Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
PLoS One ; 17(12): e0277532, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36454869

RESUMO

There are currently no preventive or disease-modifying therapies for Parkinson's Disease (PD). Failures in clinical trials necessitate a re-evaluation of existing pre-clinical models in order to adopt systems that better recapitulate underlying disease mechanisms and better predict clinical outcomes. In recent years, models utilizing patient-derived induced pluripotent stem cells (iPSC) have emerged as attractive models to recapitulate disease-relevant neuropathology in vitro without exogenous overexpression of disease-related pathologic proteins. Here, we utilized iPSC derived from patients with early-onset PD and dementia phenotypes that harbored either a point mutation (A53T) or multiplication at the α-synuclein/SNCA gene locus. We generated a three-dimensional (3D) cortical neurosphere culture model to better mimic the tissue microenvironment of the brain. We extensively characterized the differentiation process using quantitative PCR, Western immunoblotting and immunofluorescence staining. Differentiated and aged neurospheres revealed alterations in fatty acid profiles and elevated total and pathogenic phospho-α-synuclein levels in both A53T and the triplication lines compared to their isogenic control lines. Furthermore, treatment of the neurospheres with a small molecule inhibitor of stearoyl CoA desaturase (SCD) attenuated the protein accumulation and aberrant fatty acid profile phenotypes. Our findings suggest that the 3D cortical neurosphere model is a useful tool to interrogate targets for PD and amenable to test small molecule therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Humanos , alfa-Sinucleína/genética , Doença de Parkinson/genética , Organoides , Ácidos Graxos
3.
Neurotherapeutics ; 19(3): 1018-1036, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35445353

RESUMO

Increasing evidence has shown that Parkinson's disease (PD) impairs midbrain dopaminergic, cortical and other neuronal subtypes in large part due to the build-up of lipid- and vesicle-rich α-synuclein (αSyn) cytotoxic inclusions. We previously identified stearoyl-CoA desaturase (SCD) as a potential therapeutic target for synucleinopathies. A brain-penetrant SCD inhibitor, YTX-7739, was developed and has entered Phase 1 clinical trials. Here, we report the efficacy of YTX-7739 in reversing pathological αSyn phenotypes in various in vitro and in vivo PD models. In cell-based assays, YTX-7739 decreased αSyn-mediated neuronal death, reversed the abnormal membrane interaction of amplified E46K ("3K") αSyn, and prevented pathological phenotypes in A53T and αSyn triplication patient-derived neurospheres, including dysregulated fatty acid profiles and pS129 αSyn accumulation. In 3K PD-like mice, YTX-7739 crossed the blood-brain barrier, decreased unsaturated fatty acids, and prevented progressive motor deficits. Both YTX-7739 treatment and decreasing SCD activity through deletion of one copy of the SCD1 gene (SKO) restored the physiological αSyn tetramer-to-monomer ratio, dopaminergic integrity, and neuronal survival in 3K αSyn mice. YTX-7739 efficiently reduced pS129 + and PK-resistant αSyn in both human wild-type αSyn and 3K mutant mice similar to the level of 3K-SKO. Together, these data provide further validation of SCD as a PD therapeutic target and YTX-7739 as a clinical candidate for treating human α-synucleinopathies.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Encéfalo/metabolismo , Humanos , Camundongos , Neurônios/metabolismo , Doença de Parkinson/genética , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Mol Neurobiol ; 59(4): 2171-2189, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35060064

RESUMO

Stearoyl-CoA desaturase (SCD) is a potential therapeutic target for Parkinson's and related neurodegenerative diseases. SCD inhibition ameliorates neuronal toxicity caused by aberrant α-synuclein, a lipid-binding protein implicated in Parkinson's disease. Its inhibition depletes monounsaturated fatty acids, which may modulate α-synuclein conformations and membrane interactions. Herein, we characterize the pharmacokinetic and pharmacodynamic properties of YTX-7739, a clinical-stage SCD inhibitor. Administration of YTX-7739 to rats and monkeys for 15 days caused a dose-dependent increase in YTX-7739 concentrations that were well-tolerated and associated with concentration-dependent reductions in the fatty acid desaturation index (FADI), the ratio of monounsaturated to saturated fatty acids. An approximate 50% maximal reduction in the carbon-16 desaturation index was observed in the brain, with comparable responses in the plasma and skin. A study with a diet supplemented in SCD products indicates that changes in brain C16 desaturation were due to local SCD inhibition, rather than to changes in systemic fatty acids that reach the brain. Assessment of pharmacodynamic response onset and reversibility kinetics indicated that approximately 7 days of dosing were required to achieve maximal responses, which persisted for at least 2 days after cessation of dosing. YTX-7739 thus achieved sufficient concentrations in the brain to inhibit SCD and produce pharmacodynamic responses that were well-tolerated in rats and monkeys. These results provide a framework for evaluating YTX-7739 pharmacology clinically as a disease-modifying therapy to treat synucleinopathies.


Assuntos
Doença de Parkinson , Estearoil-CoA Dessaturase , Animais , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Metabolismo dos Lipídeos/fisiologia , Ratos , Estearoil-CoA Dessaturase/metabolismo , alfa-Sinucleína/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-33468560

RESUMO

OBJECTIVE: To test the hypothesis that dimethyl fumarate (DMF, Tecfidera) elicits different biological changes from DMF combined with monoethyl fumarate (MEF) (Fumaderm, a psoriasis therapy), we investigated DMF and MEF in rodents and cynomolgus monkeys. Possible translatability of findings was explored with lymphocyte counts from a retrospective cohort of patients with MS. METHODS: In rodents, we evaluated pharmacokinetic and pharmacodynamic effects induced by DMF and MEF monotherapies or in combination (DMF/MEF). Clinical implications were investigated in a retrospective, observational analysis of patients with MS treated with DMF/MEF (n = 36). RESULTS: In rodents and cynomolgus monkeys, monomethyl fumarate (MMF, the primary metabolite of DMF) exhibited higher brain penetration, whereas MEF was preferentially partitioned into the kidney. In mice, transcriptional profiling for DMF and MEF alone identified both common and distinct pharmacodynamic responses, with almost no overlap between DMF- and MEF-induced differentially expressed gene profiles in immune tissues. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated oxidative stress response pathway was exclusively regulated by DMF, whereas apoptosis pathways were activated by MEF. DMF/MEF treatment demonstrated that DMF and MEF functionally interact to modify DMF- and MEF-specific responses in unpredictable ways. In patients with MS, DMF/MEF treatment led to early and pronounced suppression of lymphocytes, predominantly CD8+ T cells. In a multivariate regression analysis, the absolute lymphocyte count (ALC) was associated with age at therapy start, baseline ALC, and DMF/MEF dosage but not with previous immunosuppressive medication and sex. Furthermore, the ALC increased in a small cohort of patients with MS (n = 6/7) after switching from DMF/MEF to DMF monotherapy. CONCLUSIONS: Fumaric acid esters exhibit different biodistribution and may elicit different biological responses; furthermore, pharmacodynamic effects of combinations differ unpredictably from monotherapy. The strong potential to induce lymphopenia in patients with MS may be a result of activation of apoptosis pathways by MEF compared with DMF.


Assuntos
Fumarato de Dimetilo/química , Fumarato de Dimetilo/farmacologia , Fumaratos/química , Fumaratos/farmacologia , Esclerose Múltipla/tratamento farmacológico , Animais , Estudos Transversais , Fumarato de Dimetilo/uso terapêutico , Feminino , Fumaratos/uso terapêutico , Perfilação da Expressão Gênica/métodos , Humanos , Imunossupressores/química , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/sangue , Esclerose Múltipla/genética , Ratos , Ratos Sprague-Dawley , Estudos Retrospectivos
6.
Cell Rep ; 25(10): 2742-2754.e31, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517862

RESUMO

The lack of disease-modifying treatments for neurodegenerative disease stems in part from our rudimentary understanding of disease mechanisms and the paucity of targets for therapeutic intervention. Here we used an integrated discovery paradigm to identify a new therapeutic target for diseases caused by α-synuclein (α-syn), a small lipid-binding protein that misfolds and aggregates in Parkinson's disease and other disorders. Using unbiased phenotypic screening, we identified a series of compounds that were cytoprotective against α-syn-mediated toxicity by inhibiting the highly conserved enzyme stearoyl-CoA desaturase (SCD). Critically, reducing the levels of unsaturated membrane lipids by inhibiting SCD reduced α-syn toxicity in human induced pluripotent stem cell (iPSC) neuronal models. Taken together, these findings suggest that inhibition of fatty acid desaturation has potential as a therapeutic approach for the treatment of Parkinson's disease and other synucleinopathies.


Assuntos
Estearoil-CoA Dessaturase/antagonistas & inibidores , alfa-Sinucleína/toxicidade , Animais , Citoproteção/efeitos dos fármacos , Ácidos Graxos/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxidiazóis/química , Oxidiazóis/farmacologia , Agregados Proteicos , Ratos , Saccharomyces cerevisiae/efeitos dos fármacos , Estearoil-CoA Dessaturase/metabolismo , Triglicerídeos/metabolismo
7.
Sci Rep ; 8(1): 13438, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30194389

RESUMO

Protein interacting with C kinase (PICK1) is a scaffolding protein that is present in dendritic spines and interacts with a wide array of proteins through its PDZ domain. The best understood function of PICK1 is regulation of trafficking of AMPA receptors at neuronal synapses via its specific interaction with the AMPA GluA2 subunit. Disrupting the PICK1-GluA2 interaction has been shown to alter synaptic plasticity, a molecular mechanism of learning and memory. Lack of potent, selective inhibitors of the PICK1 PDZ domain has hindered efforts at exploring the PICK1-GluA2 interaction as a therapeutic target for neurological diseases. Here, we report the discovery of PICK1 small molecule inhibitors using a structure-based drug design strategy. The inhibitors stabilized surface GluA2, reduced Aß-induced rise in intracellular calcium concentrations in cultured neurons, and blocked long term depression in brain slices. These findings demonstrate that it is possible to identify potent, selective PICK1-GluA2 inhibitors which may prove useful for treatment of neurodegenerative disorders.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Espinhas Dendríticas/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Sinapses/metabolismo , Animais , Encéfalo/patologia , Cálcio/metabolismo , Sinalização do Cálcio , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Espinhas Dendríticas/patologia , Desenho de Fármacos , Camundongos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/patologia , Proteínas Nucleares/metabolismo , Domínios PDZ , Receptores de AMPA/metabolismo , Sinapses/patologia
8.
Curr Opin Chem Biol ; 44: 66-74, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29902695

RESUMO

Neurodegenerative diseases can arise from a multitude of different pathological drivers, however protein misfolding appears to be a common molecular feature central to several disorders. Protein folding, and attainment of correct secondary and tertiary structure, is essential for proper protein function. Protein misfolding gives rise to structural perturbations that can result in loss of protein function or a gain of toxic function, such as through aggregation, either of which can initiate and propagate biological responses that are deleterious to cells. Several neurodegenerative diseases, such as Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease and Parkinson's disease, each have identified molecular components in which protein misfolding perturbs cellular systems that ultimately lead to cell death, and this predominately occurs in neurons. Current efforts focused on developing therapies for protein misfolding disorders have employed diverse strategies; inhibiting the production of disease-relevant proteins prone to misfolding, inhibiting the aggregation of misfolded proteins, removing and preventing spread of aggregated misfolded proteins and manipulating cellular systems to mitigate the toxic effects of misfolded proteins. Each of these strategies has yielded therapeutic agents that have transitioned from preclinical proof of concept studies into human clinical testing. These approaches and therapies are described herein.


Assuntos
Descoberta de Drogas/métodos , Doenças Neurodegenerativas/tratamento farmacológico , Deficiências na Proteostase/tratamento farmacológico , Animais , Humanos , Modelos Moleculares , Terapia de Alvo Molecular/métodos , Doenças Neurodegenerativas/patologia , Agregados Proteicos/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Deficiências na Proteostase/patologia
9.
J Cereb Blood Flow Metab ; 38(8): 1354-1370, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28606007

RESUMO

The brain's white matter is highly vulnerable to reductions in cerebral blood flow via mechanisms that may involve elevated microgliosis and pro-inflammatory pathways. In the present study, the effects of severe cerebral hypoperfusion were investigated on white matter function and inflammation. Male C57Bl/6J mice underwent bilateral common carotid artery stenosis and white matter function was assessed at seven days with electrophysiology in response to evoked compound action potentials (CAPs) in the corpus callosum. The peak latency of CAPs and axonal refractoriness was increased following hypoperfusion, indicating a marked functional impairment in white matter, which was paralleled by axonal and myelin pathology and increased density and numbers of microglia/macrophages. The functional impairment in peak latency was significantly correlated with increased microglia/macrophages. Dimethyl fumarate (DMF; 100 mg/kg), a drug with anti-inflammatory properties, was found to reduce peak latency but not axonal refractoriness. DMF had no effect on hypoperfusion-induced axonal and myelin pathology. The density of microglia/macrophages was significantly increased in vehicle-treated hypoperfused mice, whereas DMF-treated hypoperfused mice had similar levels to that of sham-treated mice. The study suggests that increased microglia/macrophages following cerebral hypoperfusion contributes to the functional impairment in white matter that may be amenable to modulation by DMF.


Assuntos
Transtornos Cerebrovasculares/tratamento farmacológico , Fumarato de Dimetilo/uso terapêutico , Imunossupressores/uso terapêutico , Inflamação/tratamento farmacológico , Microglia/efeitos dos fármacos , Substância Branca/irrigação sanguínea , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/patologia , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/imunologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/patologia , Substância Branca/imunologia , Substância Branca/patologia
10.
J Biol Chem ; 292(43): 17963-17974, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-28860188

RESUMO

Aberrant activation of matrix metalloproteinases (MMPs) is a common feature of pathological cascades observed in diverse disorders, such as cancer, fibrosis, immune dysregulation, and neurodegenerative diseases. MMP-9, in particular, is highly dynamically regulated in several pathological processes. Development of MMP inhibitors has therefore been an attractive strategy for therapeutic intervention. However, a long history of failed clinical trials has demonstrated that broad-spectrum MMP inhibitors have limited clinical utility, which has spurred the development of inhibitors selective for individual MMPs. Attaining selectivity has been technically challenging because of sequence and structural conservation across the various MMPs. Here, through a biochemical and structural screening paradigm, we have identified JNJ0966, a highly selective compound that inhibited activation of MMP-9 zymogen and subsequent generation of catalytically active enzyme. JNJ0966 had no effect on MMP-1, MMP-2, MMP-3, MMP-9, or MMP-14 catalytic activity and did not inhibit activation of the highly related MMP-2 zymogen. The molecular basis for this activity was characterized as an interaction of JNJ0966 with a structural pocket in proximity to the MMP-9 zymogen cleavage site near Arg-106, which is distinct from the catalytic domain. JNJ0966 was efficacious in reducing disease severity in a mouse experimental autoimmune encephalomyelitis model, demonstrating the viability of this therapeutic approach. This discovery reveals an unprecedented pharmacological approach to MMP inhibition, providing an opportunity to improve selectivity of future clinical drug candidates. Targeting zymogen activation in this manner may also allow for pharmaceutical exploration of other enzymes previously viewed as intractable drug targets.


Assuntos
Precursores Enzimáticos/antagonistas & inibidores , Precursores Enzimáticos/química , Metaloproteinase 9 da Matriz/química , Inibidores de Metaloproteinases de Matriz/química , Regulação Alostérica , Animais , Células COS , Domínio Catalítico , Chlorocebus aethiops , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Domínios Proteicos
12.
Sci Rep ; 7: 42054, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28181536

RESUMO

Dimethyl fumarate (DMF) is indicated for the treatment of relapsing multiple sclerosis and may exert therapeutic effects via activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2) pathway. Following oral DMF administration, central nervous system (CNS) tissue is predominantly exposed to monomethyl fumarate (MMF), the bioactive metabolite of DMF, which can stabilize NRF2 and induce antioxidant gene expression; however, the detailed NRF2-dependent mechanisms modulated by MMF that lead to cytoprotection are unknown. Our data identify a mechanism for MMF-mediated cytoprotection in human astrocytes that functions in an OSGIN1-dependent manner, specifically via upregulation of the OSGIN1-61 kDa isoform. NRF2-dependent OSGIN1 expression induced P53 nuclear translocation following MMF administration, leading to cell-cycle inhibition and cell protection against oxidative challenge. This study provides mechanistic insight into MMF-mediated cytoprotection via NRF2, OSGIN1, and P53 in human CNS-derived cells and contributes to our understanding of how DMF may act clinically to ameliorate pathological processes in neurodegenerative disease.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Citoproteção , Fumaratos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas/metabolismo , Proteínas Reguladoras de Apoptose , Células Cultivadas , Humanos
13.
J Neuroimmunol ; 299: 35-44, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27725119

RESUMO

Delayed-release dimethyl fumarate (DMF) is an approved treatment for multiple sclerosis (MS). Microglia are considered central to MS pathophysiology, however the effects of DMF and the primary metabolite monomethyl fumarate (MMF) on microglia are not well characterized. We demonstrated that DMF and MMF altered transcriptional responses in primary microglia related to the nuclear factor (erythroid-derived 2)-like 2 pathway. Additionally, through an NRF2 independent manner, DMF, but not MMF significantly reduced production of proinflammatory mediators in classically activated microglia, and further rescued mitochondrial respiratory deficits in primary cortical neurons that were induced by activated microglia. These data suggest the mechanism of action of DMF may involve modulation of microglia inflammatory responses and attenuation of neurotoxicity.


Assuntos
Microambiente Celular/efeitos dos fármacos , Fumarato de Dimetilo/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Células Cultivadas , Microambiente Celular/fisiologia , Fumaratos/farmacologia , Imunossupressores/farmacologia , Mediadores da Inflamação/metabolismo , Masculino , Maleatos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley
14.
Nature ; 537(7618): 50-6, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582220

RESUMO

Alzheimer's disease (AD) is characterized by deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aß to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aß. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aß, and reduce soluble and insoluble Aß in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aß in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating-Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais Humanizados/uso terapêutico , Placa Amiloide/tratamento farmacológico , Placa Amiloide/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/efeitos dos fármacos , Amiloide/metabolismo , Peptídeos beta-Amiloides/química , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ensaios Clínicos Fase III como Assunto , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Modelos Biológicos , Placa Amiloide/patologia , Agregação Patológica de Proteínas/tratamento farmacológico , Solubilidade
15.
Proc Natl Acad Sci U S A ; 113(17): 4777-82, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27078105

RESUMO

Dimethyl fumarate (DMF) (BG-12, Tecfidera) is a fumaric acid ester (FAE) that was advanced as a multiple sclerosis (MS) therapy largely for potential neuroprotection as it was recognized that FAEs are capable of activating the antioxidative transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway. However, DMF treatment in randomized controlled MS trials was associated with marked reductions in relapse rate and development of active brain MRI lesions, measures considered to reflect CNS inflammation. Here, we investigated the antiinflammatory contribution of Nrf2 in DMF treatment of the MS model, experimental autoimmune encephalomyelitis (EAE). C57BL/6 wild-type (WT) and Nrf2-deficient (Nrf2(-/-)) mice were immunized with myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 (p35-55) for EAE induction and treated with oral DMF or vehicle daily. DMF protected WT and Nrf2(-/-) mice equally well from development of clinical and histologic EAE. The beneficial effect of DMF treatment in Nrf2(-/-) and WT mice was accompanied by reduced frequencies of IFN-γ and IL-17-producing CD4(+) cells and induction of antiinflammatory M2 (type II) monocytes. DMF also modulated B-cell MHC II expression and reduced the incidence of clinical disease in a B-cell-dependent model of spontaneous CNS autoimmunity. Our observations that oral DMF treatment promoted immune modulation and provided equal clinical benefit in acute EAE in Nrf2(-/-) and WT mice, suggest that the antiinflammatory activity of DMF in treatment of MS patients may occur through alternative pathways, independent of Nrf2.


Assuntos
Imunidade Adaptativa/imunologia , Fumarato de Dimetilo/administração & dosagem , Imunidade Inata/imunologia , Imunomodulação/imunologia , Fator 2 Relacionado a NF-E2/imunologia , Baço/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Administração Oral , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/administração & dosagem , Imunomodulação/efeitos dos fármacos , Imunossupressores/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/efeitos dos fármacos
16.
Antioxid Redox Signal ; 24(18): 1058-71, 2016 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-26980071

RESUMO

AIMS: Gastro-resistant dimethyl fumarate (DMF) is an oral therapeutic indicated for the treatment of relapsing multiple sclerosis. Recent data suggest that a primary pharmacodynamic response to DMF treatment is activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2) pathway; however, the gene targets modulated downstream of NRF2 that contribute to DMF-dependent effects are poorly understood. RESULTS: Using wild-type and NRF2 knockout mice, we characterized DMF transcriptional responses throughout the brain and periphery to understand DMF effects in vivo and to explore the necessity of NRF2 in this process. Our findings identified tissue-specific expression of NRF2 target genes as well as NRF2-dependent and -independent gene regulation after DMF administration. Furthermore, using gene ontology, we identified common biological pathways that may be regulated by DMF and contribute to in vivo functional effects. INNOVATION: Together, these data suggest that DMF modulates transcription through multiple pathways, which has implications for the cytoprotective, immunomodulatory, and clinical properties of DMF. CONCLUSION: These findings provide further understanding of the DMF mechanism of action and propose potential therapeutic targets that warrant further investigation for treating neurodegenerative diseases. Antioxid. Redox Signal. 24, 1058-1071.


Assuntos
Anti-Inflamatórios/farmacocinética , Fumarato de Dimetilo/farmacocinética , Fator 2 Relacionado a NF-E2/metabolismo , Administração Oral , Animais , Anti-Inflamatórios/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Fumarato de Dimetilo/administração & dosagem , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Distribuição Tecidual , Transcriptoma/efeitos dos fármacos
17.
Antioxid Redox Signal ; 25(2): 61-77, 2016 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-27009601

RESUMO

AIMS: This preclinical study was aimed at determining whether pharmacological targeting of transcription factor NRF2, a master controller of many homeostatic genes, might provide a disease-modifying therapy in the animal model of Parkinson's disease (PD) that best reproduces the main hallmark of this pathology, that is, α-synucleinopathy, and associated events, including nigral dopaminergic cell death, oxidative stress, and neuroinflammation. RESULTS: Pharmacological activation of NRF2 was achieved at the basal ganglia by repurposing dimethyl fumarate (DMF), a drug already in use for the treatment of multiple sclerosis. Daily oral gavage of DMF protected nigral dopaminergic neurons against α-SYN toxicity and decreased astrocytosis and microgliosis after 1, 3, and 8 weeks from stereotaxic delivery to the ventral midbrain of recombinant adeno-associated viral vector expressing human α-synuclein. This protective effect was not observed in Nrf2-knockout mice. In vitro studies indicated that this neuroprotective effect was correlated with altered regulation of autophagy markers SQTSM1/p62 and LC3 in MN9D, BV2, and IMA 2.1 and with a shift in microglial dynamics toward a less pro-inflammatory and a more wound-healing phenotype. In postmortem samples of PD patients, the cytoprotective proteins associated with NRF2 expression, NQO1 and p62, were partly sequestered in Lewy bodies, suggesting impaired neuroprotective capacity of the NRF2 signature. INNOVATION: These experiments provide a compelling rationale for targeting NRF2 with DMF as a therapeutic strategy to reinforce endogenous brain defense mechanisms against PD-associated synucleinopathy. CONCLUSION: DMF is ready for clinical validation in PD. Antioxid. Redox Signal. 25, 61-77.


Assuntos
Fumarato de Dimetilo/farmacologia , Reposicionamento de Medicamentos , Fator 2 Relacionado a NF-E2/agonistas , Doença de Parkinson/metabolismo , Sinucleínas/metabolismo , Animais , Autofagia , Encéfalo/metabolismo , Fumarato de Dimetilo/administração & dosagem , Modelos Animais de Doenças , Expressão Gênica , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinucleínas/genética
18.
Neuropharmacology ; 103: 57-68, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26690893

RESUMO

Reducing the production of larger aggregation-prone amyloid ß-peptides (Aß) remains an untested therapeutic approach for reducing the appearance and growth of Aß plaques in the brain, which are a hallmark pathological feature of Alzheimer's disease. γ-Secretase modulators (GSMs) are therapeutics that impact γ-secretase-dependent cleavage of amyloid precursor protein to promote the production of shorter Aß peptides that are less prone to aggregation and plaque deposition. This is accomplished without inhibiting overall γ-secretase function and cleavage of other substrates, which is believed to be a source of deleterious side effects. Here, we report the pharmacokinetic and pharmacodynamic properties of BIIB042, a novel bioavailable and brain-penetrant GSM. In cell-based assays, BIIB042 reduced the levels of Aß42, increased the levels of Aß38 and had little effect on the levels of Aß40, the most abundant Aß species. Similar pharmacodynamic properties were confirmed in the central nervous system and in plasma of mice and rats, and also in plasma of cynomolgus monkeys after a single oral dose of BIIB042. BIIB042 reduced Aß42 levels and Aß plaque burden in Tg2576 mice, which overexpress human amyloid precursor protein and serve as a model system for Alzheimer's disease. BIIB042 did not inhibit cleavage of other γ-secretase substrates in cell-based and in vivo signaling and cleavage assays. The pharmacodynamic effects of lowering Aß42 in the central nervous system coupled with demonstrated efficacy in reducing plaque pathology suggests modulation of γ-secretase, with molecules like BIIB042, is a compelling therapeutic approach for the treatment of Alzheimer's disease.


Assuntos
Aldeídos/farmacocinética , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Aldeídos/administração & dosagem , Peptídeos beta-Amiloides/sangue , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Macaca fascicularis , Masculino , Camundongos , Placa Amiloide/metabolismo , Isoformas de Proteínas/sangue , Ratos , Ratos Endogâmicos F344
19.
Nat Commun ; 6: 7066, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25967870

RESUMO

Forebrain neurons have weak intrinsic antioxidant defences compared with astrocytes, but the molecular basis and purpose of this is poorly understood. We show that early in mouse cortical neuronal development in vitro and in vivo, expression of the master-regulator of antioxidant genes, transcription factor NF-E2-related-factor-2 (Nrf2), is repressed by epigenetic inactivation of its promoter. Consequently, in contrast to astrocytes or young neurons, maturing neurons possess negligible Nrf2-dependent antioxidant defences, and exhibit no transcriptional responses to Nrf2 activators, or to ablation of Nrf2's inhibitor Keap1. Neuronal Nrf2 inactivation seems to be required for proper development: in maturing neurons, ectopic Nrf2 expression inhibits neurite outgrowth and aborization, and electrophysiological maturation, including synaptogenesis. These defects arise because Nrf2 activity buffers neuronal redox status, inhibiting maturation processes dependent on redox-sensitive JNK and Wnt pathways. Thus, developmental epigenetic Nrf2 repression weakens neuronal antioxidant defences but is necessary to create an environment that supports neuronal development.


Assuntos
Antioxidantes/metabolismo , Córtex Cerebral/citologia , Epigênese Genética/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Córtex Cerebral/embriologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Fenômenos Eletrofisiológicos , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética
20.
J Neuroimmunol ; 283: 74-85, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26004161

RESUMO

Fumarate-containing pharmaceuticals are potent therapeutic agents that influence multiple cellular pathways. Despite proven clinical efficacy, there is a significant lack of data that directly defines the molecular mechanisms of action of related, yet distinct fumarate compounds. We systematically compared the impact of dimethyl fumarate (DMF), monomethyl fumarate (MMF) and a mixture of monoethyl fumarate salts (Ca(++), Mg(++), Zn(++); MEF) on defined cellular responses. We demonstrate that DMF inhibited NF-κB-driven cytokine production and nuclear translocation of p65 and p52 in an Nrf2-independent manner. Equivalent doses of MMF and MEF did not affect NF-κB signaling. These results highlight a key difference in the biological impact of related, yet distinct fumarate compounds.


Assuntos
Fumaratos/farmacologia , NF-kappa B/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Neoplasias Ósseas/patologia , Linfoma de Burkitt/patologia , Cátions/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/metabolismo , Fumarato de Dimetilo , Humanos , Técnicas In Vitro , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Maleatos/farmacologia , Camundongos , Camundongos Knockout , Estrutura Molecular , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/fisiologia , Subunidade p52 de NF-kappa B/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Osteossarcoma/patologia , Transdução de Sinais/efeitos dos fármacos , Baço/citologia , Fator de Transcrição RelA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA