Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Ther ; 31(7): 2014-2027, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-36932675

RESUMO

Blue cone monochromacy (BCM) is a rare X-linked retinal disease characterized by the absence of L- and M-opsin in cone photoreceptors, considered a potential gene therapy candidate. However, most experimental ocular gene therapies utilize subretinal vector injection which would pose a risk to the fragile central retinal structure of BCM patients. Here we describe the use of ADVM-062, a vector optimized for cone-specific expression of human L-opsin and administered using a single intravitreal (IVT) injection. Pharmacological activity of ADVM-062 was established in gerbils, whose cone-rich retina naturally lacks L-opsin. A single IVT administration dose of ADVM-062 effectively transduced gerbil cone photoreceptors and produced a de novo response to long-wavelength stimuli. To identify potential first-in-human doses we evaluated ADVM-062 in non-human primates. Cone-specific expression of ADVM-062 in primates was confirmed using ADVM-062.myc, a vector engineered with the same regulatory elements as ADVM-062. Enumeration of human OPN1LW.myc-positive cones demonstrated that doses ≥3 × 1010 vg/eye resulted in transduction of 18%-85% of foveal cones. A Good Laboratory Practice (GLP) toxicology study established that IVT administration of ADVM-062 was well tolerated at doses that could potentially achieve clinically meaningful effect, thus supporting the potential of ADVM-062 as a one-time IVT gene therapy for BCM.


Assuntos
Opsinas , Células Fotorreceptoras Retinianas Cones , Animais , Humanos , Células Fotorreceptoras Retinianas Cones/metabolismo , Opsinas/genética , Primatas/genética , Primatas/metabolismo , Opsinas de Bastonetes/genética , Opsinas de Bastonetes/metabolismo , Terapia Genética/métodos
2.
Sci Adv ; 7(12)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33741591

RESUMO

Neuronal tau reduction confers resilience against ß-amyloid and tau-related neurotoxicity in vitro and in vivo. Here, we introduce a novel translational approach to lower expression of the tau gene MAPT at the transcriptional level using gene-silencing zinc finger protein transcription factors (ZFP-TFs). Following a single administration of adeno-associated virus (AAV), either locally into the hippocampus or intravenously to enable whole-brain transduction, we selectively reduced tau messenger RNA and protein by 50 to 80% out to 11 months, the longest time point studied. Sustained tau lowering was achieved without detectable off-target effects, overt histopathological changes, or molecular alterations. Tau reduction with AAV ZFP-TFs was able to rescue neuronal damage around amyloid plaques in a mouse model of Alzheimer's disease (APP/PS1 line). The highly specific, durable, and controlled knockdown of endogenous tau makes AAV-delivered ZFP-TFs a promising approach for the treatment of tau-related human brain diseases.


Assuntos
Doença de Alzheimer , Fatores de Transcrição , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Camundongos , Placa Amiloide/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Dedos de Zinco/genética , Proteínas tau/genética , Proteínas tau/metabolismo
4.
PLoS One ; 9(8): e102909, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170892

RESUMO

Detailed analysis of disease-affected tissue provides insight into molecular mechanisms contributing to pathogenesis. Substantia nigra, striatum, and cortex are functionally connected with increasing degrees of alpha-synuclein pathology in Parkinson's disease. We undertook functional and causal pathway analysis of gene expression and proteomic alterations in these three regions, and the data revealed pathways that correlated with disease progression. In addition, microarray and RNAseq experiments revealed previously unidentified causal changes related to oligodendrocyte function and synaptic vesicle release, and these and other changes were reflected across all brain regions. Importantly, subsets of these changes were replicated in Parkinson's disease blood; suggesting peripheral tissue may provide important avenues for understanding and measuring disease status and progression. Proteomic assessment revealed alterations in mitochondria and vesicular transport proteins that preceded gene expression changes indicating defects in translation and/or protein turnover. Our combined approach of proteomics, RNAseq and microarray analyses provides a comprehensive view of the molecular changes that accompany functional loss and alpha-synuclein pathology in Parkinson's disease, and may be instrumental to understand, diagnose and follow Parkinson's disease progression.


Assuntos
Encéfalo/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Animais , Encéfalo/metabolismo , Progressão da Doença , Regulação da Expressão Gênica , Humanos , Análise em Microsséries , Proteínas/análise , Proteínas/genética , Proteínas/metabolismo , Proteômica , Análise de Sequência de RNA , Transdução de Sinais , alfa-Sinucleína/análise , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
5.
PLoS One ; 8(7): e66879, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23861750

RESUMO

Surface Plasmon Resonance (SPR) is rarely used as a primary High-throughput Screening (HTS) tool in fragment-based approaches. With SPR instruments becoming increasingly high-throughput it is now possible to use SPR as a primary tool for fragment finding. SPR becomes, therefore, a valuable tool in the screening of difficult targets such as the ubiquitin E3 ligase Parkin. As a prerequisite for the screen, a large number of SPR tests were performed to characterize and validate the active form of Parkin. A set of compounds was designed and used to define optimal SPR assay conditions for this fragment screen. Using these conditions, more than 5000 pre-selected fragments from our in-house library were screened for binding to Parkin. Additionally, all fragments were simultaneously screened for binding to two off target proteins to exclude promiscuous binding compounds. A low hit rate was observed that is in line with hit rates usually obtained by other HTS screening assays. All hits were further tested in dose responses on the target protein by SPR for confirmation before channeling the hits into Nuclear Magnetic Resonance (NMR) and other hit-confirmation assays.


Assuntos
Ensaios de Triagem em Larga Escala , Fragmentos de Peptídeos/química , Ressonância de Plasmônio de Superfície , Ubiquitina-Proteína Ligases/química , Ditiotreitol/química , Ditiotreitol/metabolismo , Descoberta de Drogas , Ensaios de Triagem em Larga Escala/métodos , Cinética , Ligantes , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Substâncias Redutoras/química , Substâncias Redutoras/metabolismo , Ressonância de Plasmônio de Superfície/métodos , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo
6.
J Cell Biol ; 196(5): 573-87, 2012 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-22371559

RESUMO

Pathognomonic accumulation of ubiquitin (Ub) conjugates in human neurodegenerative diseases, such as Huntington's disease, suggests that highly aggregated proteins interfere with 26S proteasome activity. In this paper, we examine possible mechanisms by which an N-terminal fragment of mutant huntingtin (htt; N-htt) inhibits 26S function. We show that ubiquitinated N-htt-whether aggregated or not-did not choke or clog the proteasome. Both Ub-dependent and Ub-independent proteasome reporters accumulated when the concentration of mutant N-htt exceeded a solubility threshold, indicating that stabilization of 26S substrates is not linked to impaired Ub conjugation. Above this solubility threshold, mutant N-htt was rapidly recruited to cytoplasmic inclusions that were initially devoid of Ub. Although synthetically polyubiquitinated N-htt competed with other Ub conjugates for access to the proteasome, the vast majority of mutant N-htt in cells was not Ub conjugated. Our data confirm that proteasomes are not directly impaired by aggregated N-terminal fragments of htt; instead, our data suggest that Ub accumulation is linked to impaired function of the cellular proteostasis network.


Assuntos
Doença de Huntington/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Ubiquitina/metabolismo , Animais , Linhagem Celular , Estabilidade Enzimática , Genes Reporter , Células HEK293 , Humanos , Proteína Huntingtina , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitina/genética , Ubiquitinação
7.
Nat Methods ; 8(8): 691-6, 2011 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-21743460

RESUMO

The protein ubiquitin is an important post-translational modifier that regulates a wide variety of biological processes. In cells, ubiquitin is apportioned among distinct pools, which include a variety of free and conjugated species. Although maintenance of a dynamic and complex equilibrium among ubiquitin pools is crucial for cell survival, the tools necessary to quantify each cellular ubiquitin pool have been limited. We have developed a quantitative mass spectrometry approach to measure cellular concentrations of ubiquitin species using isotope-labeled protein standards and applied it to characterize ubiquitin pools in cells and tissues. Our method is convenient, adaptable and should be a valuable tool to facilitate our understanding of this important signaling molecule.


Assuntos
Marcação por Isótopo/métodos , Espectrometria de Massas/métodos , Espectrometria de Massas/normas , Frações Subcelulares/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Estados Unidos
9.
J Cell Biol ; 191(3): 537-52, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21041446

RESUMO

Genetic ablation of autophagy in mice leads to liver and brain degeneration accompanied by the appearance of ubiquitin (Ub) inclusions, which has been considered to support the hypothesis that ubiquitination serves as a cis-acting signal for selective autophagy. We show that tissue-specific disruption of the essential autophagy genes Atg5 and Atg7 leads to the accumulation of all detectable Ub-Ub topologies, arguing against the hypothesis that any particular Ub linkage serves as a specific autophagy signal. The increase in Ub conjugates in Atg7(-/-) liver and brain is completely suppressed by simultaneous knockout of either p62 or Nrf2. We exploit a novel assay for selective autophagy in cell culture, which shows that inactivation of Atg5 leads to the selective accumulation of aggregation-prone proteins, and this does not correlate with an increase in substrate ubiquitination. We propose that protein oligomerization drives autophagic substrate selection and that the accumulation of poly-Ub chains in autophagy-deficient circumstances is an indirect consequence of activation of Nrf2-dependent stress response pathways.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Estresse Fisiológico/fisiologia , Ubiquitina/metabolismo , Animais , Autofagia , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Células Cultivadas , Camundongos , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Ligação Proteica , Especificidade por Substrato
10.
Genes Dev ; 20(16): 2183-92, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16912271

RESUMO

The polyglutamine disorders are a class of nine neuro-degenerative disorders that are inherited gain-of-function diseases caused by expansion of a translated CAG repeat. Even though the disease-causing proteins are widely expressed, specific collections of neurons are more susceptible in each disease, resulting in characteristic patterns of pathology and clinical symptoms. One hypothesis poses that altered protein function is fundamental to pathogenesis, with protein context of the expanded polyglutamine having key roles in disease-specific processes. This review will focus on the role of the disease-causing polyglutamine proteins in gene transcription and the extent to which the mutant proteins induce disruption of transcription.


Assuntos
Regulação da Expressão Gênica , Doenças Neurodegenerativas/genética , Peptídeos/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Ligação Proteica , Expansão das Repetições de Trinucleotídeos
12.
J Biol Chem ; 280(48): 40282-92, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16192271

RESUMO

CNS neurons are endowed with the ability to recover from cytotoxic insults associated with the accumulation of proteinaceous polyglutamine aggregates via a process that appears to involve capture and degradation of aggregates by autophagy. The ubiquitin-proteasome system protects cells against proteotoxicity by degrading soluble monomeric misfolded aggregation-prone proteins but is ineffective against, and impaired by, non-native protein oligomers. Here we show that autophagy is induced in response to impaired ubiquitin proteasome system activity. We show that ATG proteins, molecular determinants of autophagic vacuole formation, and lysosomes are recruited to pericentriolar cytoplasmic inclusion bodies by a process requiring an intact microtubule cytoskeleton and the cytoplasmic deacetylase HDAC6. These data suggest that HDAC6-dependent retrograde transport on microtubules is used by cells to increase the efficiency and selectivity of autophagic degradation.


Assuntos
Autofagia , Histona Desacetilases/fisiologia , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/química , Transporte Biológico , Linhagem Celular , Citoplasma/metabolismo , Citoesqueleto/metabolismo , DNA Complementar/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Desacetilase 6 de Histona , Histona Desacetilases/metabolismo , Humanos , Proteína Huntingtina , Lisossomos/metabolismo , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Interferência de RNA , Fatores de Tempo , Transcrição Gênica , Transfecção , Ubiquitina/química , Vacúolos/metabolismo
13.
J Biol Chem ; 280(23): 21942-8, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15824120

RESUMO

SUMO (small ubiquitin-like modifier) is a member of the ubiquitin family of proteins. SUMO targets include proteins involved in numerous roles including nuclear transport and transcriptional regulation. The previous finding that mutant ataxin-1[82Q] disrupted promyelocytic leukemia (PML) oncogenic domains prompted us to determine whether ataxin-1 disrupts another component of PML oncogenic domains, Sp100 (100-kDa Speckled protein). Similar to the PML protein, mutant ataxin-1[82Q] redistributed Sp100 to mutant ataxin-1[82Q] nuclear inclusions. Based on the ability of PML and Sp100 to be covalently modified by SUMO, we investigated the ability of ataxin-1 to be SUMOylated. SUMO-1 was found to covalently modify the polyglutamine repeat protein ataxin-1. There was a decrease in ataxin-1 SUMOylation in the presence of the expanded polyglutamine tract, ataxin-1[82Q]. The phospho-mutant, ataxin-1[82Q]-S776A, restored SUMO levels to those of wild-type ataxin-1[30Q]. SUMOylation of ataxin-1 was dependent on a functional nuclear localization signal. Ataxin-1 SUMOylation was mapped to at least five lysine residues. Lys(16), Lys(194) preceding the polyglutamine tract, Lys(610)/Lys(697) in the C-terminal ataxin high mobility group domain, and Lys(746) all contribute to ataxin-1 SUMOylation.


Assuntos
Núcleo Celular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Animais , Antígenos Nucleares/metabolismo , Ataxina-1 , Ataxinas , Autoantígenos/metabolismo , Sítios de Ligação , Células COS , Linhagem Celular , Detergentes/farmacologia , Imunoprecipitação , Lisina/química , Microscopia de Fluorescência , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/química , Sinais de Localização Nuclear , Proteínas Nucleares/química , Octoxinol/farmacologia , Peptídeos/química , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Proteínas Supressoras de Tumor
14.
J Biol Chem ; 279(40): 42290-301, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15280365

RESUMO

The ataxin-1 interacting ubiquitin-like protein (A1Up) contains an amino-terminal ubiquitin-like (UbL) region, four stress-inducible, heat shock chaperonin-binding motifs (STI1), and an ubiquitin-associated domain (UBA) at the carboxyl terminus of A1Up. Although proteins that have both an UbL and UBA domain are thought to play a crucial role in proteasome-mediated activities, few are characterized, except for hHR23A/B. Similar to other UbL-containing proteins, the UbL of A1Up is essential for the interaction of A1Up with the S5a subunit of the 19S proteasome. Importantly, the interaction with the 19S proteasome was disrupted in the presence of the polyglutamine repeat protein, ataxin-1. The UbL domain of A1Up is ubiquitinated by both Lys(48)-linked and Lys(63)-linked chains. Intact A1Up is stable, suggesting that ubiquitination of A1Up is important for degradation-independent targeting of A1Up to the 19S proteasome. The UBA domain of A1Up binds polyubiquitin chains and has a role in the stability of A1Up and in the subcellular localization of A1Up. When the UBA domain was deleted, the localization of A1Up was entirely cytoplasmic, and it co-localized with the proteasome. Interestingly, the interaction between A1Up and mutant ataxin-1-(82Q) increased the half-life of A1Up, whereas nonpathogenic wild-type ataxin-1-(30Q) or ataxin-1-(82Q)-A776 did not.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Peptídeos , Animais , Ataxina-1 , Ataxinas , Proteínas de Transporte/química , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/química , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Transfecção , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA