Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Huntingtons Dis ; 11(3): 269-280, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35848031

RESUMO

Huntingtin is a 3144 amino acid protein defined as a scaffold protein with many intracellular locations that suggest functions in these compartments. Expansion of the CAG DNA tract in the huntingtin first exon is the cause of Huntington's disease. An important tool in understanding the biological functions of huntingtin is molecular imaging at the single-cell level by microscopy and nanoscopy. The evolution of these technologies has accelerated since the Nobel Prize in Chemistry was awarded in 2014 for super-resolution nanoscopy. We are in a new era of light imaging at the single-cell level, not just for protein location, but also for protein conformation and biochemical function. Large-scale microscopy-based screening is also being accelerated by a coincident development of machine-based learning that offers a framework for truly unbiased data acquisition and analysis at very large scales. This review will summarize the newest technologies in light, electron, and atomic force microscopy in the context of unique challenges with huntingtin cell biology and biochemistry.


Assuntos
Doença de Huntington , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/genética , Doença de Huntington/metabolismo , Microscopia , Proteínas do Tecido Nervoso/genética
2.
Hum Mol Genet ; 30(8): 706-715, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-33772540

RESUMO

Spinocerebellar Ataxia Type 1 (SCA1) is an autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion in the ataxin-1 protein. Recent genetic correlational studies have implicated DNA damage repair pathways in modifying the age at onset of disease symptoms in SCA1 and Huntington's Disease, another polyglutamine expansion disease. We demonstrate that both endogenous and transfected ataxin-1 localizes to sites of DNA damage, which is impaired by polyglutamine expansion. This response is dependent on ataxia-telangiectasia mutated (ATM) kinase activity. Further, we characterize an ATM phosphorylation motif within ataxin-1 at serine 188. We show reduction of the Drosophila ATM homolog levels in a ATXN1[82Q] Drosophila model through shRNA or genetic cross ameliorates motor symptoms. These findings offer a possible explanation as to why DNA repair was implicated in SCA1 pathogenesis by past studies. The similarities between the ataxin-1 and the huntingtin responses to DNA damage provide further support for a shared pathogenic mechanism for polyglutamine expansion diseases.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Ataxina-1/genética , Dano ao DNA , Ataxias Espinocerebelares/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Ataxina-1/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Drosophila/genética , Drosophila/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Microscopia Confocal , Mutação , Peptídeos/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais/genética , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/patologia , Repetições de Trinucleotídeos/genética
3.
J Huntingtons Dis ; 10(1): 165-173, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33579859

RESUMO

The use of genome wide association studies (GWAS) in Huntington's disease (HD) research, driven by unbiased human data analysis, has transformed the focus of new targets that could affect age at onset. While there is a significant depth of information on DNA damage repair, with many drugs and drug targets, most of this development has taken place in the context of cancer therapy. DNA damage repair in neurons does not rely on DNA replication correction mechanisms. However, there is a strong connection between DNA repair and neuronal metabolism, mediated by nucleotide salvaging and the poly ADP-ribose (PAR) response, and this connection has been implicated in other age-onset neurodegenerative diseases. Validation of leads including the mismatch repair protein MSH3, and interstrand cross-link repair protein FAN1, suggest the mechanism is driven by somatic CAG instability, which is supported by the protective effect of CAA substitutions in the CAG tract. We currently do not understand: how somatic instability is triggered; the state of DNA damage within expanding alleles in the brain; whether this damage induces mismatch repair and interstrand cross-link pathways; whether instability mediates toxicity, and how this relates to human ageing. We discuss DNA damage pathways uncovered by HD GWAS, known roles of other polyglutamine disease proteins in DNA damage repair, and a panel of hypotheses for pathogenic mechanisms.


Assuntos
Reparo do DNA/genética , Estudo de Associação Genômica Ampla , Instabilidade Genômica/genética , Doença de Huntington/genética , Ataxias Espinocerebelares/genética , Humanos
4.
Neuron ; 107(5): 768-769, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32910889

RESUMO

Using two independent single-cell transcriptomics technologies, Lee et al. have cataloged transcriptional changes in the subset of striatal neurons hit hardest in Huntington's disease. One downregulated pathway, oxidative phosphorylation, may also explain their observed release of mitochondrial-encoded RNAs.


Assuntos
Doença de Huntington , Corpo Estriado , Humanos , Doença de Huntington/genética , Doença de Huntington/terapia , Imunidade Inata , Neurônios , RNA Mitocondrial , Transcriptoma
5.
DNA Cell Biol ; 38(1): 1-6, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30481056

RESUMO

A new hypothesis for the mechanism of Huntington's disease (HD) is driven by a small molecule lead that may connect age-associated reactive oxygen stress, oxidative DNA damage, and mitochondrial dysfunction. These pathways have also recently been defined in genome-wide association studies of cytosine-adenine-guanine-expansion polyglutamine neurodegenerative diseases, including HD and the spinocerebellar ataxias. We discuss how N6-furfuryladenine (N6FFA) nucleotide salvage and role as a kinase neosubstrate may have important mechanistic implications for both HD and familial Parkinson's disease. N6FFA highlights a mechanism of how energy dysregulation and protein misfolding in neurodegeneration may be the effect of age-associated reactive oxygen species damage to DNA and part of a feedback loop augmenting with aging.


Assuntos
Dano ao DNA , Reparo do DNA , Proteína Huntingtina/genética , Doenças Neurodegenerativas/genética , Estresse Oxidativo/genética , Animais , Humanos , Proteína Huntingtina/metabolismo , Doenças Neurodegenerativas/metabolismo , Transdução de Sinais
6.
J Biol Chem ; 294(6): 1915-1923, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30538129

RESUMO

Huntington's disease (HD) is a neurodegenerative, age-onset disorder caused by a CAG DNA expansion in exon 1 of the HTT gene, resulting in a polyglutamine expansion in the huntingtin protein. Nuclear accumulation of mutant huntingtin is a hallmark of HD, resulting in elevated mutant huntingtin levels in cell nuclei. Huntingtin is normally retained at the endoplasmic reticulum via its N17 amphipathic α-helix domain but is released by oxidation of Met-8 during reactive oxygen species (ROS) stress. Huntingtin enters the nucleus via an importin ß1- and 2-dependent proline-tyrosine nuclear localization signal (PY-NLS), which has a unique intervening sequence in huntingtin. Here, we have identified the high-mobility group box 1 (HMGB1) protein as an interactor of the intervening sequence within the PY-NLS. Nuclear levels of HMGB1 positively correlated with varying levels of nuclear huntingtin in both HD and normal human fibroblasts. We also found that HMGB1 interacts with the huntingtin N17 region and that this interaction is enhanced by the presence of ROS and phosphorylation of critical serine residues in the N17 region. We conclude that HMGB1 is a huntingtin N17/PY-NLS ROS-dependent interactor, and this protein bridging is essential for relaying ROS sensing by huntingtin to its nuclear entry during ROS stress. ROS may therefore be a critical age-onset stress that triggers nuclear accumulation of mutant huntington in Huntington's disease.


Assuntos
Transporte Ativo do Núcleo Celular , Proteína HMGB1/fisiologia , Proteína Huntingtina/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Sítios de Ligação , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Proteína Huntingtina/efeitos dos fármacos , Proteína Huntingtina/fisiologia , Sinais de Localização Nuclear , Proteínas Nucleares/metabolismo , Fosforilação , Ligação Proteica
7.
Mol Biol Cell ; 29(23): 2809-2820, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30256717

RESUMO

The huntingtin protein participates in several cellular processes that are disrupted when the polyglutamine tract is expanded beyond a threshold of 37 CAG DNA repeats in Huntington's disease (HD). Cellular biology approaches to understand these functional disruptions in HD have primarily focused on cell lines with synthetically long CAG length alleles that clinically represent outliers in this disease and a more severe form of HD that lacks age onset. Patient-derived fibroblasts are limited to a finite number of passages before succumbing to cellular senescence. We used human telomerase reverse transcriptase (hTERT) to immortalize fibroblasts taken from individuals of varying age, sex, disease onset, and CAG repeat length, which we have termed TruHD cells. TruHD cells display classic HD phenotypes of altered morphology, size and growth rate, increased sensitivity to oxidative stress, aberrant adenosine diphosphate/adenosine triphosphate (ADP/ATP) ratios, and hypophosphorylated huntingtin protein. We additionally observed dysregulated reactive oxygen species (ROS)-dependent huntingtin localization to nuclear speckles in HD cells. We report the generation and characterization of a human, clinically relevant cellular model for investigating disease mechanisms in HD at the single-cell level, which, unlike transformed cell lines, maintains functions critical for huntingtin transcriptional regulation and genomic integrity.


Assuntos
Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Adulto , Sequência de Bases/genética , Encéfalo/metabolismo , Linhagem Celular/metabolismo , Senescência Celular/genética , Feminino , Fibroblastos/metabolismo , Humanos , Doença de Huntington/fisiopatologia , Cariotipagem , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fenótipo , Cultura Primária de Células , Telomerase , Repetições de Trinucleotídeos/genética , Repetições de Trinucleotídeos/fisiologia
8.
Proc Natl Acad Sci U S A ; 115(30): E7081-E7090, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29987005

RESUMO

The huntingtin N17 domain is a modulator of mutant huntingtin toxicity and is hypophosphorylated in Huntington's disease (HD). We conducted high-content analysis to find compounds that could restore N17 phosphorylation. One lead compound from this screen was N6-furfuryladenine (N6FFA). N6FFA was protective in HD model neurons, and N6FFA treatment of an HD mouse model corrects HD phenotypes and eliminates cortical mutant huntingtin inclusions. We show that N6FFA restores N17 phosphorylation levels by being salvaged to a triphosphate form by adenine phosphoribosyltransferase (APRT) and used as a phosphate donor by casein kinase 2 (CK2). N6FFA is a naturally occurring product of oxidative DNA damage. Phosphorylated huntingtin functionally redistributes and colocalizes with CK2, APRT, and N6FFA DNA adducts at sites of induced DNA damage. We present a model in which this natural product compound is salvaged to provide a triphosphate substrate to signal huntingtin phosphorylation via CK2 during low-ATP stress under conditions of DNA damage, with protective effects in HD model systems.


Assuntos
Adenina , Adutos de DNA/metabolismo , Dano ao DNA , Doença de Huntington/tratamento farmacológico , Neurônios/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adenina/análogos & derivados , Adenina/farmacocinética , Adenina/farmacologia , Adenina Fosforribosiltransferase/genética , Adenina Fosforribosiltransferase/metabolismo , Animais , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Linhagem Celular Transformada , Adutos de DNA/genética , Modelos Animais de Doenças , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Transdução de Sinais/genética
9.
Hum Mol Genet ; 26(2): 395-406, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28017939

RESUMO

Huntington's disease (HD) is an age-dependent neurodegenerative disease. DNA repair pathways have recently been implicated as the most predominant modifiers of age of onset in HD patients. We report that endogenous huntingtin protein directly participates in oxidative DNA damage repair. Using novel chromobodies to detect endogenous human huntingtin in live cells, we show that localization of huntingtin to DNA damage sites is dependent on the kinase activity of ataxia telangiectasia mutated (ATM) protein. Super-resolution microscopy and biochemical assays revealed that huntingtin co-localizes with and scaffolds proteins of the DNA damage response pathway in response to oxidative stress. In HD patient fibroblasts bearing typical clinical HD allele lengths, we demonstrate that there is deficient oxidative DNA damage repair. We propose that DNA damage in HD is caused by dysfunction of the mutant huntingtin protein in DNA repair, and accumulation of DNA oxidative lesions due to elevated reactive oxygen species may contribute to the onset of HD.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteína Huntingtina/genética , Doença de Huntington/genética , Estresse Oxidativo/genética , Alelos , Dano ao DNA/genética , Reparo do DNA/genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo
10.
Commun Integr Biol ; 6(2): e23318, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23750301

RESUMO

Nuclear accumulation of the polyglutamine-expanded mutant huntingtin protein remains one of the most predictive cell biological phenotypes of Huntington's disease (HD) progression in patient brain samples and mouse models of the disease. Yet, the relationship between huntingtin nuclear import, neuronal dysfunction and toxicity is not fully understood and it remains unclear whether nuclear accumulation is required for disease onset. Here, we discuss several studies that have guided current understanding of this subject, and highlight our recent data detailing the discovery of a karyopherin ß1/ß2-type nuclear localization signal near the N-terminus of huntingtin. This signal can function through multiple pathways of nuclear import, and may also be responsible for huntingtin import into the primary cilium. This work represents a significant step forward in our knowledge of the regulatory pathways that govern huntingtin nuclear accumulation and will allow direct examination of both normal and mutant huntingtin nuclear function. This work also suggests a re-examination of the cell biology of any protein that contains a multi-pathway nuclear localization signal. The possibility of targeting huntingtin nuclear import therapeutically and the potential impacts of such a strategy for the treatment of HD are also discussed.

11.
Nat Chem Biol ; 7(7): 453-60, 2011 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-21623356

RESUMO

Two serine residues within the first 17 amino acid residues of huntingtin (N17) are crucial for modulation of mutant huntingtin toxicity in cell and mouse genetic models of Huntington's disease. Here we show that the stress-dependent phosphorylation of huntingtin at Ser13 and Ser16 affects N17 conformation and targets full-length huntingtin to chromatin-dependent subregions of the nucleus, the mitotic spindle and cleavage furrow during cell division. Polyglutamine-expanded mutant huntingtin is hypophosphorylated in N17 in both homozygous and heterozygous cell contexts. By high-content screening in live cells, we identified kinase inhibitors that modulated N17 phosphorylation and hence huntingtin subcellular localization. N17 phosphorylation was reduced by casein kinase-2 inhibitors. Paradoxically, IKKß kinase inhibition increased N17 phosphorylation, affecting huntingtin nuclear and subnuclear localization. These data indicate that huntingtin phosphorylation at Ser13 and Ser16 can be modulated by small-molecule drugs, which may have therapeutic potential in Huntington's disease.


Assuntos
Caseína Quinase II/antagonistas & inibidores , Doença de Huntington/metabolismo , Quinase I-kappa B/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Inibidores de Proteínas Quinases/farmacologia , Animais , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Imunofluorescência , Proteína Huntingtina , Doença de Huntington/enzimologia , Doença de Huntington/genética , Camundongos , Mutação , Fosforilação , Serina/genética , Fuso Acromático/metabolismo , Transfecção
12.
J Clin Invest ; 120(9): 3310-25, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20697156

RESUMO

Some cancers have been stratified into subclasses based on their unique involvement of specific signaling pathways. The mapping of human cancer genomes is revealing a vast number of somatic alterations; however, the identification of clinically relevant molecular tumor subclasses and their respective driver genes presents challenges. This information is key to developing more targeted and personalized cancer therapies. Here, we generate a new mouse model of genomically unstable osteosarcoma (OSA) that phenocopies the human disease. Integrative oncogenomics pinpointed cAMP-dependent protein kinase type I, alpha regulatory subunit (Prkar1a) gene deletions at 11qE1 as a recurrent genetic trait for a molecularly distinct subclass of mouse OSA featuring RANKL overexpression. Using mouse genetics, we established that Prkar1a is a bone tumor suppressor gene capable of directing subclass development and driving RANKL overexpression during OSA tumorigenesis. Finally, we uncovered evidence for a PRKAR1A-low subset of human OSA with distinct clinical behavior. Thus, tumor subclasses develop in mice and can potentially provide information toward the molecular stratification of human cancers.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Genes Supressores de Tumor , Osteossarcoma/genética , Animais , Modelos Animais de Doenças , Deleção de Genes , Camundongos , Camundongos Transgênicos , Fenótipo
13.
J Biol Chem ; 285(20): 15038-15047, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20223817

RESUMO

The phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB)/Akt-PTEN signal transduction pathway orchestrates a variety of fundamental cell processes and its deregulation is implicated in many human diseases. Although the importance of this pathway to many cellular functions is well established, the mechanisms by which it achieves context-specific physiological outcomes in response to a variety of stimuli, using a relatively limited pool of effectors, remain largely unknown. Spatial restriction of signaling events is one means by which cells coordinate specific responses using common molecules. To investigate the subcellular location-specific roles of the major PI3K effector PKB/Akt in various cell processes, we have developed a novel experimental system employing cellular compartment-directed PKB/Akt pseudosubstrate inhibitors. Subcellular location-restricted PKB/Akt inhibition in the 3T3L1 adipocyte differentiation model revealed that nuclear and plasma membrane, but not cytoplasmic, PKB/Akt activity is required for terminal adipocyte differentiation. Nuclear and plasma membrane pools of PKB/Akt were found to contribute to distinct stages of adipocyte differentiation, revealing that PKB/Akt activity impacts multiple points of this program. Our work establishes the use of localized pseudosubstrate PKB/Akt inhibitors as an effective method for the dissection of PKB/Akt signaling.


Assuntos
Adipócitos/citologia , Diferenciação Celular , Proteínas Proto-Oncogênicas c-akt/metabolismo , Frações Subcelulares/enzimologia , Células 3T3-L1 , Adipócitos/enzimologia , Animais , Camundongos , Microscopia de Fluorescência , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA