Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Kidney Int Rep ; 4(10): 1387-1400, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31701048

RESUMO

INTRODUCTION: Therapeutic agents that target complement are increasingly available for glomerular diseases. However, the mechanisms linking glomerular complement deposition with inflammation and damage are incompletely understood. Complement factor H-related protein 5 (FHR5) interacts with complement C3 and is considered to promote activation. Circulating and glomerular FHR5 associates with IgA nephropathy and abnormal FHR5 associates with familial C3 glomerulopathy (C3G). We characterized glomerular FHR5 staining in C3G and assessed its relationships with histological features of glomerular injury and clinical outcome. METHODS: We developed FHR5 staining protocols for formalin-fixed paraffin-embedded (FFPE) renal tissue and applied them to surplus biopsy sections from a C3G cohort. RESULTS: Glomerular FHR5 was highly prevalent in native and transplant C3G and correlated with glomerular C3 and C5b-9 staining. Glomerular FHR5 staining correlated negatively with estimated glomerular filtration rate (eGFR) (P = 0.04, difference of medians 19.7 ml/min per 1.73 m2; 95% confidence interval [CI] 1.1-43.0) and positively with a membranoproliferative glomerulonephritis pattern at diagnostic biopsy (odds ratio 18; 95% CI 1.6-201; P = 0.049). Glomerular FHR5 staining intensity positively correlated with glomerular complement C3b/iC3b/C3c (Pearson's correlation coefficient [R] = 0.59; P = 0.0008), C3dg (R = 0.47; P = 0.02) and C5b9 (R = 0.44, P = 0.02). CONCLUSIONS: Glomerular FHR5 is highly prevalent in C3G, interacts with glomerular C3, and is associated with markers of disease severity. Glomerular FHR5 likely exacerbates complement-mediated glomerular damage in C3G and its interaction with glomerular complement might be exploited to target complement therapeutic agents.

2.
Biochem Biophys Res Commun ; 406(3): 471-7, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21334309

RESUMO

The FAM69 family of cysteine-rich type II transmembrane proteins comprises three members in all vertebrates except fish, and orthologues with a conserved structure are present throughout metazoa. All three murine FAM69 proteins (FAM69A, FAM69B, FAM69C) localise to the endoplasmic reticulum (ER) in cultured cells, probably via N-terminal di-arginine motifs. Mammalian FAM69A is ubiquitously expressed, FAM69B is strongly expressed in the brain and in peripheral endothelial cells, and FAM69C in the brain and eye. Antibodies against mouse FAM69B strongly stain the ER of a subset of neurons in the brain. FAM69 proteins are likely to play a fundamental and highly conserved role in the ER of most metazoan cells, with additional specialised roles in the vertebrate nervous system.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Biologia Computacional , Olho/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/classificação , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Filogenia , Alinhamento de Sequência
3.
Proc Natl Acad Sci U S A ; 107(17): 7927-32, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20378838

RESUMO

Huntington's disease (HD), an incurable neurodegenerative disorder, has a complex pathogenesis including protein aggregation and the dysregulation of neuronal transcription and metabolism. Here, we demonstrate that inhibition of sirtuin 2 (SIRT2) achieves neuroprotection in cellular and invertebrate models of HD. Genetic or pharmacologic inhibition of SIRT2 in a striatal neuron model of HD resulted in gene expression changes including significant down-regulation of RNAs responsible for sterol biosynthesis. Whereas mutant huntingtin fragments increased sterols in neuronal cells, SIRT2 inhibition reduced sterol levels via decreased nuclear trafficking of SREBP-2. Importantly, manipulation of sterol biosynthesis at the transcriptional level mimicked SIRT2 inhibition, demonstrating that the metabolic effects of SIRT2 inhibition are sufficient to diminish mutant huntingtin toxicity. These data identify SIRT2 inhibition as a promising avenue for HD therapy and elucidate a unique mechanism of SIRT2-inhibitor-mediated neuroprotection. Furthermore, the ascertainment of SIRT2's role in regulating cellular metabolism demonstrates a central function shared with other sirtuin proteins.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Doença de Huntington/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Sirtuína 2/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Esteróis/biossíntese , Análise de Variância , Animais , Western Blotting , Caenorhabditis elegans , Drosophila , Perfilação da Expressão Gênica , Imuno-Histoquímica , Camundongos , Microscopia Confocal
4.
J Biol Chem ; 285(12): 8808-23, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20086007

RESUMO

Huntingtin proteolysis has been implicated in the molecular pathogenesis of Huntington disease (HD). Despite an intense effort, the identity of the pathogenic smallest N-terminal fragment has not been determined. Using a panel of anti-huntingtin antibodies, we employed an unbiased approach to generate proteolytic cleavage maps of mutant and wild-type huntingtin in the HdhQ150 knock-in mouse model of HD. We identified 14 prominent N-terminal fragments, which, in addition to the full-length protein, can be readily detected in cytoplasmic but not nuclear fractions. These fragments were detected at all ages and are not a consequence of the pathogenic process. We demonstrated that the smallest fragment is an exon 1 huntingtin protein, known to contain a potent nuclear export signal. Prior to the onset of behavioral phenotypes, the exon 1 protein, and possibly other small fragments, accumulate in neuronal nuclei in the form of a detergent insoluble complex, visualized as diffuse granular nuclear staining in tissue sections. This methodology can be used to validate the inhibition of specific proteases as therapeutic targets for HD by pharmacological or genetic approaches.


