Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Parkinsons Dis ; 14(2): 325-334, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38251063

RESUMO

Background: Pre-clinical studies suggest that c-Abl activation may play an important role in the etiology of Parkinson's disease, making c-Abl an important target to evaluate for potential disease-modification. Objective: To assess safety, tolerability, and pharmacokinetics of the c-Abl inhibitor risvodetinib (IkT-148009) in healthy subjects and participants with Parkinson's disease. Methods: Part 1 (single ascending dose (SAD)) and Part 2 (7-day multiple ascending dose (MAD)) studies were in healthy volunteers. Participants were randomized 3 : 1 across 9 SAD doses and 3 MAD doses of risvodetinib (IkT-148009) or placebo. Part 3 was a MAD study conducted at two doses in 14 participants with mild-to-moderate PD (MAD-PD). Primary outcome measures were safety, tolerability and pharmacokinetics. Exploratory outcomes in PD participants included clinical measures of PD state, GI function, and cerebrospinal fluid (CSF) concentration. Results: 108 patients were treated with no dropouts. The SAD tested doses ranging from 12.5 to 325 mg, while the MAD tested 25 to 200 mg and MAD-PD tested 50 to 100 mg in Parkinson's participants. All active doses had a favorable safety profile with no clinically meaningful adverse events. Single dose pharmacokinetics were approximately linear between 12.5 mg and 200 mg for both Cmax and AUC0 - inf without distinction between healthy volunteers and participants with PD. Exposures at each dose were high relative to other drugs in the same kinase inhibitor class. Conclusions: Risvodetinib (IkT-148009) was well tolerated, had a favorable safety and pharmacology profile over 7-day dosing, did not induce serious adverse events and did not appear to induce deleterious side-effects in participants administered anti-PD medications.


Assuntos
Doença de Parkinson , Idoso , Humanos , Área Sob a Curva , Voluntários Saudáveis , Doença de Parkinson/tratamento farmacológico
2.
Sci Transl Med ; 15(679): eabp9352, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36652533

RESUMO

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease of the central nervous system, with an estimated 5,000,000 cases worldwide. PD pathology is characterized by the accumulation of misfolded α-synuclein, which is thought to play a critical role in the pathogenesis of the disease. Animal models of PD suggest that activation of Abelson tyrosine kinase (c-Abl) plays an essential role in the initiation and progression of α-synuclein pathology and initiates processes leading to degeneration of dopaminergic and nondopaminergic neurons. Given the potential role of c-Abl in PD, a c-Abl inhibitor library was developed to identify orally bioavailable c-Abl inhibitors capable of crossing the blood-brain barrier based on predefined characteristics, leading to the discovery of IkT-148009. IkT-148009, a brain-penetrant c-Abl inhibitor with a favorable toxicology profile, was analyzed for therapeutic potential in animal models of slowly progressive, α-synuclein-dependent PD. In mouse models of both inherited and sporadic PD, IkT-148009 suppressed c-Abl activation to baseline and substantially protected dopaminergic neurons from degeneration when administered therapeutically by once daily oral gavage beginning 4 weeks after disease initiation. Recovery of motor function in PD mice occurred within 8 weeks of initiating treatment concomitantly with a reduction in α-synuclein pathology in the mouse brain. These findings suggest that IkT-148009 may have potential as a disease-modifying therapy in PD.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Sinucleinopatias , Camundongos , Animais , alfa-Sinucleína/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Doenças Neurodegenerativas/patologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo
3.
Mov Disord ; 37(1): 6-15, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34816484

RESUMO

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease of the central nervous system, with an estimated 5 000 000 cases worldwide. Historically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, PD pathology is now known to be widespread and to affect serotonin, cholinergic and norepinephrine neurons as well as nerve cells in the olfactory system, cerebral hemisphere, brain stem, spinal cord, and peripheral autonomic nervous system. PD pathology is characterized by the accumulation of misfolded α-synuclein, which is thought to play a critical role in the etiopathogenesis of the disease. Animal models of PD suggest that activation of the Abelson tyrosine kinase (c-Abl) plays an essential role in the initiation and progression of α-synuclein pathology and neurodegeneration. These studies demonstrate that internalization of misfolded α-synuclein activates c-Abl, which phosphorylates α-synuclein and promotes α-synuclein pathology within the affected neurons. Additionally, c-Abl inactivates parkin, disrupting mitochondrial quality control and biogenesis, promoting neurodegeneration. Post-mortem studies of PD patients demonstrate increased levels of tyrosine phosphorylated α-synuclein, consistent with the activation of c-Abl in human disease. Although the c-Abl inhibitor nilotinib failed to demonstrate clinical benefit in two double-blind trials, novel c-Abl inhibitors have been developed that accumulate in the brain and may inhibit c-Abl at saturating levels. These novel inhibitors have demonstrated benefits in animal models of PD and have now entered clinical development. Here, we review the role of c-Abl in the neurodegenerative disease process and consider the translational potential of c-Abl inhibitors from model studies to disease-modifying therapies for Parkinson's disease. © 2021 Inhibikase Therapeutics, Inc. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Animais , Encéfalo/patologia , Neurônios Dopaminérgicos/metabolismo , Humanos , Doenças Neurodegenerativas/patologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto
4.
Parkinsonism Relat Disord ; 90: 134-141, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34561166

RESUMO

OBJECTIVE: Disease modification in Parkinson disease (PD) has remained an elusive goal, in spite of large investments over several decades. Following a large meeting of experts, this review article discusses the state of the science, possible reasons for past PD trials' failures to demonstrate disease-modifying benefit, and potential solutions. METHODS: The National Institute of Neurological Disorders and Stroke (NINDS) convened a meeting including leaders in the field and representatives of key stakeholder groups to discuss drug therapy with the goal of disease modification in PD. RESULTS: Important lessons can be learned from previous attempts, as well as from other fields. The selection process for therapeutic targets and agents differs among various organizations committed to therapeutic development. The areas identified as critical to target in future research include the development of relevant biomarkers, refinements of the targeted patient populations, considerations of novel trial designs, and improving collaborations between all stakeholders. CONCLUSIONS: We identify potential barriers to progress in disease modification for Parkinson's and propose a set of research priorities that may improve the likelihood of success.


Assuntos
Descoberta de Drogas , Doença de Parkinson/tratamento farmacológico , Biomarcadores/análise , Humanos , National Institute of Neurological Disorders and Stroke (USA) , Estados Unidos
5.
Neurobiol Dis ; 148: 105184, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33221532

RESUMO

Multiple system atrophy (MSA) is a rare and extremely debilitating progressive neurodegenerative disease characterized by variable combinations of parkinsonism, cerebellar ataxia, dysautonomia, and pyramidal dysfunction. MSA is a unique synucleinopathy, in which alpha synuclein-rich aggregates are present in the cytoplasm of oligodendroglia. The precise origin of the alpha synuclein (aSyn) found in the glial cytoplasmic inclusions (GCIs) as well the mechanisms of neurodegeneration in MSA remain unclear. Despite this fact, cell and animal models of MSA rely on oligodendroglial overexpression of aSyn. In the present study, we utilized a novel oligotrophic AAV, Olig001, to overexpress aSyn specifically in striatal oligodendrocytes of rats and nonhuman primates in an effort to further characterize our novel viral vector-mediated MSA animal models. Using two cohorts of animals with 10-fold differences in Olig001 vector titers, we show a dose-dependent formation of MSA-like pathology in rats. High titer of Olig001-aSyn in these animals were required to produce the formation of pS129+ and proteinase K resistant aSyn-rich GCIs, demyelination, and neurodegeneration. Using this knowledge, we injected high titer Olig001 in the putamen of cynomolgus macaques. After six months, histological analysis showed that oligodendroglial overexpression of aSyn resulted in the formation of hallmark GCIs throughout the putamen, demyelination, a 44% reduction of striatal neurons and a 12% loss of nigral neurons. Furthermore, a robust inflammatory response similar to MSA was produced in Olig001-aSyn NHPs, including microglial activation, astrogliosis, and a robust infiltration of T cells into the CNS. Taken together, oligodendroglial-specific viral vector-mediated overexpression of aSyn in rats and nonhuman primates faithfully reproduces many of the pathological disease hallmarks found in MSA. Future studies utilizing these large animal models of MSA would prove extremely valuable as a pre-clinical platform to test novel therapeutics that are so desperately needed for MSA.


Assuntos
Modelos Animais de Doenças , Atrofia de Múltiplos Sistemas/genética , Neostriado/patologia , Neurônios/patologia , Oligodendroglia/patologia , Putamen/patologia , alfa-Sinucleína/genética , Animais , Dependovirus , Vetores Genéticos , Humanos , Macaca fascicularis , Atrofia de Múltiplos Sistemas/metabolismo , Atrofia de Múltiplos Sistemas/patologia , Oligodendroglia/metabolismo , Ratos , Técnicas Estereotáxicas , alfa-Sinucleína/metabolismo
6.
BMC Pharmacol Toxicol ; 19(1): 80, 2018 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-30514402

RESUMO

BACKGROUND: Several tyrosine kinase inhibitors (TKIs) developed as anti-cancer drugs, also have anti-viral activity due to their ability to disrupt productive replication and dissemination in infected cells. Consequently, such drugs are attractive candidates for "repurposing" as anti-viral agents. However, clinical evaluation of therapeutics against infectious agents associated with high mortality, but low or infrequent incidence, is often unfeasible. The United States Food and Drug Administration formulated the "Animal Rule" to facilitate use of validated animal models for conducting anti-viral efficacy studies. METHODS: To enable such efficacy studies of two clinically approved TKIs, nilotinib, and imatinib, we first conducted comprehensive pharmacokinetic (PK) studies in relevant rodent and non-rodent animal models. PK of these agents following intravenous and oral dosing were evaluated in C57BL/6 mice, prairie dogs, guinea pigs and Cynomolgus monkeys. Plasma samples were analyzed using an LC-MS/MS method. Secondarily, we evaluated the utility of allometry-based inter-species scaling derived from previously published data to predict the PK parameters, systemic clearance (CL) and the steady state volume of distribution (Vss) of these two drugs in prairie dogs, an animal model not tested thus far. RESULTS: Marked inter-species variability in PK parameters and resulting oral bioavailability was observed. In general, elimination half-lives of these agents in mice and guinea pigs were much shorter (1-3 h) relative to those in larger species such as prairie dogs and monkeys. The longer nilotinib elimination half-life in prairie dogs (i.v., 6.5 h and oral, 7.5 h), facilitated multiple dosing PK and safety assessment. The allometry-based predicted values of the Vss and CL were within 2.0 and 2.5-fold, respectively, of the observed values. CONCLUSIONS: Our results suggest that prairie dogs and monkeys may be suitable rodent and non-rodent species to perform further efficacy testing of these TKIs against orthopoxvirus infections. The use of rodent models such as C57BL/6 mice and guinea pigs for assessing pre-clinical anti-viral efficacy of these two TKIs may be limited due to short elimination and/or low oral bioavailability. Allometry-based correlations, derived from existing literature data, may provide initial estimates, which may serve as a useful guide for pre-clinical PK studies in untested animal models.


Assuntos
Antineoplásicos/farmacocinética , Antivirais/farmacocinética , Mesilato de Imatinib/farmacocinética , Proteínas Tirosina Quinases/farmacocinética , Pirimidinas/farmacocinética , Administração Intravenosa , Administração Oral , Animais , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Feminino , Cobaias , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Sciuridae
7.
J Neurovirol ; 22(6): 871-875, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27198748

RESUMO

Sixty-three natalizumab-treated patients with relapsing multiple sclerosis were screened for JC polyomavirus (JCV) viruria. Urinary-positive patients were longitudinally sampled for up to 24 weeks. Using methods that distinguish encapsidated virus from naked viral DNA, 17.5 % of patients were found to excrete virus, consistent with the prevalence of urinary excretion in the general population. Unexpectedly, urinary excretion was predominantly seen (>73 %) in patients with high JC antibody index (≥2.0). Active JCV infection, therefore, tends to occur in natalizumab patients that carry a high risk factor for the development of disease, directly linking JC infection to the risk factors for PML development.


Assuntos
DNA Viral/urina , Fatores Imunológicos/uso terapêutico , Vírus JC/patogenicidade , Leucoencefalopatia Multifocal Progressiva/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Natalizumab/uso terapêutico , Anticorpos Antivirais/urina , Humanos , Vírus JC/imunologia , Leucoencefalopatia Multifocal Progressiva/etiologia , Leucoencefalopatia Multifocal Progressiva/urina , Leucoencefalopatia Multifocal Progressiva/virologia , Estudos Longitudinais , Esclerose Múltipla Recidivante-Remitente/complicações , Esclerose Múltipla Recidivante-Remitente/urina , Esclerose Múltipla Recidivante-Remitente/virologia , Fatores de Risco , Urinálise
8.
Pharm Dev Technol ; 20(2): 169-75, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24218989

RESUMO

The main objective of the present study was to develop formulations of noscapine hydrochloride hydrate with enhanced solubility and bioavailability using co-solvent- and cyclodextrin-based approaches. Different combinations of co-solvents, which were selected on the basis of high-throughput solubility screening, were subjected to in vitro intestinal drug permeability studies conducted with Ussing chambers. Vitamin E tocopherol polyethylene glycol succinate and propylene glycol based co-solvent formulations provided the maximum permeability coefficient for the drug. Inclusion complexes of the drug were prepared using hydroxypropyl-ß-cyclodextrin and sulphobutylether cyclodextrins. Pharmacokinetic studies were carried out in male Sprague-Dawley rats for the selected formulations. The relative bioavailabilities of the drug with the co-solvent- and cyclodextrin-based formulations were found to be similar.


Assuntos
Antivirais/administração & dosagem , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Excipientes/química , Noscapina/administração & dosagem , Solventes/química , Administração Oral , Animais , Antivirais/sangue , Antivirais/química , Antivirais/farmacologia , Disponibilidade Biológica , Humanos , Influenza Humana/tratamento farmacológico , Absorção Intestinal , Masculino , Noscapina/sangue , Noscapina/química , Noscapina/farmacologia , Ratos Sprague-Dawley , Solubilidade
9.
Haematologica ; 93(11): 1728-33, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18790797

RESUMO

An inducible model for conditional expression of AML1-ETO in myeloid U-937 cells was generated previously to determine cellular effects of AML1-ETO and to identify target genes. Induction of AML1-ETO expression in U-937 resulted in reduced cell growth, G1 arrest and apoptosis. Microarray analysis showed more genes up-regulated than down-regulated (180 vs. 69). Clustering of AML1-ETO-positive and -negative cell lines was possible based on these differentially expressed genes. p21/WAF/Cip1 (CDKN1A) was up-regulated 4.6-fold upon induction of AML1-ETO which was confirmed in additional experiments. Knock-down of AML1-ETO by siRNA could reduce p21/WAF/Cip1 expression in Kasumi-1 cells. mRNA expression analysis of p21/WAF/Cip1 in a large cohort of acute myeloid leukemia patients demonstrated a significantly higher expression in AML1-ETO-positive leukemia. The increased expression of p21/WAF/Cip1 in primary leukemic blasts suggests that elevated p21/WAF/Cip1 levels may contribute to specific features observed in AML1-ETO positive leukemia.


Assuntos
Crise Blástica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteínas de Ligação a DNA/genética , Leucemia Mieloide/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Transcrição Gênica , Proteínas rho de Ligação ao GTP/genética , Crise Blástica/patologia , Northern Blotting , Mapeamento Cromossômico , Cromossomos Humanos Par 21 , Cromossomos Humanos Par 8 , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Genes p53 , Humanos , Leucemia Mieloide/patologia , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Proteína 1 Parceira de Translocação de RUNX1 , Translocação Genética , Regulação para Cima
10.
Mol Microbiol ; 69(2): 331-43, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18485078

RESUMO

Transcriptional activation often employs a direct interaction between an activator and RNA polymerase. For activation of its middle genes, bacteriophage T4 appropriates Escherichia coli RNA polymerase through the action of two phage-encoded proteins, MotA and AsiA. Alone, AsiA inhibits transcription from a large class of host promoters by structurally remodelling region 4 of sigma(70), the primary specificity subunit of E. coli RNA polymerase. MotA interacts both with sigma(70) region 4 and with a DNA element present in T4 middle promoters. AsiA-induced remodelling is proposed to make the far C-terminus of sigma(70) region 4 accessible for MotA binding. Here, NMR chemical shift analysis indicates that MotA uses a 'basic/hydrophobic' cleft to interact with the C-terminus of AsiA-remodelled sigma(70), but MotA does not interact with AsiA itself. Mutations within this cleft, at residues K3, K28 and Q76, both impair the interaction of MotA with sigma(70) region 4 and MotA-dependent activation. Furthermore, mutations at these residues greatly decrease phage viability. Most previously described activators that target sigma(70) directly use acidic residues to engage a basic surface of region 4. Our work supports accumulated evidence indicating that 'sigma appropriation' by MotA and AsiA uses a fundamentally different mechanism to activate transcription.


Assuntos
Bacteriófago T4/fisiologia , Proteínas de Ligação a DNA/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Escherichia coli/virologia , Mapeamento de Interação de Proteínas , Fator sigma/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Fusão Gênica Artificial , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Genes Reporter , Teste de Complementação Genética , Espectroscopia de Ressonância Magnética , Viabilidade Microbiana , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
11.
Nat Struct Mol Biol ; 14(7): 653-61, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17572682

RESUMO

The eight twenty-one protein, ETO, is implicated in 12%-15% of acute human leukemias as part of a gene fusion with RUNX1 (also called AML1). Of the four ETO domains related to Drosophila melanogaster Nervy, only two are required to induce spontaneous myeloid leukemia upon transplantation into the mouse. One of these domains is related in sequence to TAF4, a component of TFIID. The structure of this domain, ETO-TAFH, is similar to yeast Rpb4 and to Escherichia coli sigma(70); it is the first TAF-related protein with structural similarity to the multisubunit RNA polymerases. Overlapping surfaces of ETO-TAFH interact with an autonomous repression domain of the nuclear receptor corepressor N-CoR and with a conserved activation domain from the E-box family of transcription factors. Thus, ETO-TAFH acts as a structural platform that can interchange negative and positive coregulatory proteins to control transcription.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas Proto-Oncogênicas/química , Proteínas Repressoras/química , Fator de Transcrição TFIID/química , Fatores de Transcrição/química , Animais , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Elementos E-Box , Regulação da Expressão Gênica , Sequências Hélice-Alça-Hélice , Humanos , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteína 1 Parceira de Translocação de RUNX1 , Proteínas Repressoras/genética , Homologia de Sequência de Aminoácidos , Fator de Transcrição TFIID/genética , Fatores de Transcrição/genética , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido , Regulação para Cima
13.
Cell Cycle ; 5(20): 2332-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17102623

RESUMO

The Fas-associated death domain protein FADD is best known as an adaptor protein that senses a signal received at a death receptor and nucleates the assembly of the death-inducing signaling complex. Recent work reveals unexpected properties for this signaling protein, suggesting new roles for FADD in apoptotic signaling and in non-apoptotic functions linked to chemical modification of the FADD C-terminus. These new studies suggest novel types of high valency complexes may form in the plasma membrane and in the nucleus, raising intriguing questions as to how FADD senses the environment and responds to different signaling inputs to promote a biochemical response. In particular, we discuss the role of FADD in death receptor avidity and examine the relationship between FADD phosphorylation and subcellular localization with respect to various biological functions. Since FADD serves to modulate both apoptosis and cell cycle progression, these new findings promote the concept that differential complex assembly dictates disparate cellular processes mediated by this adaptor molecule.


Assuntos
Ciclo Celular , Proteína de Domínio de Morte Associada a Fas/fisiologia , Receptores de Morte Celular/metabolismo , Apoptose , Proteína de Domínio de Morte Associada a Fas/metabolismo , Complexos Multiproteicos/fisiologia , Transdução de Sinais
14.
Mol Cell ; 22(5): 599-610, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16762833

RESUMO

The structure of FADD has been solved in solution, revealing that the death effector domain (DED) and death domain (DD) are aligned with one another in an orthogonal, tail-to-tail fashion. Mutagenesis of FADD and functional reconstitution with its binding partners define the interaction with the intracellular domain of CD95 and the prodomain of procaspase-8 and reveal a self-association surface necessary to form a productive complex with an activated "death receptor." The identification of a procaspase-specific binding surface on the FADD DED suggests a preferential interaction with one, but not both, of the DEDs of procaspase-8 in a perpendicular arrangement. FADD self-association is mediated by a "hydrophobic patch" in the vicinity of F25 in the DED. The structure of FADD and its functional characterization, therefore, illustrate the architecture of key components in the death-inducing signaling complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Caspases/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Caspase 8 , Caspases/metabolismo , Proteína de Domínio de Morte Associada a Fas , Humanos , Células Jurkat , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Transdução de Sinais , Transfecção , Receptor fas/metabolismo
15.
J Biomol NMR ; 35(1): 17-25, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16791737

RESUMO

Measurement of residual dipolar couplings (RDCs) has become an important method for the determination and validation of protein or nucleic acid structures by NMRf spectroscopy. A number of toolkits have been devised for the handling of RDC data which run in the Linux/Unix operating environment and require specifically formatted input files. The outputs from these programs, while informative, require format modification prior to the incorporation of this data into commonly used personal computer programs for manuscript preparation. To bridge the gap between analysis and publication, an easy-to-use, comprehensive toolkit for RDC analysis has been created, iDC. iDC is written for the WaveMetrics Igor Pro mathematics program, a widely used graphing and data analysis software program that runs on both Windows PC and Mac OS X computers. Experimental RDC values can be loaded into iDC using simple data formats accessible to Igor's tabular data function. The program can perform most useful RDC analyses, including alignment tensor estimation from a histogram of RDC occurrence versus values and order tensor analysis by singular value decomposition (SVD). SVD analysis can be performed on an entire structure family at once, a feature missing in other applications of this kind. iDC can also import from and export to several different commonly used programs for the analysis of RDC data (DC, PALES, REDCAT) and can prepare formatted files for RDC-based refinement of macromolecular structures using XPLOR-NIH, CNS and ARIA. The graphical user interface provides an easy-to-use I/O for data, structures and formatted outputs.


Assuntos
Algoritmos , Ressonância Magnética Nuclear Biomolecular/métodos , Software , Conformação de Ácido Nucleico , Ácidos Nucleicos/química , Conformação Proteica , Proteínas/química , Interface Usuário-Computador
16.
J Biol Chem ; 280(36): 31974-80, 2005 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-16006552

RESUMO

The death domain and death effector domain are two common motifs that mediate protein-protein interactions between components of cell death signaling complexes. The mechanism by which these domains engage their binding partners has been explored by extensive mutagenesis of two death adaptors, FADD and TRADD, suggesting that a death adaptor can discriminate its intended binding partners from other proteins harboring similar motifs. Death adaptors are found to utilize one of two topologically conserved surfaces for protein-protein interaction, whether that partner is another adaptor or its cognate receptor. These surfaces are topologically related to the interaction between death domains observed in the x-ray crystal structure of the Drosophila adaptor Tube bound to Pelle kinase. Comparing the topology of protein-protein interactions for FADD complexes to TRADD complexes reveals that FADD uses a Tube-like surface in each of its death motifs to engage either CD95 or TRADD. TRADD reverses these roles, employing a Pelle-like surface to interact with either receptor TNFR1 or adaptor FADD. Since death adaptors display a Tube-like or Pelle-like preference for engaging their binding partners, Tube/Pelle-like pairing provides a mechanism for death adaptor discrimination of death receptors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Apoptose/fisiologia , Linhagem Celular , Proteína de Domínio de Morte Associada a Fas , Humanos , Células Jurkat , Dados de Sequência Molecular , Mutação , Ligação Proteica/fisiologia , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Transdução de Sinais/fisiologia , Receptor fas/metabolismo
17.
J Biol Chem ; 279(47): 49281-8, 2004 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-15377655

RESUMO

The acute human leukemias are associated with the presence of chimeric gene products that arise from spontaneous chromosomal translocations. The t(8;21) translocation gene product led to the discovery of the Eight Twenty-One (ETO) gene. When fused to RUNX1, ETO is thought to mediate the formation of a repressive complex at RUNX1-dependent genes. ETO has also been found to act as a co-repressor of the promyelocytic zinc finger and Bcl-6 oncoproteins, suggesting that it may play a common role as a transcriptional co-repressor leading to human disease. An analysis of ETO-mediated repression revealed that one of the key binding partners of ETO is the nuclear receptor co-repressor (N-CoR). It is shown that two highly conserved domains of ETO interact with repression domains I and III of N-CoR. One of the ETO domains displays significant homology to Drosophila TAF(II)110, whereas the other is a predicted zinc binding motif that engages a conserved PPLXP motif in repression domain III of N-CoR. Together, these domains of ETO cooperate in repression with N-CoR and the binding sites in N-CoR overlap with those for other repressive factors. Thus, ETO has the potential to participate in a number of repressive complexes, which can be distinguished by their binding partners and target genes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/fisiologia , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Linhagem Celular , Cisteína/química , DNA/metabolismo , Proteínas de Ligação a DNA/química , Relação Dose-Resposta a Droga , Drosophila , Deleção de Genes , Genes Reporter , Glutationa Transferase/metabolismo , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/química , Correpressor 1 de Receptor Nuclear , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/química , Proteína 1 Parceira de Translocação de RUNX1 , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/química , Homologia de Sequência de Aminoácidos , Serina/química , Fatores de Transcrição/química , Transcrição Gênica , Transfecção , Translocação Genética , Zinco/química , Dedos de Zinco
18.
EMBO J ; 23(15): 2952-62, 2004 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-15257291

RESUMO

Bacteriophage T4 AsiA is a versatile transcription factor capable of inhibiting host gene expression as an 'anti-sigma' factor while simultaneously promoting gene-specific expression of T4 middle genes in conjunction with T4 MotA. To accomplish this task, AsiA engages conserved region 4 of Eschericia coli sigma70, blocking recognition of most host promoters by sequestering the DNA-binding surface at the AsiA/sigma70 interface. The three-dimensional structure of an AsiA/region 4 complex reveals that the C-terminal alpha helix of region 4 is unstructured, while four other helices adopt a completely different conformation relative to the canonical structure of unbound region 4. That AsiA induces, rather than merely stabilizes, this rearrangement can be realized by comparison to the homologous structures of region 4 solved in a variety of contexts, including the structure of Thermotoga maritima sigmaA region 4 described herein. AsiA simultaneously occupies the surface of region 4 that ordinarily contacts core RNA polymerase (RNAP), suggesting that an AsiA-bound sigma70 may also undergo conformational changes in the context of the RNAP holoenzyme.


Assuntos
DNA Bacteriano/metabolismo , RNA Polimerases Dirigidas por DNA/química , RNA Polimerases Dirigidas por DNA/metabolismo , Fator sigma/química , Fator sigma/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Escherichia coli/química , Escherichia coli/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Dobramento de Proteína , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência , Especificidade por Substrato
19.
J Biol Chem ; 279(2): 1474-81, 2004 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-14573612

RESUMO

The initiation of programmed cell death at CD95 (Fas, Apo-1) is achieved by forming a death-inducing signaling complex (DISC) at the cytoplasmic membrane surface. Assembly of the DISC has been proposed to occur via homotypic interactions between the death domain (DD) of FADD and the cytoplasmic domain of CD95. Previous analysis of the FADD/CD95 interaction led to the identification of a putative CD95 binding surface within FADD DD formed by alpha helices 2 and 3. More detailed analysis of the CD95/FADD DD interaction now demonstrates that a bimodal surface exists in the FADD DD for interaction with CD95. An expansive surface on one side of the domain is composed of elements in alpha helices 1, 2, 3, 5, and 6. This major surface is common to many proteins harboring this motif, whether or not they are associated with programmed cell death. A secondary surface resides on the opposite face of the domain and involves residues in helices 3 and 4. The major surface is topologically similar to the protein interaction surface identified in Drosophila Tube DD and the death effector domain of hamster PEA-15, two physiologically unrelated proteins which interact with structurally unrelated binding partners. These results demonstrate the presence of a structurally conserved surface within the DD which can mediate protein recognition with homo- and heterotypic binding partners, whereas a second surface may be responsible for stabilizing the higher order complex in the DISC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/química , Receptor fas/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sobrevivência Celular , Citoplasma/metabolismo , DNA Complementar/metabolismo , Drosophila/metabolismo , Proteína de Domínio de Morte Associada a Fas , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Testes de Precipitina , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo , Receptor fas/química , Receptor fas/metabolismo
20.
J Biol Chem ; 278(52): 52587-97, 2003 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-14506247

RESUMO

Activation of Raf-1 suppresses integrin activation, potentially through the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). However, bulk ERK1/2 activation does not correlate with suppression. PEA-15 reverses suppression of integrin activation and binds ERK1/2. Here we report that PEA-15 reversal of integrin suppression depends on its capacity to bind ERK1/2, indicating that ERK1/2 function is indeed required for suppression. Mutations in either the death effector domain or C-terminal tail of PEA-15 that block ERK1/2 binding abrogated the reversal of integrin suppression. Furthermore, we used ERK/p38 chimeras and site-directed mutagenesis to identify ERK1/2 residues required for binding PEA-15. Mutations of residues that precede the alphaG helix and within the mitogen-activated protein kinase insert blocked ERK2 binding to PEA-15, but not activation of ERK2. These ERK2 mutants blocked the ability of PEA-15 to reverse suppression of integrin activation. Thus, PEA-15 regulation of integrin activation depends on its binding to ERK1/2. To directly test the role of ERK1/2 localization in suppression, we enforced membrane association of ERK1 and 2 by joining a membrane-targeting CAAX box sequence to them. Both ERK1-CAAX and ERK2-CAAX were membrane-localized and suppressed integrin activation. In contrast to suppression by membrane-targeted Raf-CAAX, suppression by ERK1/2-CAAX was not reversed by PEA-15. Thus, ERK1/2 are the Raf effectors for suppression of integrin activation, and PEA-15 reverses suppression by binding ERK1/2.


Assuntos
Integrinas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Motivos de Aminoácidos , Animais , Células CHO , Membrana Celular/metabolismo , Separação Celular , Cricetinae , DNA Complementar/metabolismo , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Proteínas de Fluorescência Verde , Immunoblotting , Ligantes , Proteínas Luminescentes/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Modelos Genéticos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Fosfoproteínas/química , Mutação Puntual , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA