Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
PLoS One ; 14(3): e0213521, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30913220

RESUMO

Huntington's disease (HD) is a monogenic neurodegenerative disorder caused by an expansion of the CAG trinucleotide repeat domain in the huntingtin (HTT) gene, leading to an expanded poly-glutamine (polyQ) stretch in the HTT protein. This mutant HTT (mHTT) protein is highly prone to intracellular aggregation, causing significant damage and cellular loss in the striatal, cortical, and other regions of the brain. Therefore, modulation of mHTT levels in these brain regions in order to reduce intracellular mHTT and aggregate levels represents a direct approach in the development of HD therapeutics. To this end, assays that can be used to detect changes in HTT levels in biological samples are invaluable tools to assess target engagement and guide dose selection in clinical trials. The Meso Scale Discovery (MSD) ELISA-based assay platform is a robust and sensitive method previously employed for the quantification of HTT. However, the currently available MSD assays for HTT are primarily detecting the monomeric soluble form of the protein, but not aggregated species. In this study, we describe the development of novel MSD assays preferentially detecting mHTT in an aggregated form. Recombinant monomeric HTT(1-97)-Q46, which forms aggregates in a time-dependent manner, was used to characterize the ability of each established assay to distinguish between HTT monomers and HTT in a higher assembly state. Further validation of these assays was performed using brain lysates from R6/2, zQ175 knock-in, and BACHD mouse models, to replicate a previously well-characterized age-dependent increase in brain aggregate signals, as well as a significant reduction of aggregate levels in the striatum following mHTT knockdown with a CAG-directed allele-specific zinc-finger repressor protein (ZFP). Lastly, size exclusion chromatography was used to separate and characterize HTT species from brain tissue lysates to demonstrate specificity of the assays for the fractions containing aggregated HTT. In summary, we demonstrate that the newly developed assays preferentially detect aggregated HTT with improved performance in comparison to previous assay technologies. These assays complement the existing MSD platform assays specific for soluble HTT monomers, allowing for a more comprehensive analysis of disease-relevant HTT species in preclinical models of HD.


Assuntos
Bioensaio , Encéfalo/metabolismo , Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Peptídeos/genética , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia
2.
J Med Chem ; 59(7): 3489-98, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27011007

RESUMO

Herein, we describe the development of a functionally selective liver X receptor ß (LXRß) agonist series optimized for Emax selectivity, solubility, and physical properties to allow efficacy and safety studies in vivo. Compound 9 showed central pharmacodynamic effects in rodent models, evidenced by statistically significant increases in apolipoprotein E (apoE) and ATP-binding cassette transporter levels in the brain, along with a greatly improved peripheral lipid safety profile when compared to those of full dual agonists. These findings were replicated by subchronic dosing studies in non-human primates, where cerebrospinal fluid levels of apoE and amyloid-ß peptides were increased concomitantly with an improved peripheral lipid profile relative to that of nonselective compounds. These results suggest that optimization of LXR agonists for Emax selectivity may have the potential to circumvent the adverse lipid-related effects of hepatic LXR activity.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Apolipoproteínas E/líquido cefalorraquidiano , Benzamidas/química , Benzamidas/farmacologia , Receptores Nucleares Órfãos/agonistas , Piperidinas/química , Piperidinas/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cães , Células Hep G2 , Humanos , Lipídeos/análise , Fígado/efeitos dos fármacos , Fígado/metabolismo , Receptores X do Fígado , Locomoção/efeitos dos fármacos , Macaca mulatta , Células Madin Darby de Rim Canino , Camundongos , Camundongos Transgênicos
3.
PLoS One ; 9(7): e103187, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25051234

RESUMO

Sporadic or late-onset Alzheimer's disease (AD) is expected to affect 50% of individuals reaching 85 years of age. The most significant genetic risk factor for late-onset AD is the e4 allele of APOE gene encoding apolipoprotein E, a lipid carrier shown to modulate brain amyloid burden. Recent genome-wide association studies have uncovered additional single nucleotide polymorphisms (SNPs) linked to AD susceptibility, including those in the CLU and BIN1 genes encoding for clusterin (CLU) and the bridging integrator 1 (BIN1) proteins, respectively. Because CLU has been implicated in brain amyloid-ß (Aß) clearance in mouse models of amyloid deposition, we sought to investigate whether an AD-linked SNP in the CLU gene altered Aß42 biomarker levels in the cerebrospinal fluid (CSF). Instead, we found that the CLU rs11136000 SNP modified CSF levels of the microtubule-associated protein Tau in AD patients. We also found that an intracellular form of CLU (iCLU) was upregulated in the brain of Tau overexpressing Tg4510 mice, but not in Tg2576 amyloid mouse model. By overexpressing iCLU and Tau in cell culture systems we discovered that iCLU was a Tau-interacting protein and that iCLU associated with brain-specific isoforms of BIN1, also recently identified as a Tau-binding protein. Through expression analysis of CLU and BIN1 variants, we found that CLU and BIN1 interacted via their coiled-coil motifs. In co-immunoprecipitation studies using human brain tissue, we showed that iCLU and the major BIN1 isoform expressed in neurons were associated with modified Tau species found in AD. Finally, we showed that expression of certain coding CLU variants linked to AD risk led to increased levels of iCLU. Together, our findings suggest that iCLU and BIN1 interaction might impact Tau function in neurons and uncover potential new mechanisms underlying the etiology of Tau pathology in AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Clusterina/metabolismo , Proteínas Nucleares/metabolismo , Mapas de Interação de Proteínas , Proteínas Supressoras de Tumor/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Células Cultivadas , Clusterina/análise , Humanos , Camundongos , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Proteínas tau/análise
4.
PLoS Biol ; 9(1): e1000575, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21264353

RESUMO

Obesity is a growing epidemic characterized by excess fat storage in adipocytes. Although lipoprotein receptors play important roles in lipid uptake, their role in controlling food intake and obesity is not known. Here we show that the lipoprotein receptor LRP1 regulates leptin signaling and energy homeostasis. Conditional deletion of the Lrp1 gene in the brain resulted in an obese phenotype characterized by increased food intake, decreased energy consumption, and decreased leptin signaling. LRP1 directly binds to leptin and the leptin receptor complex and is required for leptin receptor phosphorylation and Stat3 activation. We further showed that deletion of the Lrp1 gene specifically in the hypothalamus by Cre lentivirus injection is sufficient to trigger accelerated weight gain. Together, our results demonstrate that the lipoprotein receptor LRP1, which is critical in lipid metabolism, also regulates food intake and energy homeostasis in the adult central nervous system.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético , Leptina/fisiologia , Receptores de LDL/deficiência , Proteínas Supressoras de Tumor/deficiência , Proteína Relacionada com Agouti/biossíntese , Proteína Relacionada com Agouti/genética , Animais , Regulação do Apetite , Linhagem Celular , Feminino , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Homeostase , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Hipotálamo/metabolismo , Resistência à Insulina , Metabolismo dos Lipídeos/efeitos dos fármacos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos , Camundongos Knockout , Neuropeptídeo Y/biossíntese , Neuropeptídeo Y/genética , Obesidade/genética , Obesidade/metabolismo , Receptores de LDL/genética , Proteínas Supressoras de Tumor/genética , Regulação para Cima
5.
Neuron ; 56(1): 66-78, 2007 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17920016

RESUMO

Mutations in the amyloid precursor protein (APP) cause early-onset Alzheimer's disease (AD), but the only genetic risk factor for late-onset AD is the varepsilon4 allele of apolipoprotein E (apoE), a major cholesterol carrier. Using Cre-lox conditional knockout mice, we demonstrate that lipoprotein receptor LRP1 expression regulates apoE and cholesterol levels within the CNS. We also found that deletion of APP and its homolog APLP2, or components of the gamma-secretase complex, significantly enhanced the expression and function of LRP1, which was reversed by forced expression of the APP intracellular domain (AICD). We further show that AICD, together with Fe65 and Tip60, interacts with the LRP1 promoter and suppresses its transcription. Together, our findings support that the gamma-secretase cleavage of APP plays a central role in regulating apoE and cholesterol metabolism in the CNS via LRP1 and establish a biological linkage between APP and apoE, the two major genetic determinants of AD.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Colesterol/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de LDL/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/deficiência , Animais , Animais Recém-Nascidos , Linhagem Celular Transformada , Imunoprecipitação da Cromatina , Cricetinae , Citidina Desaminase/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica no Desenvolvimento/genética , Histona Acetiltransferases/farmacologia , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Lisina Acetiltransferase 5 , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/farmacologia , Proteínas Nucleares/farmacologia , RNA Mensageiro/biossíntese , Receptores de LDL/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transativadores , Transfecção/métodos , Proteínas Supressoras de Tumor/deficiência
6.
Ann N Y Acad Sci ; 1086: 35-53, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17185504

