Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
J Drugs Dermatol ; 18(5): 448-453, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-31141850

RESUMO

Background: Studies have identified numerous genetic polymorphisms associated with increased risk of melanoma and non-melanoma skin cancer (NMSC). In this pilot study, we aimed to examine whether previously identified melanoma and non-melanoma associated single nucleotide polymorphisms (SNPs) which were found to be associated with cutaneous malignancy were also present in a relatively heterogeneous population with a history of skin cancer versus an age and environmental matched controls. The undertaking of this project serves to further the current understanding of the genetic profile for those at higher risk for developing skin cancer. Methods: Nineteen NMSC patients and their age-matched and environmental controls underwent genotyping of 7 previously discovered SNPs associated with melanoma and NMSC. Results: In a random, heterogeneous population in Southern California, SNP's Chr1, PAD16, PIGU, TDG had a similar association with NMSC previously reported in prior studies. Due to small trial size, no conclusions or observable associations could be drawn from the SNPs MC1R, TP53, and XRCC1. Conclusion: This data supports that 4 of the 7 SNP's studied had similar associations and could potentially be predictive tool of NMSC risk in this patient population. The remaining three SNP's did not have a definitive association with malignancy. Larger studies are needed to further elucidate the specific roles of these SNPs collectively and ultimately to develop a genetic profile for those patients at increased risk of developing skin cancer. J Drugs Dermatol. 2019;18(5):448-453.


Assuntos
Predisposição Genética para Doença , Melanoma/epidemiologia , Mucosa Bucal/patologia , Polimorfismo de Nucleotídeo Único/genética , Neoplasias Cutâneas/epidemiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , California/epidemiologia , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Melanoma/etiologia , Melanoma/genética , Pessoa de Meia-Idade , Projetos Piloto , Valor Preditivo dos Testes , Estudos Prospectivos , Fatores de Risco , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética , Manejo de Espécimes , Adulto Jovem
2.
Clin Ther ; 40(7): 1060-1065, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30049501

RESUMO

Cell therapy has existed since the first bone marrow transplant in the 1950s involving identical twins. The blood-forming stem cells were used to restore healthy blood cells for the twin with leukemia. It was not until 1968 that genetic matching (known as human leukocyte antigen matching) was known to be important, and not until 1973 that bone marrow transplants were performed from non-twin-related and nonrelated donors. The most important application of human stem cells is for the generation of cells and tissues for cell-based therapies. Currently, donated organs and tissues are often the only option to replace diseased, injured, or destroyed tissue. The availability for these transplantable tissues and organs is very limited, however. To satisfy the demand for a source for these cells and tissues, induced pluripotent stem cells that have been differentiated into specific cell types can serve as a renewable source of replacement cells and tissues. A bank of suitable human leukocyte antigen-matched cells will be an important source providing immediate availability of cells that are readily scalable, economical, and well characterized. Areas of active pursuit with stem cell therapy is being investigated for treating diseases such as macular degeneration, spinal cord injury, stroke, burns, heart disease, diabetes, osteoarthritis, rheumatoid arthritis, and neurodegenerative diseases. This article describes the advantages and hurdles for the use of induced pluripotent cells as the starting material for a source of replacement cells for regenerative medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco , História do Século XX , História do Século XXI , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Japão , Medicina Regenerativa/história , Traumatismos da Medula Espinal/história , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/história , Bancos de Tecidos/história , Estados Unidos
3.
Bioorg Med Chem Lett ; 27(23): 5144-5148, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29103974

RESUMO

Spinal muscular atrophy (SMA) is a neurodegenerative disorder that results from mutations in the SMN1 gene, leading to survival motor neuron (SMN) protein deficiency. One therapeutic strategy for SMA is to identify compounds that enhance the expression of the SMN2 gene, which normally only is a minor contributor to functional SMN protein production, but which is unaffected in SMA. A recent high-throughput screening campaign identified a 3,4-dihydro-4-phenyl-2(1H)-quinolinone derivative (2) that increases the expression of SMN2 by 2-fold with an EC50 = 8.3 µM. A structure-activity relationship (SAR) study revealed that the array of tolerated substituents, on either the benzo portion of the quinolinone or the 4-phenyl, was very narrow. However, the lactam ring of the quinolinone was more amenable to modifications. For example, the quinazolinone (9a) and the benzoxazepin-2(3H)-one (19) demonstrated improved potency and efficacy for increase in SMN2 expression as compared to 2.


Assuntos
Quinolonas/química , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo , Animais , Linhagem Celular , Ciclização , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Microssomos Hepáticos/metabolismo , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patologia , Quinolonas/farmacologia , RNA Mensageiro/metabolismo , Solubilidade , Relação Estrutura-Atividade , Proteína 2 de Sobrevivência do Neurônio Motor/genética
4.
Science ; 357(6354): 891-898, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860381

RESUMO

Copy number mutations implicate excess production of α-synuclein as a possibly causative factor in Parkinson's disease (PD). Using an unbiased screen targeting endogenous gene expression, we discovered that the ß2-adrenoreceptor (ß2AR) is a regulator of the α-synuclein gene (SNCA). ß2AR ligands modulate SNCA transcription through histone 3 lysine 27 acetylation of its promoter and enhancers. Over 11 years of follow-up in 4 million Norwegians, the ß2AR agonist salbutamol, a brain-penetrant asthma medication, was associated with reduced risk of developing PD (rate ratio, 0.66; 95% confidence interval, 0.58 to 0.76). Conversely, a ß2AR antagonist correlated with increased risk. ß2AR activation protected model mice and patient-derived cells. Thus, ß2AR is linked to transcription of α-synuclein and risk of PD in a ligand-specific fashion and constitutes a potential target for therapies.


Assuntos
Regulação da Expressão Gênica , Doença de Parkinson/etnologia , Doença de Parkinson/genética , Receptores Adrenérgicos beta 2/metabolismo , alfa-Sinucleína/genética , Acetilação , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Albuterol/farmacologia , Albuterol/uso terapêutico , Animais , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Ligantes , Camundongos , Fármacos Neuroprotetores/farmacologia , Noruega/etnologia , Doença de Parkinson/tratamento farmacológico , Regiões Promotoras Genéticas , Propranolol/farmacologia , Propranolol/uso terapêutico , Receptores Adrenérgicos beta 2/genética , Risco , Substância Negra/metabolismo , Transcrição Gênica/efeitos dos fármacos
5.
J Med Chem ; 60(11): 4594-4610, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28481536

RESUMO

Spinal muscular atrophy (SMA) is the leading genetic cause of infant death. We previously developed a high-throughput assay that employs an SMN2-luciferase reporter allowing identification of compounds that act transcriptionally, enhance exon recognition, or stabilize the SMN protein. We describe optimization and characterization of an analog suitable for in vivo testing. Initially, we identified analog 4m that had good in vitro properties but low plasma and brain exposure in a mouse PK experiment due to short plasma stability; this was overcome by reversing the amide bond and changing the heterocycle. Thiazole 27 showed excellent in vitro properties and a promising mouse PK profile, making it suitable for in vivo testing. This series post-translationally stabilizes the SMN protein, unrelated to global proteasome or autophagy inhibition, revealing a novel therapeutic mechanism that should complement other modalities for treatment of SMA.


Assuntos
Anilidas/farmacologia , Benzamidas/farmacologia , Isoxazóis/farmacologia , Sondas Moleculares , Atrofia Muscular Espinal/terapia , Processamento de Proteína Pós-Traducional , Quinolonas/farmacologia , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Tiazóis/farmacologia , Anilidas/farmacocinética , Anilidas/uso terapêutico , Área Sob a Curva , Benzamidas/farmacocinética , Benzamidas/uso terapêutico , Linhagem Celular , Descoberta de Drogas , Meia-Vida , Humanos , Isoxazóis/farmacocinética , Isoxazóis/uso terapêutico , Estabilidade Proteica , Quinolonas/farmacocinética , Quinolonas/uso terapêutico , Relação Estrutura-Atividade , Tiazóis/farmacocinética , Tiazóis/uso terapêutico
6.
Sci Rep ; 6: 30263, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27457881

RESUMO

Pre-treatment or priming of mesenchymal stem cells (MSC) prior to transplantation can significantly augment the immunosuppressive effect of MSC-based therapies. In this study, we screened a library of 1402 FDA-approved bioactive compounds to prime MSC. We identified tetrandrine as a potential hit that activates the secretion of prostaglandin E2 (PGE2), a potent immunosuppressive agent, by MSC. Tetrandrine increased MSC PGE2 secretion through the NF-κB/COX-2 signaling pathway. When co-cultured with mouse macrophages (RAW264.7), tetrandrine-primed MSC attenuated the level of TNF-α secreted by RAW264.7. Furthermore, systemic transplantation of primed MSC into a mouse ear skin inflammation model significantly reduced the level of TNF-α in the inflamed ear, compared to unprimed cells. Screening of small molecules to pre-condition cells prior to transplantation represents a promising strategy to boost the therapeutic potential of cell therapy.


Assuntos
Benzilisoquinolinas/farmacologia , Imunossupressores/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Ciclo-Oxigenase 2/imunologia , Humanos , Imunomodulação/efeitos dos fármacos , Programas de Rastreamento , Células-Tronco Mesenquimais/imunologia , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Bibliotecas de Moléculas Pequenas
8.
PLoS One ; 10(7): e0131297, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26176772

RESUMO

Currently there is no effective treatment available for major neurodegenerative disorders associated to protein misfolding, including Alzheimer's and Parkinson's disease. One of most promising therapeutic approaches under development focuses on inhibiting the misfolding and aggregation pathway. However, it is likely that by the time clinical symptoms appear, there is a large accumulation of misfolded aggregates and a very substantial damage to the brain. Thus, it seems that at the clinical stage of the disease it is necessary also to develop strategies aiming to prevent the neuronal damage produced by already formed misfolded aggregates. Chronic activation of calcineurin (CaN), a type IIB phosphatase, has been implicated as a pivotal molecule connecting synaptic loss and neuronal damage to protein misfolding. The fact that the crystal structure of CaN is also well established makes it an ideal target for drug discovery. CaN activity assays for High Throughput Screening (HTS) reported so far are based on absorbance. In this article we report the development of a fluorescent quenching based CaN activity assay suitable for robotic screening of large chemical libraries to find novel inhibitors. The assay yielded a Z score of 0.84 with coefficient of variance ≤ 15%. Our results also show that this assay can be used to identify CaN inhibitors with a wide range of potencies.


Assuntos
Inibidores de Calcineurina/química , Avaliação Pré-Clínica de Medicamentos , Ensaios Enzimáticos , Corantes Fluorescentes/química , Ensaios de Triagem em Larga Escala , Humanos , Cinética , Corantes de Rosanilina/química , Bibliotecas de Moléculas Pequenas/química , Espectrometria de Fluorescência
9.
PLoS One ; 10(7): e0132604, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26161952

RESUMO

Microgliosis is a major hallmark of Alzheimer's disease (AD) brain pathology. Aß peptide is hypothesized to act as a stimulus for microglia leading to activation of non-receptor tyrosine kinases and subsequent secretion of pro-inflammatory cytokines. Therefore, the signaling pathways mediating microglial activation may be important therapeutic targets of anti-inflammatory therapy for AD. Four novel compounds were chosen after high throughput screening kinase activity assays determined them as potential Lyn kinase inhibitors. Their kinase inhibitory and anti-inflammatory effect on Aß-stimulated activation was assessed using the murine microglial cell line, BV2. Cells were treated with the compounds to determine effects on active, phosphorylated levels of Src family kinases, Src and Lyn, as well as MAP kinases ERK, JNK and p38. Only one compound, LDDN-0003499, produced a dose dependent decrease in basal levels of active, phosphorylated Src and Lyn in the BV2 cells. LDDN-0003499 treatment also attenuated the Aß-stimulated increase in active, phosphorylated levels of Lyn/Src and TNFα and IL-6 secretion. This study identifies a novel small molecule Src family tyrosine kinase inhibitor with anti-inflammatory effects in response to Aß stimulation of microglia. Further in vitro/in vivo characterization of LDDN-0003499 as well as structural modification may provide a new tool for attenuating microglial-mediated brain inflammatory conditions such as that occurring in AD.


Assuntos
Gliose/patologia , Quinases da Família src/antagonistas & inibidores , Administração Oral , Peptídeos beta-Amiloides/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Células CACO-2 , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gliose/enzimologia , Humanos , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/metabolismo
10.
J Exp Med ; 212(3): 319-32, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25711212

RESUMO

Glutamatergic systems play a critical role in cognitive functions and are known to be defective in Alzheimer's disease (AD) patients. Previous literature has indicated that glial glutamate transporter EAAT2 plays an essential role in cognitive functions and that loss of EAAT2 protein is a common phenomenon observed in AD patients and animal models. In the current study, we investigated whether restored EAAT2 protein and function could benefit cognitive functions and pathology in APPSw,Ind mice, an animal model of AD. A transgenic mouse approach via crossing EAAT2 transgenic mice with APPSw,Ind. mice and a pharmacological approach using a novel EAAT2 translational activator, LDN/OSU-0212320, were conducted. Findings from both approaches demonstrated that restored EAAT2 protein function significantly improved cognitive functions, restored synaptic integrity, and reduced amyloid plaques. Importantly, the observed benefits were sustained one month after compound treatment cessation, suggesting that EAAT2 is a potential disease modifier with therapeutic potential for AD.


Assuntos
Doença de Alzheimer/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Piridazinas/farmacologia , Piridinas/farmacologia , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Cognição/efeitos dos fármacos , Cognição/fisiologia , Modelos Animais de Doenças , Transportador 2 de Aminoácido Excitatório/genética , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/genética , Placa Amiloide/metabolismo
11.
Antiviral Res ; 112: 113-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25453344

RESUMO

INTRODUCTION: This study evaluates polyomavirus JC (JCV) large T antigen (LTA) as a potential target for drug development. LTA is a hexameric protein with a helicase activity that is powered by ATP binding and hydrolysis. The helicase and ATPase function is critical for viral replication. METHODS: Recombinant JCV LTA was produced in an Escherichia coli based expression plasmid. ATPase activity was measured using the malachite green assay. A high throughput screen was completed using a brain-biased library of 75,000 drug-like compounds selected for physicochemical properties consistent with blood-brain barrier permeability. RESULTS: Five compounds showed non-competitive inhibition of ATPase activity with an EC50 ⩽ 15 µM. Modest antiviral activity was demonstrated in an immunofluorescence assay for JCV VP-1 expression in COS7 cells (EC50 15, 18, 20, 27, and 52 µM respectively). The compounds also inhibited viral replication in a real time PCR assay at comparable concentrations. LD50 in the MTS96 and Cell TiterGlo assays was >100 µM for all compounds in COS7 as well as HEK293 cells. However, two compounds inhibited cell proliferation in culture with IC50 values of 43 and 34 µM respectively. Despite substantial amino acid similarity between polyomavirus JC, BK and SV40 proteins, these compounds differ from those previously reported to inhibit SV40 LTA ATPase in chemical structure as well as a non-competitive mechanism of inhibition. CONCLUSION: LTA ATPase is a valid target for discovery. Additional screening and chemical optimization is needed to develop clinically useful compounds with less toxicity, which should be measured by metabolic as well as cell proliferation assays.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Antígenos Virais de Tumores/metabolismo , Antivirais/isolamento & purificação , Antivirais/farmacologia , Inibidores Enzimáticos/isolamento & purificação , Inibidores Enzimáticos/farmacologia , Vírus JC/enzimologia , Adenosina Trifosfatases/genética , Animais , Antígenos Virais de Tumores/genética , Linhagem Celular , Chlorocebus aethiops , Colorimetria/métodos , Avaliação Pré-Clínica de Medicamentos , Escherichia coli/genética , Escherichia coli/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Vírus JC/efeitos dos fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Replicação Viral/efeitos dos fármacos
12.
PLoS Biol ; 12(8): e1001923, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25093460

RESUMO

STEP (STriatal-Enriched protein tyrosine Phosphatase) is a neuron-specific phosphatase that regulates N-methyl-D-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking, as well as ERK1/2, p38, Fyn, and Pyk2 activity. STEP is overactive in several neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease (AD). The increase in STEP activity likely disrupts synaptic function and contributes to the cognitive deficits in AD. AD mice lacking STEP have restored levels of glutamate receptors on synaptosomal membranes and improved cognitive function, results that suggest STEP as a novel therapeutic target for AD. Here we describe the first large-scale effort to identify and characterize small-molecule STEP inhibitors. We identified the benzopentathiepin 8-(trifluoromethyl)-1,2,3,4,5-benzopentathiepin-6-amine hydrochloride (known as TC-2153) as an inhibitor of STEP with an IC50 of 24.6 nM. TC-2153 represents a novel class of PTP inhibitors based upon a cyclic polysulfide pharmacophore that forms a reversible covalent bond with the catalytic cysteine in STEP. In cell-based secondary assays, TC-2153 increased tyrosine phosphorylation of STEP substrates ERK1/2, Pyk2, and GluN2B, and exhibited no toxicity in cortical cultures. Validation and specificity experiments performed in wild-type (WT) and STEP knockout (KO) cortical cells and in vivo in WT and STEP KO mice suggest specificity of inhibitors towards STEP compared to highly homologous tyrosine phosphatases. Furthermore, TC-2153 improved cognitive function in several cognitive tasks in 6- and 12-mo-old triple transgenic AD (3xTg-AD) mice, with no change in beta amyloid and phospho-tau levels.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/enzimologia , Inibidores Enzimáticos/uso terapêutico , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Animais , Benzotiepinas/farmacologia , Benzotiepinas/uso terapêutico , Domínio Catalítico , Morte Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Transtornos Cognitivos/complicações , Transtornos Cognitivos/patologia , Cisteína/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Especificidade por Substrato/efeitos dos fármacos
13.
Hum Mol Genet ; 23(18): 4887-905, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24794857

RESUMO

Mutations in LRRK2 are one of the primary genetic causes of Parkinson's disease (PD). LRRK2 contains a kinase and a GTPase domain, and familial PD mutations affect both enzymatic activities. However, the signaling mechanisms regulating LRRK2 and the pathogenic effects of familial mutations remain unknown. Identifying the signaling proteins that regulate LRRK2 function and toxicity remains a critical goal for the development of effective therapeutic strategies. In this study, we apply systems biology tools to human PD brain and blood transcriptomes to reverse-engineer a LRRK2-centered gene regulatory network. This network identifies several putative master regulators of LRRK2 function. In particular, the signaling gene RGS2, which encodes for a GTPase-activating protein (GAP), is a key regulatory hub connecting the familial PD-associated genes DJ-1 and PINK1 with LRRK2 in the network. RGS2 expression levels are reduced in the striata of LRRK2 and sporadic PD patients. We identify RGS2 as a novel interacting partner of LRRK2 in vivo. RGS2 regulates both the GTPase and kinase activities of LRRK2. We show in mammalian neurons that RGS2 regulates LRRK2 function in the control of neuronal process length. RGS2 is also protective against neuronal toxicity of the most prevalent mutation in LRRK2, G2019S. We find that RGS2 regulates LRRK2 function and neuronal toxicity through its effects on kinase activity and independently of GTPase activity, which reveals a novel mode of action for GAP proteins. This work identifies RGS2 as a promising target for interfering with neurodegeneration due to LRRK2 mutations in PD patients.


Assuntos
Redes Reguladoras de Genes , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas RGS/metabolismo , Animais , Encéfalo/metabolismo , Caenorhabditis elegans/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Neurônios/metabolismo , Proteínas Oncogênicas/metabolismo , Doença de Parkinson/sangue , Proteína Desglicase DJ-1 , Proteínas Quinases/metabolismo , Biologia de Sistemas/métodos , Transcriptoma
14.
J Biol Chem ; 289(19): 13042-53, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24695735

RESUMO

The effect of leucine-rich repeat kinase 2 (LRRK2) mutation I2020T on its kinase activity has been controversial, with both increased and decreased effects being reported. We conducted steady-state and pre-steady-state kinetic studies on LRRKtide and its analog LRRKtide(S). Their phosphorylation differs by the rate-limiting steps: product release is rate-limiting for LRRKtide and phosphoryl transfer is rate-limiting for LRRKtide(S). As a result, we observed that the I2020T mutant is more active than wild type (WT) LRRK2 for LRRKtide(S) phosphorylation, whereas it is less active than WT for LRRKtide phosphorylation. Our pre-steady-state kinetic data suggest that (i) the I2020T mutant accelerates the rates of phosphoryl transfer of both reactions by 3-7-fold; (ii) this increase is masked by a rate-limiting product release step for LRRKtide phosphorylation; and (iii) the observed lower activity of the mutant for LRRKtide phosphorylation is a consequence of its instability: the concentration of the active form of the mutant is 3-fold lower than WT. The I2020T mutant has a dramatically low KATP and therefore leads to resistance to ATP competitive inhibitors. Two well known DFG-out or type II inhibitors are also weaker toward the mutant because they inhibit the mutant in an unexpected ATP competitive mechanism. The I2020 residue lies next to the DYG motif of the activation loop of the LRRK2 kinase domain. Our modeling and metadynamic simulations suggest that the I2020T mutant stabilizes the DYG-in active conformation and creates an unusual allosteric pocket that can bind type II inhibitors but in an ATP competitive fashion.


Assuntos
Modelos Biológicos , Modelos Moleculares , Mutação de Sentido Incorreto , Doença de Parkinson , Proteínas Serina-Treonina Quinases , Trifosfato de Adenosina , Substituição de Aminoácidos , Animais , Estabilidade Enzimática/genética , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Doença de Parkinson/enzimologia , Doença de Parkinson/genética , Fosforilação/genética , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína
15.
J Biomol Screen ; 19(7): 1024-34, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24598103

RESUMO

PERK is serine/threonine kinase localized to the endoplasmic reticulum (ER) membrane. PERK is activated and contributes to cell survival in response to a variety of physiological stresses that affect protein quality control in the ER, such as hypoxia, glucose depravation, increased lipid biosynthesis, and increased protein translation. Pro-survival functions of PERK are triggered by such stresses, suggesting that development of small-molecule inhibitors of PERK may be efficacious in a variety of disease scenarios. Hence, we have conducted a detailed enzymatic characterization of the PERK kinase to develop a high-throughput-screening assay (HTS) that will permit the identification of small-molecule PERK inhibitors. In addition to establishing the K(m) of PERK for both its primary substrate, eIF2α, and for adenosine triphosphate, further mechanistic studies revealed that PERK targets its substrate via either a random/steady-state ordered mechanism. For HTS, we developed a time-resolved fluorescence resonance energy transfer-based assay that yielded a robust Z' factor and percent coefficient of variation value, enabling the successful screening of 79,552 compounds. This approach yielded one compound that exhibited good in vitro and cellular activity. These results demonstrate the validity of this screen and represent starting points for drug discovery efforts.


Assuntos
Descoberta de Drogas/métodos , Inibidores Enzimáticos/química , Ensaios de Triagem em Larga Escala/métodos , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/química , Animais , Simulação por Computador , Desenho de Fármacos , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/química , Fibroblastos/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Camundongos , Fenótipo , Fosforilação , Transdução de Sinais
16.
J Clin Invest ; 124(3): 1255-67, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24569372

RESUMO

Glial glutamate transporter EAAT2 plays a major role in glutamate clearance in synaptic clefts. Several lines of evidence indicate that strategies designed to increase EAAT2 expression have potential for preventing excitotoxicity, which contributes to neuronal injury and death in neurodegenerative diseases. We previously discovered several classes of compounds that can increase EAAT2 expression through translational activation. Here, we present efficacy studies of the compound LDN/OSU-0212320, which is a pyridazine derivative from one of our lead series. In a murine model, LDN/OSU-0212320 had good potency, adequate pharmacokinetic properties, no observed toxicity at the doses examined, and low side effect/toxicity potential. Additionally, LDN/OSU-0212320 protected cultured neurons from glutamate-mediated excitotoxic injury and death via EAAT2 activation. Importantly, LDN/OSU-0212320 markedly delayed motor function decline and extended lifespan in an animal model of amyotrophic lateral sclerosis (ALS). We also found that LDN/OSU-0212320 substantially reduced mortality, neuronal death, and spontaneous recurrent seizures in a pilocarpine-induced temporal lobe epilepsy model. Moreover, our study demonstrated that LDN/OSU-0212320 treatment results in activation of PKC and subsequent Y-box-binding protein 1 (YB-1) activation, which regulates activation of EAAT2 translation. Our data indicate that the use of small molecules to enhance EAAT2 translation may be a therapeutic strategy for the treatment of neurodegenerative diseases.


Assuntos
Transportador 2 de Aminoácido Excitatório/genética , Fármacos Neuroprotetores/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Piridazinas/farmacologia , Piridinas/farmacologia , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/patologia , Animais , Células do Corno Anterior/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular , Técnicas de Cocultura , Ativação Enzimática/efeitos dos fármacos , Transportador 2 de Aminoácido Excitatório/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Mutação de Sentido Incorreto , Fármacos Neuroprotetores/farmacocinética , Pilocarpina , Proteína Quinase C/metabolismo , Piridazinas/farmacocinética , Piridinas/farmacocinética , Ratos , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/patologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Distribuição Tecidual , Fatores de Transcrição/metabolismo
17.
Expert Opin Drug Discov ; 9(2): 215-20, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24444088

RESUMO

The following conference was the 14th International Conference on Alzheimer's Drug Discovery held 9 - 10 September 2013, in Jersey City, NJ. The conference attracted about 140 attendees with 49% from academia, 36% from industry and private practice, 10% from nonprofit organizations and 2% from the government. The meeting had two plenary speakers that kicked off each morning of the conference and then two sessions each day to cover different aspects of Alzheimer's disease drug discovery. There were sessions on neuroprotection, mitochondrial function, biomarkers, ApoE, tau and protein clearance. The conference was organized by the Alzheimer's Drug Discovery Foundation (ADDF) with all of the presenters supported by grants awarded by the ADDF. The conference had financial support from the pharmaceutical companies Merck & Co., Eli Lilly & Co. and Pfizer, Inc. Friends, exhibitors and media partners also helped financially support the conference.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Animais , Descoberta de Drogas , Humanos , Fármacos Neuroprotetores/uso terapêutico
18.
J Biomol Screen ; 19(1): 44-56, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24019256

RESUMO

TDP-43 is an RNA binding protein found to accumulate in the cytoplasm of brain and spinal cord from patients affected with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Nuclear TDP-43 protein regulates transcription through several mechanisms, and under stressed conditions, it forms cytoplasmic aggregates that co-localize with stress granule (SG) proteins in cell culture. These granules are also found in the brain and spinal cord of patients affected with ALS and FTLD. The mechanism through which TDP-43 might contribute to neurodegenerative diseases is poorly understood. To investigate the pathophysiology of TDP-43 aggregation and to isolate potential therapeutic targets, we screened a chemical library of 75,000 compounds using high-content analysis with PC12 cells that inducibly express human TDP-43 tagged with green fluorescent protein (GFP). The screen identified 16 compounds that dose-dependently decreased the TDP-43 inclusions without significant cellular toxicity or changes in total TDP-43 expression levels. To validate the effect, we tested compounds by Western blot analysis and in a Caenorhabditis elegans model that replicates some of the relevant disease phenotypes. The hits from this assay will be useful for elucidating regulation of TDP-43, stress granule response, and possible ALS therapeutics.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala , Estresse Fisiológico/efeitos dos fármacos , Animais , Animais Geneticamente Modificados , Arsenitos/farmacologia , Caenorhabditis elegans , Linhagem Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Descoberta de Drogas/métodos , Expressão Gênica , Genes Reporter , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Fármacos Neuroprotetores/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Bibliotecas de Moléculas Pequenas , Compostos de Sódio/farmacologia
19.
EMBO Mol Med ; 5(7): 1103-18, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23740718

RESUMO

Spinal muscular atrophy (SMA) is a neurodegenerative disease that causes progressive muscle weakness, which primarily targets proximal muscles. About 95% of SMA cases are caused by the loss of both copies of the SMN1 gene. SMN2 is a nearly identical copy of SMN1, which expresses much less functional SMN protein. SMN2 is unable to fully compensate for the loss of SMN1 in motor neurons but does provide an excellent target for therapeutic intervention. Increased expression of functional full-length SMN protein from the endogenous SMN2 gene should lessen disease severity. We have developed and implemented a new high-throughput screening assay to identify small molecules that increase the expression of full-length SMN from a SMN2 reporter gene. Here, we characterize two novel compounds that increased SMN protein levels in both reporter cells and SMA fibroblasts and show that one increases lifespan, motor function, and SMN protein levels in a severe mouse model of SMA.


Assuntos
Descoberta de Drogas , Atrofia Muscular Espinal/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/uso terapêutico , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Regulação para Cima/efeitos dos fármacos , Animais , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatologia , RNA Mensageiro/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína 1 de Sobrevivência do Neurônio Motor/análise , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/análise
20.
Future Med Chem ; 4(13): 1671-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22924505

RESUMO

The majority of neurodegenerative diseases have an important age component, and thus, understanding the molecular changes that occur during normal aging of the brain is of utmost relevance. In search for the basis of the age-related cognitive decline found in humans, monkeys and rodents, we study the rhesus monkey. Surprisingly, there is no loss of neurons in aged monkey brains. However, we reported white matter and myelin abnormalities in aged monkeys, similar to those observed in Alzheimer's disease and multiple sclerosis patients. In a microarray analysis comparing young and old monkey white matter, we discovered that Klotho is downregulated in the aged brain. We then asked whether there is a connection between the age-related cognitive decline, myelin abnormalities and Klotho downregulation. If such a connection is found, compounds that upregulate Klotho expression could become of therapeutic interest for the treatment of multiple sclerosis, and perhaps even Alzheimer's disease.


Assuntos
Descoberta de Drogas/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronidase/genética , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Envelhecimento/genética , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Células HEK293 , Humanos , Proteínas Klotho , Macaca mulatta , Doenças Neurodegenerativas/patologia , RNA Mensageiro/genética , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...