Assuntos
Doença de Huntington/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Animais , Células COS , Calpaína/química , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Modelos Animais de Doenças , Éxons , Genótipo , Proteína Huntingtina , Camundongos , Estrutura Terciária de Proteína
5.
PLoS One ; 4(11): e8025, 2009 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-19956633

RESUMO

BACKGROUND: Huntington's disease (HD) is an inherited progressive neurodegenerative disorder caused by a CAG repeat expansion in the ubiquitously expressed HD gene resulting in an abnormally long polyglutamine repeat in the huntingtin protein. Polyglutamine inclusions are a hallmark of the neuropathology of HD. We have previously shown that inclusion pathology is also present in the peripheral tissues of the R6/2 mouse model of HD which expresses a small N-terminal fragment of mutant huntingtin. To determine whether this peripheral pathology is a consequence of the aberrant expression of this N-terminal fragment, we extend this analysis to the genetically precise knock-in mouse model of HD, HdhQ150, which expresses mutant mouse huntingtin. METHODOLOGY/PRINCIPAL FINDINGS: We have previously standardized the CAG repeat size and strain background of the R6/2 and HdhQ150 knock-in mouse models and found that they develop a comparable and widespread neuropathology. To determine whether HdhQ150 knock-in mice also develop peripheral inclusion pathology, homozygous Hdh(Q150/Q150) mice were perfusion fixed at 22 months of age, and tissues were processed for histology and immunohistochemistry with the anti-huntingtin antibody S830. The peripheral inclusion pathology was almost identical to that found in R6/2 mice at 12 weeks of age with minor differences in inclusion abundance. CONCLUSIONS/SIGNIFICANCE: The highly comparable peripheral inclusion pathology that is present in both the R6/2 and HdhQ150 knock-in models of HD indicates that the presence of peripheral inclusions in R6/2 mice is not a consequence of the aberrant expression of an N-terminal huntingtin protein. It remains to be determined whether peripheral inclusions are a pathological feature of the human disease. Both mouse models carry CAG repeats that cause childhood disease in humans, and therefore, inclusion pathology may be a feature of the childhood rather than the adult forms of HD. It is important to establish the extent to which peripheral pathology causes the peripheral symptoms of HD from the perspective of a mechanistic understanding and future treatment options.


Assuntos
Doença de Huntington/genética , Peptídeos/metabolismo , Glândulas Suprarrenais/metabolismo , Animais , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Mucosa Gástrica/metabolismo , Proteína Huntingtina , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Músculo Esquelético/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Estrutura Terciária de Proteína
6.
PLoS One ; 4(6): e5747, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19484127

RESUMO

Huntington's disease (HD) is an inherited, progressive neurological disorder caused by a CAG/polyglutamine repeat expansion, for which there is no effective disease modifying therapy. In recent years, transcriptional dysregulation has emerged as a pathogenic process that appears early in disease progression. Administration of histone deacetylase (HDAC) inhibitors such as suberoylanilide hydroxamic acid (SAHA) have consistently shown therapeutic potential in models of HD, at least partly through increasing the association of acetylated histones with down-regulated genes and by correcting mRNA abnormalities. The HDAC enzyme through which SAHA mediates its beneficial effects in the R6/2 mouse model of HD is not known. Therefore, we have embarked on a series of genetic studies to uncover the HDAC target that is relevant to therapeutic development for HD. HDAC7 is of interest in this context because SAHA has been shown to decrease HDAC7 expression in cell culture systems in addition to inhibiting enzyme activity. After confirming that expression levels of Hdac7 are decreased in the brains of wild type and R6/2 mice after SAHA administration, we performed a genetic cross to determine whether genetic reduction of Hdac7 would alleviate phenotypes in the R6/2 mice. We found no improvement in a number of physiological or behavioral phenotypes. Similarly, the dysregulated expression levels of a number of genes of interest were not improved suggesting that reduction in Hdac7 does not alleviate the R6/2 HD-related transcriptional dysregulation. Therefore, we conclude that the beneficial effects of HDAC inhibitors are not predominantly mediated through the inhibition of HDAC7.


Assuntos
Histona Desacetilases/genética , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Genótipo , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Fenótipo , Transcrição Gênica
7.
Proc Natl Acad Sci U S A ; 105(9): 3467-72, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18299573

RESUMO

Huntington's disease (HD) is caused by a CAG repeat expansion that is unstable upon germ-line transmission and exhibits mosaicism in somatic tissues. We show that region-specific CAG repeat mosaicism profiles are conserved between several mouse models of HD and therefore develop in a predetermined manner. Furthermore, we demonstrate that these synchronous, radical changes in CAG repeat size occur in terminally differentiated neurons. In HD this ongoing mutation of the repeat continuously generates genetically distinct neuronal populations in the adult brain of mouse models and HD patients. The neuronal population of the striatum is particularly distinguished by a high rate of CAG repeat allele instability and expression driving the repeat upwards and would be expected to enhance its toxicity. In both mice and humans, neurons are distinguished from nonneuronal cells by expression of MSH3, which provides a permissive environment for genetic instability independent of pathology. The neuronal mutations described here accumulate to generate genetically discrete populations of cells in the absence of selection. This is in contrast to the traditional view in which genetically discrete cellular populations are generated by the sequence of random variation, selection, and clonal proliferation. We are unaware of any previous demonstration that mutations can occur in terminally differentiated neurons and provide a proof of principle that, dependent on a specific set of conditions, functional DNA polymorphisms can be produced in adult neurons.


Assuntos
Doença de Huntington/etiologia , Neurônios/patologia , Expansão das Repetições de Trinucleotídeos , Animais , Encéfalo/patologia , Diferenciação Celular , Humanos , Doença de Huntington/genética , Doença de Huntington/patologia , Camundongos , Mitose , Mosaicismo
8.
Brain Res Bull ; 72(2-3): 83-97, 2007 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-17352931

RESUMO

The identification of the Huntington's disease (HD) mutation as a CAG/polyglutamine repeat expansion enabled the generation of transgenic rodent models and gene-targeted mouse models of HD. Of these, mice that are transgenic for an N-terminal huntingtin fragment have been used most extensively because they develop phenotypes with relatively early ages of onset and rapid disease progression. Although the fragment models have led to novel insights into the pathophysiology of HD, it is important that models expressing a mutant version of the full-length protein are analysed in parallel. We have generated congenic C57BL/6 and CBA strains for the HdhQ150 knock-in mouse model of HD so that homozygotes can be analysed on an F1 hybrid background. Although a significant impairment in grip strength could be detected from a very early age, the performance of these mice in the quantitative behavioural tests most frequently used in preclinical efficacy trials indicates that they are unlikely to be useful for preclinical screening using a battery of conventional tests. However, at 22 months of age, the Hdh(Q150/Q150) homozygotes showed unexpected widespread aggregate deposition throughout the brain, transcriptional dysregulation in the striatum and cerebellum and decreased levels of specific chaperones, all well-characterised molecular phenotypes present in R6/2 mice aged 12 weeks. Therefore, when strain background and CAG repeat length are controlled for, the knock-in and fragment models develop comparable phenotypes. This supports the continued use of the more high-throughput fragment models to identify mechanisms of pathogenesis and for preclinical screening.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Camundongos , Animais , Western Blotting , Éxons , Feminino , Proteína Huntingtina , Imuno-Histoquímica , Masculino , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Expansão das Repetições de Trinucleotídeos
9.
Neurobiol Dis ; 21(1): 228-36, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16111888

RESUMO

Huntington's disease (HD) is an inherited progressive neurological disorder for which there is no effective therapy. It is caused by a CAG/polyglutamine repeat expansion that leads to abnormal protein aggregation and deposition in the brain. Several compounds have been shown to disrupt the aggregation process in vitro, including a number of benzothiazoles. To further explore the therapeutic potential of the benzothiazole aggregation inhibitors, we assessed PGL-135 and riluzole in hippocampal slice cultures derived from the R6/2 mouse, confirming their ability to inhibit aggregation with an EC50 of 40 microM in this system. Preliminary pharmacological work showed that PGL-135 was metabolically unstable, and therefore, we conducted a preclinical trial in the R6/2 mouse with riluzole. At the maximum tolerated dose, we achieved steady-state riluzole levels of 100 microM in brain. However, this was insufficient to inhibit aggregation in vivo and we found no improvement in the disease phenotype.


Assuntos
Doença de Huntington/tratamento farmacológico , Fármacos Neuroprotetores/farmacocinética , Riluzol/farmacocinética , Tiazóis/metabolismo , Tiazóis/farmacologia , Animais , Benzotiazóis , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Genótipo , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Fármacos Neuroprotetores/química , Técnicas de Cultura de Órgãos , Riluzol/química , Tiazóis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...