RESUMO

Amyloid-beta peptide (Abeta) production and accumulation in the brain is a central event in the pathogenesis of Alzheimer's disease (AD). Recent studies have shown that apolipoprotein E (apoE) receptors, members of the low-density lipoprotein receptor (LDLR) family, modulate Abeta production as well as Abeta cellular uptake. Abeta is derived from proteolytic processing of the amyloid precursor protein (APP), which interacts with several members of the LDLR family. Studies from our laboratory have focused on two members of the LDLR family, the LDLR-related protein (LRP) and LRP1B. Our in vitro studies have shown that while LRP's rapid endocytosis facilitates APP endocytic trafficking and processing to Abeta, LRP1B's slow endocytosis inhibits these processes. In addition to modulating APP endocytic trafficking, LRP's rapid endocytosis also facilitates Abeta cellular uptake by binding to Abeta either directly or via LRP ligands such as apoE. Our in vivo studies using transgenic mice have shown that overexpression of LRP in central nervous system (CNS) neurons increases soluble brain Abeta and this increase correlates with deficits in memory. Together our studies demonstrate that members of the LDLR family modulate APP processing and Abeta metabolism by several independent mechanisms. Understanding the pathways that modulate brain Abeta metabolism may enable the rational design of molecular medicine to treat AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Endocitose/fisiologia , Endossomos/metabolismo , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Receptores de LDL/genética , Proteínas Supressoras de Tumor/genética
7.
J Biol Chem ; 281(47): 36180-6, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17012232

RESUMO

The low density lipoprotein receptor-related protein (LRP) is highly expressed in the brain and has been shown to alter the metabolism of amyloid precursor protein and amyloid-beta peptide (Abeta) in vitro. Previously we developed mice that overexpress a functional LRP minireceptor (mLRP2) in their brains and crossed them to the PDAPP mouse model of Alzheimer disease. Overexpression of mLRP2 in 22-month-old PDAPP mice with amyloid plaques increased a pool of carbonate-soluble Abeta in the brain and worsened memory-related behavior. In the current study, we examined the effects of mLRP2 overexpression on 3-month-old PDAPP mice that had not yet developed amyloid plaques. We found significantly higher levels of membrane-associated Abeta42 in the hippocampus of mice that overexpressed mLRP2. Using immunohistochemical methods, we observed significant intraneuronal Abeta42 in the hippocampus and frontal cortex of PDAPP mice, which frequently co-localized with the lysosomal marker LAMP-1. Interestingly, PDAPP mice lacking apolipoprotein E (apoE) had much less intraneuronal Abeta42. We also found that PC12 cells overexpressing mLRP2 cleared Abeta42 and Abeta40 more rapidly from media than PC12 cells transfected with the vector only. Preincubation of apoE3 or apoE4 with Abeta42 increased the rate of Abeta clearance, and this effect was partially blocked by receptor-associated protein. Our results support the hypothesis that LRP binds and endocytoses Abeta42 both directly and via apoE but that endocytosed Abeta42 is not completely degraded and accumulates in intraneuronal lysosomes.


Assuntos
Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/fisiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Neurônios/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Animais , Encéfalo/metabolismo , Endocitose , Hipocampo/metabolismo , Humanos , Imuno-Histoquímica , Lisossomos/metabolismo , Camundongos , Camundongos Transgênicos , Células PC12 , Ratos
8.
J Biol Chem ; 279(28): 29639-46, 2004 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-15126508

RESUMO

The low density lipoprotein (LDL) receptor-related protein 1B (LRP1B) is a newly identified member of the LDL receptor family that shares high homology with the LDL receptor-related protein (LRP). LRP1B was originally described as a putative tumor suppressor in lung cancer cells; however, its expression profile in several regions of adult human brain suggests it may have additional functions in the central nervous system. Since LRP1B has overlapping ligand binding properties with LRP, we investigated whether LRP1B, like LRP, could interact with the beta-amyloid precursor protein (APP) and modulate its processing to amyloid-beta peptides (Abetas). Using an LRP1B minireceptor (mLRP1B4) generated to study the trafficking of LRP1B, we found that mLRP1B4 and APP form an immunoprecipitable complex. Furthermore mLRP1B4 bound and facilitated the degradation of a soluble isoform of APP containing a Kunitz proteinase inhibitor domain but not soluble APP lacking a Kunitz proteinase inhibitor domain. A functional consequence of mLRP1B4 expression was a significant accumulation of APP at the cell surface, which is likely related to the slow endocytosis rate of LRP1B. More importantly, mLRP1B4-expressing cells that accumulated cell surface APP produced less Abeta and secreted more soluble APP. These findings reveal that LRP1B is a novel binding partner of APP that functions to decrease APP processing to Abeta. Consequently LRP1B expression could function to protect against the pathogenesis of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Receptores de LDL/metabolismo , Adulto , Animais , Encéfalo/metabolismo , Células CHO , Membrana Celular/química , Membrana Celular/metabolismo , Cricetinae , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Camundongos , Estrutura Terciária de Proteína , Receptores de LDL/genética
9.
Am J Physiol Cell Physiol ; 284(2): C555-61, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12388112

RESUMO

It is postulated that macrophage-derived foam cells accumulate in the arterial wall because they lose the ability to migrate after excessive ingestion of modified forms of low-density lipoproteins (LDL). To assess changes in locomotor force generating capacity of foam cells, we measured isometric forces in J774A.1 macrophages after cholesterol loading with oxidized (Ox-LDL) or aggregated (Agg-LDL) LDL using a novel magnetic force transducer. Ox-LDL loading reduced the ability of J774A.1 macrophages to generate isometric forces by 50% relative to control cells. Changes in force frequency consistent with reduced motility were detected as well. Agg-LDL loading was also detrimental to J774A.1 motility but to a lesser extent than Ox-LDL. Ox-LDL loading significantly reduced total actin levels and induced changes in the F-actin to G-actin distribution, whereas Agg-LDL loaded cells had significantly increased levels of total actin. These data provide evidence that cholesterol loading and subsequent accumulation decreases macrophage motility by reducing the cells' force generating capacity and that Ox-LDL appears to be more effective than Agg-LDL in disrupting the locomotor machinery.


Assuntos
Actinas/metabolismo , Arteriosclerose/metabolismo , Movimento Celular/fisiologia , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Actinas/efeitos dos fármacos , Animais , Arteriosclerose/fisiopatologia , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Colesterol/metabolismo , Células Espumosas/citologia , Células Espumosas/metabolismo , Humanos , Contração Isométrica/efeitos dos fármacos , Contração Isométrica/fisiologia , Lipoproteínas LDL/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Microscopia Confocal , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Estresse Mecânico
10.
Arq. bras. cardiol ; 52(5): 291-295, maio 1989. tab
Artigo em Português | LILACS | ID: lil-87315

RESUMO

Lavastatina, um inibidor competitivo da HMGCoA redutase, foi utilizada no tratamento de 26 pacientes portadores de hipercolesterolemia primária, após um período inicial de placebo de 4 semanas. A resposta terapêutica foi analisada durante 11 semanas. As reduçöes do colesteroltotal com 20 e 40 mg/dia foram 17% e 31% e de LDL-C de 24% e 41%, respectivamente. Cinco pacientes mantiveram a dose de 20 mg/dia durante as 12 semanas com reduçöes mais significativas na semana 12 em relaçäo à semana 6. Näo se observou alteraçäo dos níveis de triglicérides, HDL-C e VLDL-C. Comparando-se os dois grupos, isto é, hipercolesterolemia familiar e poligênica, a resposta à droga foi semelhante. Näo ocorreram alteraçöes clínicas ou efeitos colaterais significantes durante o período analisado


Assuntos
Humanos , Masculino , Feminino , Adolescente , Adulto , Pessoa de Meia-Idade , Lovastatina/uso terapêutico , Hipercolesterolemia/tratamento farmacológico , Lovastatina/administração & dosagem , Colesterol/biossíntese , Colesterol/sangue , Ensaios Clínicos como Assunto , LDL-Colesterol/sangue , Química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA