Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 18(7): e1010302, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35853002

RESUMO

Perturbation of huntingtin (HTT)'s physiological function is one postulated pathogenic factor in Huntington's disease (HD). However, little is known how HTT is regulated in vivo. In a proteomic study, we isolated a novel ~40kDa protein as a strong binding partner of Drosophila HTT and demonstrated it was the functional ortholog of HAP40, an HTT associated protein shown recently to modulate HTT's conformation but with unclear physiological and pathologic roles. We showed that in both flies and human cells, HAP40 maintained conserved physical and functional interactions with HTT. Additionally, loss of HAP40 resulted in similar phenotypes as HTT knockout. More strikingly, HAP40 strongly affected HTT's stability, as depletion of HAP40 significantly reduced the levels of endogenous HTT protein while HAP40 overexpression markedly extended its half-life. Conversely, in the absence of HTT, the majority of HAP40 protein were degraded, likely through the proteasome. Further, the affinity between HTT and HAP40 was not significantly affected by polyglutamine expansion in HTT, and contrary to an early report, there were no abnormal accumulations of endogenous HAP40 protein in HD cells from mouse HD models or human patients. Lastly, when tested in Drosophila models of HD, HAP40 partially modulated the neurodegeneration induced by full-length mutant HTT while showed no apparent effect on the toxicity of mutant HTT exon 1 fragment. Together, our study uncovers a conserved mechanism governing the stability and in vivo functions of HTT and demonstrates that HAP40 is a central and positive regulator of endogenous HTT. Further, our results support that mutant HTT is toxic regardless of the presence of its partner HAP40, and implicate HAP40 as a potential modulator of HD pathogenesis through its multiplex effect on HTT's function, stability and the potency of mutant HTT's toxicity.


Assuntos
Proteína Huntingtina , Doença de Huntington , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares , Animais , Modelos Animais de Doenças , Drosophila/genética , Drosophila/metabolismo , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Proteômica
2.
J Med Chem ; 66(18): 13205-13246, 2023 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-37712656

RESUMO

Huntington's disease (HD) is caused by an expanded CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. We report the design of a series of HTT pre-mRNA splicing modulators that lower huntingtin (HTT) protein, including the toxic mutant huntingtin (mHTT), by promoting insertion of a pseudoexon containing a premature termination codon at the exon 49-50 junction. The resulting transcript undergoes nonsense-mediated decay, leading to a reduction of HTT mRNA transcripts and protein levels. The starting benzamide core was modified to pyrazine amide and further optimized to give a potent, CNS-penetrant, and orally bioavailable HTT-splicing modulator 27. This compound reduced canonical splicing of the HTT RNA exon 49-50 and demonstrated significant HTT-lowering in both human HD stem cells and mouse BACHD models. Compound 27 is a structurally diverse HTT-splicing modulator that may help understand the mechanism of adverse effects such as peripheral neuropathy associated with branaplam.

3.
PLoS One ; 17(4): e0266812, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35395060

RESUMO

Huntington's disease (HD) is caused by an expansion of the CAG trinucleotide repeat domain in the huntingtin gene that results in expression of a mutant huntingtin protein (mHTT) containing an expanded polyglutamine tract in the amino terminus. A number of therapeutic approaches that aim to reduce mHTT expression either locally in the CNS or systemically are in clinical development. We have previously described sensitive and selective assays that measure human HTT proteins either in a polyglutamine-independent (detecting both mutant expanded and non-expanded proteins) or in a polyglutamine length-dependent manner (detecting the disease-causing polyglutamine repeats) on the electrochemiluminescence Meso Scale Discovery detection platform. These original assays relied upon polyclonal antibodies. To ensure an accessible and sustainable resource for the HD field, we developed similar assays employing monoclonal antibodies. We demonstrate that these assays have equivalent sensitivity compared to our previous assays through the evaluation of cellular and animal model systems, as well as HD patient biosamples. We also demonstrate cross-site validation of these assays, allowing direct comparison of studies performed in geographically distinct laboratories.


Assuntos
Doença de Huntington , Animais , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Expansão das Repetições de Trinucleotídeos
4.
J Med Chem ; 65(18): 12445-12459, 2022 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-36098485

RESUMO

Huntington's disease (HD) is a lethal autosomal dominant neurodegenerative disorder resulting from a CAG repeat expansion in the huntingtin (HTT) gene. The product of translation of this gene is a highly aggregation-prone protein containing a polyglutamine tract >35 repeats (mHTT) that has been shown to colocalize with histone deacetylase 4 (HDAC4) in cytoplasmic inclusions in HD mouse models. Genetic reduction of HDAC4 in an HD mouse model resulted in delayed aggregation of mHTT, along with amelioration of neurological phenotypes and extended lifespan. To further investigate the role of HDAC4 in cellular models of HD, we have developed bifunctional degraders of the protein and report the first potent and selective degraders of HDAC4 that show an effect in multiple cell lines, including HD mouse model-derived cortical neurons. These degraders act via the ubiquitin-proteasomal pathway and selectively degrade HDAC4 over other class IIa HDAC isoforms (HDAC5, HDAC7, and HDAC9).


Assuntos
Histona Desacetilases , Doença de Huntington , Animais , Modelos Animais de Doenças , Desenvolvimento de Medicamentos , Histona Desacetilases/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Camundongos , Neurônios/metabolismo , Proteólise , Ubiquitinas
5.
J Med Chem ; 65(14): 9819-9845, 2022 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-35816678

RESUMO

The Rho kinase (ROCK) pathway is implicated in the pathogenesis of several conditions, including neurological diseases. In Huntington's disease (HD), ROCK is implicated in mutant huntingtin (HTT) aggregation and neurotoxicity, and members of the ROCK pathway are increased in HD mouse models and patients. To validate this mode of action as a potential treatment for HD, we sought a potent, selective, central nervous system (CNS)-penetrant ROCK inhibitor. Identifying a compound that could be dosed orally in mice with selectivity against other AGC kinases, including protein kinase G (PKG), whose inhibition could potentially activate the ROCK pathway, was paramount for the program. We describe the optimization of published ligands to identify a novel series of ROCK inhibitors based on a piperazine core. Morphing of the early series developed in-house by scaffold hopping enabled the identification of a compound exhibiting high potency and desired selectivity and demonstrating a robust pharmacodynamic (PD) effect by the inhibition of ROCK-mediated substrate (MYPT1) phosphorylation after oral dosing.


Assuntos
Doença de Huntington , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Proteína Huntingtina/metabolismo , Doença de Huntington/tratamento farmacológico , Camundongos , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinases Associadas a rho
6.
ACS Med Chem Lett ; 12(3): 380-388, 2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33738065

RESUMO

Using an iterative structure-activity relationship driven approach, we identified a CNS-penetrant 5-(trifluoromethyl)-1,2,4-oxadiazole (TFMO, 12) with a pharmacokinetic profile suitable for probing class IIa histone deacetylase (HDAC) inhibition in vivo. Given the lack of understanding of endogenous class IIa HDAC substrates, we developed a surrogate readout to measure compound effects in vivo, by exploiting the >100-fold selectivity compound 12 exhibits over class I/IIb HDACs. We achieved adequate brain exposure with compound 12 in mice to estimate a class I/IIb deacetylation EC50, using class I substrate H4K12 acetylation and global acetylation levels as a pharmacodynamic readout. We observed excellent correlation between the compound 12 in vivo pharmacodynamic response and in vitro class I/IIb cellular activity. Applying the same relationship to class IIa HDAC inhibition, we estimated the compound 12 dose required to inhibit class IIa HDAC activity, for use in preclinical models of Huntington's disease.

7.
J Med Chem ; 64(8): 5018-5036, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33783225

RESUMO

Our group has recently shown that brain-penetrant ataxia telangiectasia-mutated (ATM) kinase inhibitors may have potential as novel therapeutics for the treatment of Huntington's disease (HD). However, the previously described pyranone-thioxanthenes (e.g., 4) failed to afford selectivity over a vacuolar protein sorting 34 (Vps34) kinase, an important kinase involved with autophagy. Given that impaired autophagy has been proposed as a pathogenic mechanism of neurodegenerative diseases such as HD, achieving selectivity over Vps34 became an important objective for our program. Here, we report the successful selectivity optimization of ATM over Vps34 by using X-ray crystal structures of a Vps34-ATM protein chimera where the Vps34 ATP-binding site was mutated to approximate that of an ATM kinase. The morpholino-pyridone and morpholino-pyrimidinone series that resulted as a consequence of this selectivity optimization process have high ATM potency and good oral bioavailability and have lower molecular weight, reduced lipophilicity, higher aqueous solubility, and greater synthetic tractability compared to the pyranone-thioxanthenes.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Piridonas/química , Pirimidinonas/química , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sítios de Ligação , Encéfalo/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Cristalografia por Raios X , Desenho de Fármacos , Meia-Vida , Humanos , Doença de Huntington/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Dinâmica Molecular , Morfolinos/química , Piridonas/metabolismo , Piridonas/uso terapêutico , Pirimidinonas/metabolismo , Pirimidinonas/uso terapêutico , Relação Estrutura-Atividade
8.
J Med Chem ; 62(6): 2988-3008, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30840447

RESUMO

Genetic and pharmacological evidence indicates that the reduction of ataxia telangiectasia-mutated (ATM) kinase activity can ameliorate mutant huntingtin (mHTT) toxicity in cellular and animal models of Huntington's disease (HD), suggesting that selective inhibition of ATM could provide a novel clinical intervention to treat HD. Here, we describe the development and characterization of ATM inhibitor molecules to enable in vivo proof-of-concept studies in HD animal models. Starting from previously reported ATM inhibitors, we aimed with few modifications to increase brain exposure by decreasing P-glycoprotein liability while maintaining potency and selectivity. Here, we report brain-penetrant ATM inhibitors that have robust pharmacodynamic (PD) effects consistent with ATM kinase inhibition in the mouse brain and an understandable pharmacokinetic/PD (PK/PD) relationship. Compound 17 engages ATM kinase and shows robust dose-dependent inhibition of X-ray irradiation-induced KAP1 phosphorylation in the mouse brain. Furthermore, compound 17 protects against mHTT (Q73)-induced cytotoxicity in a cortical-striatal cell model of HD.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Doença de Huntington/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Modelos Animais de Doenças , Cães , Humanos , Células Madin Darby de Rim Canino , Camundongos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacocinética , Estudo de Prova de Conceito
9.
Neuropsychopharmacology ; 33(3): 685-700, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17487225

RESUMO

The receptor tyrosine kinase product of the anaplastic lymphoma kinase (ALK) gene has been implicated in oncogenesis as a product of several chromosomal translocations, although its endogeneous role in the hematopoietic and neural systems has remained poorly understood. We describe that the generation of animals homozygous for a deletion of the ALK tyrosine kinase domain leads to alterations in adult brain function. Evaluation of adult ALK homozygotes (HOs) revealed an age-dependent increase in basal hippocampal progenitor proliferation and alterations in behavioral tests consistent with a role for this receptor in the adult brain. ALK HO animals displayed an increased struggle time in the tail suspension test and the Porsolt swim test and enhanced performance in a novel object-recognition test. Neurochemical analysis demonstrates an increase in basal dopaminergic signalling selectively within the frontal cortex. Altogether, these results suggest that ALK functions in the adult brain to regulate the function of the frontal cortex and hippocampus and identifies ALK as a new target for psychiatric indications, such as schizophrenia and depression, with an underlying deregulated monoaminergic signalling.


Assuntos
Comportamento Animal/fisiologia , Química Encefálica/fisiologia , Proteínas Tirosina Quinases/genética , Quinase do Linfoma Anaplásico , Animais , Ansiedade/genética , Ansiedade/psicologia , Química Encefálica/genética , Bromodesoxiuridina , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Depressão/genética , Depressão/psicologia , Dopamina/metabolismo , Feminino , Citometria de Fluxo , Elevação dos Membros Posteriores , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Atividade Motora , Receptores Proteína Tirosina Quinases , Reconhecimento Psicológico/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serotonina/metabolismo , Natação/psicologia , Timidina/análogos & derivados , Timidina/farmacologia
10.
Stem Cells Dev ; 16(1): 143-65, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17233554

RESUMO

The use of neural precursor cells (NPCs) represents a promising repair strategy for many neurological disorders. However, the molecular events and biological features that control NPC proliferation and their differentiation into neurons, astrocytes, and oligodendrocytes are unclear. In the present study, we used a comparative proteomics approach to identify proteins that were differentially regulated in NPCs after short-term differentiation. We also used a subcellular fractionation technique for enrichment of nuclei and other dense organelles to identify proteins that were not readily detected in whole cell extracts. In total, 115 distinct proteins underwent expression changes during NPC differentiation. Forty one of these were only identified following subcellular fractionation. These included transcription factors, RNA-processing factors, cell cycle proteins, and proteins that translocate between the nucleus and cytoplasm. Biological network analysis showed that the differentiation of NPCs was associated with significant changes in cell cycle and protein synthesis machinery. Further characterization of these proteins could provide greater insight into the mechanisms involved in regulation of neurogenesis in the adult central nervous system (CNS) and potentially identify points of therapeutic intervention.


Assuntos
Células-Tronco Adultas/citologia , Ventrículos Laterais/citologia , Células-Tronco Multipotentes/citologia , Neurônios/citologia , Proteômica , Células-Tronco Adultas/metabolismo , Animais , Western Blotting , Técnicas de Cultura de Células , Ciclo Celular , Diferenciação Celular , Eletroforese em Gel Bidimensional/métodos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ventrículos Laterais/metabolismo , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Mapeamento de Peptídeos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
11.
J Neurosci Methods ; 157(1): 54-63, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16876875

RESUMO

Neural stem cells reside in the subventricular zone and the dentate gyrus of the hippocampus in adult mammalian brain. In the hippocampus, a number of factors are reported to modulate the rate of neural progenitor proliferation in the hippocampus, such as exercise, corticosteroids, and many pharmacological agents including several classes of antidepressants. It is currently unclear whether this increased proliferation is physiologically relevant, but it provides a potentially useful biomarker to assess novel antidepressant compounds. Changes in neurogenesis are typically quantified by administration of bromodeoxyuridine (BrdU) in vivo, and subsequent quantification of labelled nuclei. A robust and rapid means of quantifying BrdU labelling in adult hippocampus in vivo would allow higher throughput screening of potential antidepressant compounds. In this study we describe a FACS-based method for quantification of BrdU labelled cells in fixed cell suspensions from BrdU-treated adult mouse hippocampus. A variety of experimental conditions known to modulate proliferation were tested, including administration of corticosterone and the antidepressants imipramine and fluoxetine. The robust changes compared to control groups observed in these models were similar to previously reported studies, thus offering a more rapid and streamlined means to quantify effects of compounds on hippocampal proliferation.


Assuntos
Citometria de Fluxo/métodos , Hipocampo/citologia , Imuno-Histoquímica/métodos , Neurônios/fisiologia , Organogênese/fisiologia , Animais , Bromodesoxiuridina/administração & dosagem , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Dactinomicina/administração & dosagem , Dactinomicina/análogos & derivados , Relação Dose-Resposta a Droga , Camundongos , Reprodutibilidade dos Testes
12.
ACS Med Chem Lett ; 7(1): 34-9, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26819662

RESUMO

Potent and selective class IIa HDAC tetrasubstituted cyclopropane hydroxamic acid inhibitors were identified with high oral bioavailability that exhibited good brain and muscle exposure. Compound 14 displayed suitable properties for assessment of the impact of class IIa HDAC catalytic site inhibition in preclinical disease models.

13.
Assay Drug Dev Technol ; 1(1 Pt 1): 31-40, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15090154

RESUMO

We have developed an assay system suitable for assessment of compound action on the Edg4 subtype of the widely expressed lysophosphatidic acid (LPA)-responsive Edg receptor family. Edg4 was stably overexpressed in the rat hepatoma cell line Rh 7777, and a Ca(2+)-based FLIPR assay developed for measurement of functional responses. In order to investigate the mechanisms linking Edg4 activation to cytosolic Ca(2+) elevation, we have also studied LPA signalling in a human neuroblastoma cell line that endogenously expresses Edg4. LPA responses displayed similar kinetics and potency in the two cell lines. The Ca(2+) signal generated by activation of LPA-sensitive receptors in these cells is mediated primarily by endoplasmic reticulum. However, there is a substantial inhibition of the LPA response by FCCP, indicating that mitochondria also play a key role in the LPA response. Partial inhibition of the response by cyclosporin A could indicate an active Ca(2+) release role for mitochondria in the LPA response. The inositol 1,4,5-triphosphate receptor antagonist 2-aminoethyl diphenyl borate markedly inhibits, but does not abolish, the Ca(2+) response to LPA, suggesting further complexity to the signalling pathways activated by Edg receptors. In comparing Edg signalling in recombinant and native cells, there is a striking overall similarity in receptor expression pattern, agonist potency, and the effect of modulators on the Ca(2+) response. This indicates that the Edg4-overexpressing Rh7777 cell line is a very useful model system for studying receptor pharmacology and signalling mechanisms, and for investigating the Edg4 receptor's downstream effects.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Receptores Acoplados a Proteínas G/fisiologia , Cálcio/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Corantes , Retículo Endoplasmático/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Imuno-Histoquímica , Inositol 1,4,5-Trifosfato/farmacologia , Neoplasias Hepáticas/metabolismo , Lisofosfolipídeos/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores de Ácidos Lisofosfatídicos , Proteínas Recombinantes , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia
14.
Assay Drug Dev Technol ; 1(2): 239-49, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15090189

RESUMO

The superfamily of GPCRs have diverse biological roles, transducing signals from a range of stimuli, from photon recognition by opsins to neurotransmitter regulation of neuronal function. Of the many identified genes encoding GPCRs, >130 are orphan receptors ( i.e., their endogenous ligands are unknown), and this subset represents putative novel therapeutic targets for pharmaceutical intervention in a variety of diseases. As an initial step toward drug discovery, determining a biological function for these newly identified receptors is of vital importance, and thus identification of a natural ligand(s) is a primary aim. There are several established methods for doing this, but many have drawbacks and usually require some in-depth knowledge about how the receptor functions. The technique described here utilizes a transcription-based reporter assay in live cells. This allows the determination of the signal transduction pathway any given oGPCR uses, without any prior knowledge of the endogenous ligand. This can therefore reduce the redundancy of effort involved in screening ligands at a given receptor in multiple formats (i.e., Galpha(s), Galpha(i/0), and Galpha(q) assays), as well as ensuring that the receptor targeted is capable of signaling if appropriately activated. Such knowledge is often laboriously obtained, and for almost all oGPCRs, this kind of information is not yet available. This technology can also be used to develop inverse agonist as well as agonist sensitive high throughput assays for oGPCRs. The veracity of this approach is demonstrated, using a number of known GPCRs. The likely signaling pathways of the GPR3, GPR12, GPR19, GPR21, and HG55 oGPCRs are shown, and a high throughput assay for GPR26 receptors developed. The methods outlined here for elucidation of the signal transduction pathways for oGPCRs and development of functional assays should speed up the process of identification of ligands for this potentially therapeutically useful group of receptors.


Assuntos
Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Sinalização do Cálcio/fisiologia , Linhagem Celular , Cricetinae , Ciclofosfamida , Doxorrubicina , Indústria Farmacêutica/métodos , Fluorescência , Genes Reporter , Proteínas de Fluorescência Verde , Rim/citologia , Proteínas Luminescentes , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso , Receptor 5-HT2A de Serotonina/fisiologia , Receptores de Dopamina D2 , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Leucotrienos/metabolismo , Receptores de Neurotransmissores/metabolismo , Receptores de Serotonina/fisiologia , Transcrição Gênica/fisiologia , Transfecção/métodos , Vincristina
15.
Brain Res ; 1002(1-2): 1-10, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-14988027

RESUMO

Antidepressants are widely prescribed in the treatment of depression, although the mechanism of how they exert their therapeutic effects is poorly understood. To shed further light on their mode of action, we have attempted to identify a common proteomic signature in guinea pig brains after chronic treatment with two different antidepressants. Both fluoxetine and the substance P receptor (NK(1)R) antagonist (SPA) L-000760735 altered cortical expression of multiple heat shock protein 60 forms along with neurofilaments and related proteins that are critical determinants of synaptic structure and function. Analysis of NK(1)R-/- mice showed similar alterations of neurofilaments confirming the specificity of the effects observed with chronic NK(1)R antagonist treatment. To determine if these changes were associated with structural modification of synapses, we carried out electron microscopic analysis of cerebral cortices from fluoxetine-treated guinea pigs. This showed an increase in the percentage of synapses with split postsynaptic densities (PSDs), a phenomenon that is characteristic of activity-dependent synaptic rearrangement. These findings suggest that cortical alterations of the neurofilament pathway and increased synaptic remodeling are associated with the mechanism of these two antidepressant drug treatments and may contribute to their psychotherapeutic actions.


Assuntos
Antidepressivos/farmacologia , Fluoxetina/farmacologia , Proteínas de Neurofilamentos/ultraestrutura , Antagonistas dos Receptores de Neurocinina-1 , Sinapses/efeitos dos fármacos , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/ultraestrutura , Cobaias , Proteínas de Choque Térmico/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neurofilamentos/biossíntese , Receptores da Neurocinina-1/biossíntese , Receptores da Neurocinina-1/deficiência , Sinapses/metabolismo , Sinapses/ultraestrutura
16.
PLoS One ; 9(5): e96854, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24816435

RESUMO

The expansion of a CAG trinucleotide repeat in the huntingtin gene, which produces huntingtin protein with an expanded polyglutamine tract, is the cause of Huntington's disease (HD). Recent studies have reported that RNAi suppression of polyglutamine-expanded huntingtin (mutant HTT) in HD animal models can ameliorate disease phenotypes. A key requirement for such preclinical studies, as well as eventual clinical trials, aimed to reduce mutant HTT exposure is a robust method to measure HTT protein levels in select tissues. We have developed several sensitive and selective assays that measure either total human HTT or polyglutamine-expanded human HTT proteins on the electrochemiluminescence Meso Scale Discovery detection platform with an increased dynamic range over other methods. In addition, we have developed an assay to detect endogenous mouse and rat HTT proteins in pre-clinical models of HD to monitor effects on the wild type protein of both allele selective and non-selective interventions. We demonstrate the application of these assays to measure HTT protein in several HD in vitro cellular and in vivo animal model systems as well as in HD patient biosamples. Furthermore, we used purified recombinant HTT proteins as standards to quantitate the absolute amount of HTT protein in such biosamples.


Assuntos
Bioensaio/métodos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Encéfalo/metabolismo , Linhagem Celular , Feminino , Humanos , Proteína Huntingtina , Medições Luminescentes , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/imunologia , Proteínas Nucleares/química , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Ratos , Solubilidade
17.
PLoS One ; 9(2): e87923, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24503862

RESUMO

Huntington's disease (HD) is a devastating, genetic neurodegenerative disease caused by a tri-nucleotide expansion in exon 1 of the huntingtin gene. HD is clinically characterized by chorea, emotional and psychiatric disturbances and cognitive deficits with later symptoms including rigidity and dementia. Pathologically, the cortico-striatal pathway is severely dysfunctional as reflected by striatal and cortical atrophy in late-stage disease. Brain-derived neurotrophic factor (BDNF) is a neuroprotective, secreted protein that binds with high affinity to the extracellular domain of the tropomyosin-receptor kinase B (TrkB) receptor promoting neuronal cell survival by activating the receptor and down-stream signaling proteins. Reduced cortical BDNF production and transport to the striatum have been implicated in HD pathogenesis; the ability to enhance TrkB signaling using a BDNF mimetic might be beneficial in disease progression, so we explored this as a therapeutic strategy for HD. Using recombinant and native assay formats, we report here the evaluation of TrkB antibodies and a panel of reported small molecule TrkB agonists, and identify the best candidate, from those tested, for in vivo proof of concept studies in transgenic HD models.


Assuntos
Anticorpos Monoclonais/farmacologia , Doença de Huntington/metabolismo , Receptor trkB/agonistas , Receptor trkB/metabolismo , Animais , Anticorpos Monoclonais/química , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Doença de Huntington/tratamento farmacológico , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos
18.
J Med Chem ; 56(24): 9934-54, 2013 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-24261862

RESUMO

Inhibition of class IIa histone deacetylase (HDAC) enzymes have been suggested as a therapeutic strategy for a number of diseases, including Huntington's disease. Catalytic-site small molecule inhibitors of the class IIa HDAC4, -5, -7, and -9 were developed. These trisubstituted diarylcyclopropanehydroxamic acids were designed to exploit a lower pocket that is characteristic for the class IIa HDACs, not present in other HDAC classes. Selected inhibitors were cocrystallized with the catalytic domain of human HDAC4. We describe the first HDAC4 catalytic domain crystal structure in a "closed-loop" form, which in our view represents the biologically relevant conformation. We have demonstrated that these molecules can differentiate class IIa HDACs from class I and class IIb subtypes. They exhibited pharmacokinetic properties that should enable the assessment of their therapeutic benefit in both peripheral and CNS disorders. These selective inhibitors provide a means for evaluating potential efficacy in preclinical models in vivo.


Assuntos
Desenho de Fármacos , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Doença de Huntington/tratamento farmacológico , Animais , Relação Dose-Resposta a Droga , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacocinética , Histona Desacetilases/classificação , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/química , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
19.
PLoS One ; 7(9): e44498, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22973455

RESUMO

Histone deacetylase (HDAC) inhibitors have received considerable attention as potential therapeutics for a variety of cancers and neurological disorders. Recent publications on a class of pimelic diphenylamide HDAC inhibitors have highlighted their promise in the treatment of the neurodegenerative diseases Friedreich's ataxia and Huntington's disease, based on efficacy in cell and mouse models. These studies' authors have proposed that the unique action of these compounds compared to hydroxamic acid-based HDAC inhibitors results from their unusual slow-on/slow-off kinetics of binding, preferentially to HDAC3, resulting in a distinctive pharmacological profile and reduced toxicity. Here, we evaluate the HDAC subtype selectivity, cellular activity, absorption, distribution, metabolism and excretion (ADME) properties, as well as the central pharmacodynamic profile of one such compound, HDACi 4b, previously described to show efficacy in vivo in the R6/2 mouse model of Huntington's disease. Based on our data reported here, we conclude that while the in vitro selectivity and binding mode are largely in agreement with previous reports, the physicochemical properties, metabolic and p-glycoprotein (Pgp) substrate liability of HDACi 4b render this compound suboptimal to investigate central Class I HDAC inhibition in vivo in mouse per oral administration. A drug administration regimen using HDACi 4b dissolved in drinking water was used in the previous proof of concept study, casting doubt on the validation of CNS HDAC3 inhibition as a target for the treatment of Huntington's disease. We highlight physicochemical stability and metabolic issues with 4b that are likely intrinsic liabilities of the benzamide chemotype in general.


Assuntos
Sistema Nervoso Central/metabolismo , Ataxia de Friedreich/tratamento farmacológico , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Doença de Huntington/tratamento farmacológico , Ácidos Pimélicos/farmacologia , Administração Oral , Animais , Células CACO-2 , Cromatografia Líquida de Alta Pressão , Cães , Ataxia de Friedreich/enzimologia , Inibidores de Histona Desacetilases/administração & dosagem , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/farmacocinética , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Doença de Huntington/enzimologia , Células Madin Darby de Rim Canino , Camundongos , Microssomos Hepáticos/metabolismo , Ácidos Pimélicos/administração & dosagem , Ácidos Pimélicos/síntese química , Ácidos Pimélicos/farmacocinética , Ácidos Pimélicos/uso terapêutico , Espectrometria de Massas em Tandem
20.
Biomark Insights ; 5: 39-47, 2010 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-20520744

RESUMO

We describe the validation of a serum-based test developed by Rules-Based Medicine which can be used to help confirm the diagnosis of schizophrenia. In preliminary studies using multiplex immunoassay profiling technology, we identified a disease signature comprised of 51 analytes which could distinguish schizophrenia (n = 250) from control (n = 230) subjects. In the next stage, these analytes were developed as a refined 51-plex immunoassay panel for validation using a large independent cohort of schizophrenia (n = 577) and control (n = 229) subjects. The resulting test yielded an overall sensitivity of 83% and specificity of 83% with a receiver operating characteristic area under the curve (ROC-AUC) of 89%. These 51 immunoassays and the associated decision rule delivered a sensitive and specific prediction for the presence of schizophrenia in patients compared to matched healthy controls.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA