobacter and Klebsiella).


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/genética , Nitrato de Prata/farmacologia , Prata/farmacologia , Enterobacter/efeitos dos fármacos , Escherichia coli/genética , Bactérias Gram-Negativas/patogenicidade , Infecções por Bactérias Gram-Negativas/microbiologia , Humanos , Klebsiella/efeitos dos fármacos , Mutação de Sentido Incorreto/efeitos dos fármacos , Óperon , Prevalência
9.
Exp Anim ; 66(2): 137-144, 2017 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27928112

RESUMO

Camurati-Engelmann disease (CED) is a rare sclerosing bone disorder in humans with autosomal dominant inheritance. Mutations in the gene (TGFB1) that encodes transforming growth factor-ß1 (TGF-ß1) are causative for CED. TGF-ß1 signaling is enhanced by the CED-causing mutations. In this study, we performed Tgfb1 mutation screening in an ENU-mutagenized mouse genomic DNA library. We identified a missense mutation in which cysteine was substituted by serine at position 225 (p.C225S), that corresponded to the CED-causing mutation (p.C225R). TGF-ß1 mutant protein carrying p.C225S was secreted normally into the extracellular space. Reporter gene assays showed that the p.C225S mutants enhanced TGF-ß signaling at the same level as p.C225R mutants. We generated p.C225S homozygous mice and confirmed that the mature TGF-ß1 levels in the culture supernatants of the calvarial cells from the homozygotes were significantly higher than those from wild-type mice. Although the skull and femur are sclerotic in CED, these phenotypes were not observed in p.C225S homozygous mice. These results suggest that human and mouse bone tissue react differently to TGF-ß1. These findings are useful to pharmacological studies using mouse models in developing drugs that will target TGF-ß signaling.


Assuntos
Substituição de Aminoácidos/genética , Síndrome de Camurati-Engelmann/genética , Etilnitrosoureia/toxicidade , Estudos de Associação Genética , Mutação de Sentido Incorreto , Fator de Crescimento Transformador beta1/genética , Animais , Cisteína , Feminino , Biblioteca Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Terapia de Alvo Molecular , Mutação de Sentido Incorreto/efeitos dos fármacos , Fenótipo , Serina , Transdução de Sinais/genética
10.
Trends Mol Med ; 22(12): 997-999, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27793600

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2, encoding polycystin-1 and polycystin-2, respectively. Optimizing the folding environment for polycystin-1 missense mutations may have a critical effect on the progression of ADPKD in animal models and could potentially lead to tangible therapeutic options for subgroups of ADPKD patients.


Assuntos
Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Animais , Descoberta de Drogas , Humanos , Mutação de Sentido Incorreto/efeitos dos fármacos , Rim Policístico Autossômico Dominante/tratamento farmacológico , Dobramento de Proteína/efeitos dos fármacos , Canais de Cátion TRPP/química
11.
Nat Cell Biol ; 18(8): 897-909, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27347849

RESUMO

In cancer, the tumour suppressor gene TP53 undergoes frequent missense mutations that endow mutant p53 proteins with oncogenic properties. Until now, a universal mutant p53 gain-of-function program has not been defined. By means of multi-omics: proteome, DNA interactome (chromatin immunoprecipitation followed by sequencing) and transcriptome (RNA sequencing/microarray) analyses, we identified the proteasome machinery as a common target of p53 missense mutants. The mutant p53-proteasome axis globally affects protein homeostasis, inhibiting multiple tumour-suppressive pathways, including the anti-oncogenic KSRP-microRNA pathway. In cancer cells, p53 missense mutants cooperate with Nrf2 (NFE2L2) to activate proteasome gene transcription, resulting in resistance to the proteasome inhibitor carfilzomib. Combining the mutant p53-inactivating agent APR-246 (PRIMA-1MET) with the proteasome inhibitor carfilzomib is effective in overcoming chemoresistance in triple-negative breast cancer cells, creating a therapeutic opportunity for treatment of solid tumours and metastasis with mutant p53.


Assuntos
Proteínas Mutantes/efeitos dos fármacos , Mutação de Sentido Incorreto/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/farmacologia , Humanos , Camundongos , MicroRNAs/genética , Proteínas Mutantes/genética , Mutação de Sentido Incorreto/genética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/genética , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/genética , Proteoma/efeitos dos fármacos , Quinuclidinas/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Invest Ophthalmol Vis Sci ; 56(13): 7739-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26641551

RESUMO

PURPOSE: Protein misfolding, causing retention of nascent protein in the endoplasmic reticulum (ER), is the most common molecular phenotype for disease alleles of membrane proteins. Strategies are needed to identify therapeutics able to correct such folding/trafficking defects. Mutations of SLC4A11, a plasma membrane transport protein of the human corneal endothelial cell layer, cause cases of congenital hereditary endothelial dystrophy, Harboyan syndrome, and Fuchs' endothelial corneal dystrophy. Most SLC4A11 mutations induce SLC4A11 misfolding and retention in the ER. METHODS: An assay amenable to high-throughput screening was developed to quantify SLC4A11 at the plasma membrane, enabling a search for potential traffic-correcting small molecules. The assay was validated by comparing cell surface abundance of SLC4A11 mutants measured in the assay to observations from confocal immunofluorescence and values from cell surface biotinylation. Functionality of mutant proteins was assessed, using a confocal microscopic green fluorescent protein (GFP) water flux assay where relative rates of cell swelling are compared. RESULTS: A small-scale screen revealed that the nonsteroidal anti-inflammatory drugs (NSAIDs), glafenine, ibuprofen, and acetylsalicylic acid dissolved in 0.2% dimethyl sulfoxide (DMSO), partially rescued the trafficking defect in some SLC4A11 mutants, expressed in HEK293 cells. These SLC4A11 mutants retained functional activity when rescued to the plasma membrane by glafenine treatment. Glafenine was effective with an EC50 of 1.5 ± 0.7 µM. CONCLUSIONS: These data suggest that glafenine, and perhaps other NSAIDs, hold potential as therapeutics for misfolded membrane proteins, like SLC4A11. The high throughput approach described here can be modified to identify correctors of other misfolded plasma membrane proteins that cause eye disease.


Assuntos
Analgésicos não Narcóticos/farmacologia , Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Distrofias Hereditárias da Córnea/metabolismo , Glafenina/farmacologia , Mutação de Sentido Incorreto/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Proteínas de Transporte de Ânions/genética , Antiporters/genética , Linhagem Celular , Distrofias Hereditárias da Córnea/tratamento farmacológico , Distrofias Hereditárias da Córnea/genética , Células HEK293/efeitos dos fármacos , Células HEK293/metabolismo , Perda Auditiva Neurossensorial/metabolismo , Humanos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética
13.
Mycopathologia ; 179(3-4): 213-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25398256

RESUMO

The mechanism of fluconazole resistance in Candida tropicalis is still unclear. Recently, we isolated a fluconazole-resistant strain of C. tropicalis from the blood specimen of a patient with candidemia in China. In vitro antifungal susceptibility of the isolate was determined by using CLSI M27-A3 and E-test methods. The sequence of ERG11 gene was then analyzed, and the three-dimensional model of Erg11p encoded by ERG11 gene was also investigated. The sequencing of ERG11 gene revealed the mutation of A395T in this fluconazole-resistant isolate of C. tropicalis, resulting in the Y132F substitution in Erg11p. Sequence alignment and three-dimensional model comparison of Erg11ps showed high similarity between fluconazole-susceptible isolates of C. tropicalis and Candida albicans. The comparison of the three-dimensional models of Erg11ps demonstrated that the position of the Y132F substitution in this isolate of C. tropicalis is identical to the isolate of C. albicans with fluconazole resistance resulting from Y132F substitution in Erg11p. Hence, we ascertain that the Y132F substitution of Erg11p caused by A395T mutation in ERG11 gene confers the fluconazole resistance in C. tropicalis.


Assuntos
Antifúngicos/farmacologia , Candida tropicalis/efeitos dos fármacos , Candida tropicalis/genética , Candidemia/microbiologia , Candidíase/microbiologia , Sistema Enzimático do Citocromo P-450/genética , Fluconazol/farmacologia , Proteínas Fúngicas/genética , Mutação de Sentido Incorreto , Candida tropicalis/enzimologia , Candidemia/tratamento farmacológico , Candidíase/tratamento farmacológico , Sistema Enzimático do Citocromo P-450/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Mutação de Sentido Incorreto/efeitos dos fármacos
14.
J Cyst Fibros ; 14(1): 34-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25042876

RESUMO

BACKGROUND: Cystic fibrosis (CF) is rare in India. Most CF mutations identified are not yet functionally characterized. Hence, genetic counseling and adoption of therapeutic approach are particularly difficult. Our aim was to study the function and maturation of a spectrum of eleven Indian CFTR mutations from classical CF and infertile male patients with CBAVD. METHODS: We used Western blot, pharmacology and iodide efflux to study CFTR maturation and chloride transport in BHK cells expressing pEGFP-CFTR constructs for L69H, F87I, S118P, G126S, H139Q, F157C, F494L, E543A, S549N, Y852F and D1270E. RESULTS: Among these CFTR mutants, only L69H is not processed as a c-band and not functional at 37°C. However, the functions of L69H and S549N and the maturation of L69H are corrected at 27°C and by the investigational drug VX809. CONCLUSION: These data should help in developing counseling and therapeutic approaches in India. We identified L69H as a novel class II CF mutation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Regulação da Expressão Gênica , Doenças Urogenitais Masculinas/genética , Mutação de Sentido Incorreto , Ducto Deferente/anormalidades , Aminopiridinas/farmacologia , Benzodioxóis/farmacologia , Células Cultivadas/efeitos dos fármacos , Fibrose Cística/epidemiologia , Regulador de Condutância Transmembrana em Fibrose Cística/farmacologia , Humanos , Índia , Infertilidade Masculina/genética , Masculino , Mutação de Sentido Incorreto/efeitos dos fármacos , Doenças Raras , Estudos de Amostragem , Maturidade Sexual/genética
15.
J Med Case Rep ; 8: 403, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25471118

RESUMO

INTRODUCTION: Familial amyloid polyneuropathy is a rare autosomal dominant disorder caused by mutations in the transthyretin gene, TTR. Diagnosis can be challenging, especially if other family members are not affected or an obvious systemic involvement is lacking. The patients are often misdiagnosed, leading to a delay in the initiation of therapy. CASE PRESENTATION: A 35-year-old woman of Turkish origin presented to our outpatient clinic with severe polyneuropathy associated with distally pronounced tetraparesis and hypesthesia of 2 to 3 years' duration. In addition, small nerve fiber involvement with impaired detection of cold temperatures and tingling pain in the lower legs was reported. She did not complain of autonomic dysfunction or visual disturbance. Her family history was empty regarding neuromuscular disorders. The routine diagnostic work-up was unremarkable. A sural nerve biopsy disclosed amyloid deposits, which led to the identification of a rare heterozygous transthyretin mutation (p.Glu74Gly; old classification: p.Glu54Gly). CONCLUSIONS: Few cases with this very heterozygous mutation can be found in the literature. In contrast to the case of our patient, all of the previously described patients in the literature presented with additional severe autonomic symptoms, involvement of the eyes and a positive family history. In this case report, we emphasize that, in patients with progressive neuropathy with small fiber involvement, an amyloid neuropathy should be considered in the differential diagnosis, even if the family history is empty and other organs are not affected.


Assuntos
Neuropatias Amiloides Familiares/diagnóstico , Neuropatias Amiloides Familiares/genética , Benzoxazóis/uso terapêutico , Transplante de Fígado/efeitos adversos , Mutação de Sentido Incorreto , Pré-Albumina/genética , Adulto , Neuropatias Amiloides Familiares/patologia , Progressão da Doença , Evolução Fatal , Feminino , Ligação Genética , Ácido Glutâmico , Glicina , Humanos , Mutação de Sentido Incorreto/efeitos dos fármacos , Linhagem , Pré-Albumina/efeitos dos fármacos
16.
J Antimicrob Chemother ; 69(7): 1933-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24695353

RESUMO

OBJECTIVES: The reduction in mother-to-child transmission of HIV-1 by single-dose nevirapine given at birth onset is achieved at the expense of de novo HIV-1 resistance mutations. In the VITA1 study, single-dose carbamazepine accelerated nevirapine elimination, but the accompanying trend towards fewer de novo HIV-1 mutations was statistically non-significant. METHODS: We investigated if the effect of carbamazepine was confounded by the individual variability in nevirapine metabolism and transport. RESULTS: Nine of 34 (26%) single-dose nevirapine-treated women had one or more nevirapine-associated resistance mutations, compared with 3 of 34 (9%) in the single-dose nevirapine/carbamazepine arm. The genetic polymorphisms in CYP2B6 and MRP7 affected neither nevirapine kinetics nor the development of HIV-1 resistance. In contrast, the reduction in HIV-1 mutations by single-dose carbamazepine reached statistical significance at P = 0.04 with an OR of 0.1 (95% CI 0.01-0.90) upon consideration of CYP3A activity, defined as the ratio of 4ß-hydroxycholesterol to cholesterol, and it was more likely in women with higher CYP3A activity. These findings were in agreement with CYP3A induction in carbamazepine-treated patients. Likewise, carbamazepine induced CYP3A4, but not CYP2B6, in vitro when combined with nevirapine. CONCLUSIONS: The induction of nevirapine elimination reduces HIV-1 resistance mutations, but this effect is modulated by individual CYP3A activity. The study suggests that CYP3A4 activity could be monitored using an endogenous marker and, if needed, boosted to improve clinical endpoints.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Carbamazepina/administração & dosagem , Indutores do Citocromo P-450 CYP3A/administração & dosagem , Farmacorresistência Viral , HIV-1/efeitos dos fármacos , Mutação de Sentido Incorreto/efeitos dos fármacos , Nevirapina/administração & dosagem , Fármacos Anti-HIV/farmacologia , Carbamazepina/metabolismo , Quimioprevenção/métodos , Citocromo P-450 CYP3A/metabolismo , Indutores do Citocromo P-450 CYP3A/metabolismo , Feminino , Infecções por HIV/prevenção & controle , HIV-1/genética , Humanos , Nevirapina/farmacologia , Gravidez , Resultado do Tratamento
17.
Genet Mol Res ; 12(3): 2829-36, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24065639

RESUMO

This study aimed to investigate the properties of mutations of the active efflux pump genes acrA/B and tolC in Escherichia coli CVCC 1547 when induced by different drugs. The mutations were isolated in vitro by exposing E. coli CVCC 1547 to stepwise increases in the concentration of ceftriaxone (CRO), amikacin (AMK), or ciprofloxacin. The results showed that the minimum inhibitory concentrations for the corresponding drugs increased, as did the minimum inhibitory concentrations for other fluoroquinolones and ß-lactam drugs that were not inducers. DNA sequence analyses of the acrA/B and tolC genes of the mutants and comparison with the parent strain revealed that genetic variations had occurred. Three point mutations resulted in amino acid changes in the proteins expressed. Specifically, strain CRO10 had a mutation in acrA, A309G, that resulted in a Thr-103 to Ala substitution, and a mutation in tolC, G735A, that changed Ala-245 to Thr; strain AMK20 (and AMK30) had a Val-447 to Ile amino acid change in acrB. In addition to the missense mutations in these strains, we detected 7, 20, and 15 nonsense mutations in acrA, acrB, and tolC, respectively. To sum up, multiple genetic sequence variations and some changes in amino acid sequences were detected when E. coli CVCC 1547 was challenged in vitro with CRO, AMK, or ciprofloxacin. These changes may have given rise to multidrug-resistant strains.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Farmacorresistência Bacteriana Múltipla/genética , Proteínas de Escherichia coli/genética , Lipoproteínas/genética , Proteínas de Membrana Transportadoras/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Amicacina/farmacologia , Ceftriaxona/farmacologia , Ciprofloxacina/farmacologia , Códon sem Sentido/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica , Mutação de Sentido Incorreto/efeitos dos fármacos , Análise de Sequência de DNA
18.
J Neurosci ; 33(16): 6834-44, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23595742

RESUMO

The gene encoding the WD repeat-containing protein 81 (WDR81) has recently been described as the disease locus in a consanguineous family that suffers from cerebellar ataxia, mental retardation, and quadrupedal locomotion syndrome (CAMRQ2). Adult mice from the N-ethyl-N-nitrosourea-induced mutant mouse line nur5 display tremor and an abnormal gait, as well as Purkinje cell degeneration and photoreceptor cell loss. We have used polymorphic marker mapping to demonstrate that affected nur5 mice carry a missense mutation, L1349P, in the Wdr81 gene. Moreover, homozygous nur5 mice that carry a wild-type Wdr81 transgene are rescued from the abnormal phenotype, indicating that Wdr81 is the causative gene in nur5. WDR81 is expressed in Purkinje cells and photoreceptor cells, among other CNS neurons, and like the human mutation, the nur5 modification lies in the predicted major facilitator superfamily domain of the WDR81 protein. Electron microscopy analysis revealed that a subset of mitochondria in Purkinje cell dendrites of the mutant animals displayed an aberrant, large spheroid-like structure. Moreover, immunoelectron microscopy and analysis of mitochondrial-enriched cerebellum fractions indicate that WDR81 is localized in mitochondria of Purkinje cell neurons. Because the nur5 mouse mutant demonstrates phenotypic similarities to the human disease, it provides a valuable genetic model for elucidating the pathogenic mechanism of the WDR81 mutation in CAMRQ2.


Assuntos
Apraxia da Marcha/genética , Apraxia da Marcha/patologia , Proteínas Nucleares/metabolismo , Células Fotorreceptoras/metabolismo , Células de Purkinje/metabolismo , Actinas/metabolismo , Alquilantes/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Calbindinas , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cerebelo/patologia , Mapeamento Cromossômico , Modelos Animais de Doenças , Etilnitrosoureia/farmacologia , Lateralidade Funcional , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Mutagênese/efeitos dos fármacos , Mutação de Sentido Incorreto/efeitos dos fármacos , Mutação de Sentido Incorreto/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Células Fotorreceptoras/efeitos dos fármacos , Células Fotorreceptoras/ultraestrutura , Prostaglandina-Endoperóxido Sintases/metabolismo , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/ultraestrutura , RNA Mensageiro/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Análise de Sequência de DNA , Transfecção
19.
Viruses ; 5(3): 981-97, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23524389

RESUMO

DEB025 (alisporivir) is a synthetic cyclosporine with inhibitory activity against human immunodeficiency virus type-1 (HIV-1) and hepatitis C virus (HCV). It binds to cyclophilin A (CypA) and blocks essential functions of CypA in the viral replication cycles of both viruses. DEB025 inhibits clinical HIV-1 isolates in vitro and decreases HIV-1 virus load in the majority of patients. HIV-1 isolates being naturally resistant to DEB025 have been detected in vitro and in nonresponder patients. By sequence analysis of their capsid protein (CA) region, two amino acid polymorphisms that correlated with DEB025 resistance were identified: H87Q and I91N, both located in the CypA-binding loop of the CA protein of HIV-1. The H87Q change was by far more abundant than I91N. Additional polymorphisms in the CypA-binding loop (positions 86, 91 and 96), as well as in the N-terminal loop of CA were detected in resistant isolates and are assumed to contribute to the degree of resistance. These amino acid changes may modulate the conformation of the CypA-binding loop of CA in such a way that binding and/or isomerase function of CypA are no longer necessary for virus replication. The resistant HIV-1 isolates thus are CypA-independent.


Assuntos
Fármacos Anti-HIV/farmacologia , Proteínas do Capsídeo/genética , Farmacorresistência Viral , Infecções por HIV/virologia , HIV-1/genética , Polimorfismo Genético , Sequência de Aminoácidos , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Ciclosporina , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , HIV-1/metabolismo , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto/efeitos dos fármacos
20.
J Neurosci ; 33(10): 4395-404, 2013 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-23467356

RESUMO

In hair cells of the inner ear, sound or head movement increases tension in fine filaments termed tip links, which in turn convey force to mechanosensitive ion channels to open them. Tip links are formed by a tetramer of two cadherin proteins: protocadherin 15 (PCDH15) and cadherin 23 (CDH23), which have 11 and 27 extracellular cadherin (EC) repeats, respectively. Mutations in either protein cause inner ear disorders in mice and humans. We showed recently that these two cadherins bind tip-to-tip in a "handshake" mode that involves the EC1 and EC2 repeats of both proteins. However, a paucity of appropriate animal models has slowed our understanding both of the interaction and of how mutations of residues within the predicted interface compromise tip link integrity. Here, we present noddy, a new mouse model for hereditary deafness. Identified in a forward genetic screen, noddy homozygotes lack inner ear function. Mapping and sequencing showed that noddy mutant mice harbor an isoleucine-to-asparagine (I108N) mutation in the EC1 repeat of PCDH15. Residue I108 interacts with CDH23 EC2 in the handshake and its mutation impairs the interaction in vitro. The noddy mutation allowed us to determine the consequences of blocking the handshake in vivo: tip link formation and bundle morphology are disrupted, and mechanotransduction channels fail to remain open at rest. These results offer new insights into the interaction between PCDH15 and CDH23 and help explain the etiology of human deafness linked to mutations in the tip-link interface.


Assuntos
Caderinas/genética , Caderinas/metabolismo , Células Ciliadas Auditivas/metabolismo , Doenças do Labirinto , Mecanotransdução Celular/fisiologia , Mutação de Sentido Incorreto/genética , Precursores de Proteínas/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Proteínas Relacionadas a Caderinas , Cálcio/metabolismo , Células Cultivadas , Eletroencefalografia , Etilnitrosoureia/farmacologia , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Genótipo , Células Ciliadas Auditivas/patologia , Células Ciliadas Auditivas/ultraestrutura , Perda Auditiva/induzido quimicamente , Perda Auditiva/genética , Doenças do Labirinto/induzido quimicamente , Doenças do Labirinto/genética , Doenças do Labirinto/patologia , Doenças do Labirinto/fisiopatologia , Camundongos , Camundongos Transgênicos , Microscopia de Força Atômica , Mutagênicos/farmacologia , Mutação de Sentido Incorreto/efeitos dos fármacos , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Compostos de Piridínio , Compostos de Amônio Quaternário
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA