Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
G3 (Bethesda) ; 10(6): 1843-1852, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32276960

RESUMO

FUS is a nucleic acid binding protein that, when mutated, cause a subset of familial amyotrophic lateral sclerosis (ALS). Expression of FUS in yeast recapitulates several pathological features of the disease-causing mutant proteins, including nuclear to cytoplasmic translocation, formation of cytoplasmic inclusions, and cytotoxicity. Genetic screens using the yeast model of FUS have identified yeast genes and their corresponding human homologs suppressing FUS induced toxicity in yeast, neurons and animal models. To expand the search for human suppressor genes of FUS induced toxicity, we carried out a genome-scale genetic screen using a newly constructed library containing 13570 human genes cloned in an inducible yeast-expression vector. Through multiple rounds of verification, we found 37 human genes that, when overexpressed, suppress FUS induced toxicity in yeast. Human genes with DNA or RNA binding functions are overrepresented among the identified suppressor genes, supporting that perturbations of RNA metabolism is a key underlying mechanism of FUS toxicity.


Assuntos
Esclerose Lateral Amiotrófica , Proteína FUS de Ligação a RNA , Animais , Citoplasma , Humanos , Corpos de Inclusão , Mutação , Proteína FUS de Ligação a RNA/genética , Saccharomyces cerevisiae/genética
2.
J Vis Exp ; (137)2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-30035772

RESUMO

Budding yeast has been widely used as a model in studying proteins associated with human diseases. Genome-wide genetic screening is a powerful tool commonly used in yeast studies. The expression of a number of neurodegenerative disease-associated proteins in yeast causes cytotoxicity and aggregate formation, recapitulating findings seen in patients with these disorders. Here, we describe a method for screening a yeast model of the Amyotrophic Lateral Sclerosis-associated protein FUS for modifiers of its toxicity. Instead of using transformation, this new screening platform relies on the mating of yeast to introduce an arrayed library of plasmids into the yeast model. The mating method has two clear advantages: first, it is highly efficient; second, the pre-transformed arrayed library of plasmids can be stored for long-term as a glycerol stock, and quickly applied to other screens without the labor-intensive step of transformation into the yeast model each time. We demonstrate how this method can successfully be used to screen for genes that modify the toxicity of FUS.


Assuntos
Biblioteca Gênica , Doenças Neurodegenerativas/diagnóstico , Proteínas/metabolismo , Deficiências na Proteostase/diagnóstico , Saccharomyces cerevisiae/patogenicidade , Humanos
3.
Proc Natl Acad Sci U S A ; 114(20): 5065-5066, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28487485
4.
Proc Natl Acad Sci U S A ; 113(34): 9587-92, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27482083

RESUMO

The aggregation of α-synuclein (aSyn) leading to the formation of Lewy bodies is the defining pathological hallmark of Parkinson's disease (PD). Rare familial PD-associated mutations in aSyn render it aggregation-prone; however, PD patients carrying wild type (WT) aSyn also have aggregated aSyn in Lewy bodies. The mechanisms by which WT aSyn aggregates are unclear. Here, we report that inflammation can play a role in causing the aggregation of WT aSyn. We show that activation of the inflammasome with known stimuli results in the aggregation of aSyn in a neuronal cell model of PD. The insoluble aggregates are enriched with truncated aSyn as found in Lewy bodies of the PD brain. Inhibition of the inflammasome enzyme caspase-1 by chemical inhibition or genetic knockdown with shRNA abated aSyn truncation. In vitro characterization confirmed that caspase-1 directly cleaves aSyn, generating a highly aggregation-prone species. The truncation-induced aggregation of aSyn is toxic to neuronal culture, and inhibition of caspase-1 by shRNA or a specific chemical inhibitor improved the survival of a neuronal PD cell model. This study provides a molecular link for the role of inflammation in aSyn aggregation, and perhaps in the pathogenesis of sporadic PD as well.


Assuntos
Caspase 1/genética , Inflamassomos/metabolismo , Corpos de Lewy/metabolismo , Neurônios/metabolismo , Agregados Proteicos/genética , alfa-Sinucleína/genética , Compostos de Alúmen/farmacologia , Caspase 1/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dipeptídeos/farmacologia , Regulação da Expressão Gênica , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Corpos de Lewy/efeitos dos fármacos , Corpos de Lewy/patologia , Lipopolissacarídeos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Nigericina/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Vitamina K 3/farmacologia , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , para-Aminobenzoatos/farmacologia
5.
Proc Natl Acad Sci U S A ; 112(25): 7821-6, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26056265

RESUMO

Over 30% of patients with amyotrophic lateral sclerosis (ALS) exhibit cognitive deficits indicative of frontotemporal dementia (FTD), suggesting a common pathogenesis for both diseases. Consistent with this hypothesis, neuronal and glial inclusions rich in TDP43, an essential RNA-binding protein, are found in the majority of those with ALS and FTD, and mutations in TDP43 and a related RNA-binding protein, FUS, cause familial ALS and FTD. TDP43 and FUS affect the splicing of thousands of transcripts, in some cases triggering nonsense-mediated mRNA decay (NMD), a highly conserved RNA degradation pathway. Here, we take advantage of a faithful primary neuronal model of ALS and FTD to investigate and characterize the role of human up-frameshift protein 1 (hUPF1), an RNA helicase and master regulator of NMD, in these disorders. We show that hUPF1 significantly protects mammalian neurons from both TDP43- and FUS-related toxicity. Expression of hUPF2, another essential component of NMD, also improves survival, whereas inhibiting NMD prevents rescue by hUPF1, suggesting that hUPF1 acts through NMD to enhance survival. These studies emphasize the importance of RNA metabolism in ALS and FTD, and identify a uniquely effective therapeutic strategy for these disorders.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Modelos Biológicos , Neurônios/efeitos dos fármacos , Transativadores/fisiologia , Sobrevivência Celular , Humanos , Fármacos Neuroprotetores/farmacologia , Degradação do RNAm Mediada por Códon sem Sentido , RNA Helicases
6.
J Alzheimers Dis ; 32(4): 949-67, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22903131

RESUMO

Latrepirdine (Dimebon), an anti-histamine, has shown some benefits in trials of neurodegenerative diseases characterized by accumulation of aggregated or misfolded protein such as Alzheimer's disease (AD) and has been shown to promote the removal of α-synuclein protein aggregates in vivo. An important pathway for removal of aggregated or misfolded proteins is the autophagy-lysosomal pathway, which has been implicated in AD pathogenesis, and enhancing this pathway has been shown to have therapeutic potential in AD and other proteinopathies. Here we use a yeast model, Saccharomyces cerevisiae, to investigate whether latrepirdine can enhance autophagy and reduce levels of amyloid-ß (Aß)42 aggregates. Latrepirdine was shown to upregulate yeast vacuolar (lysosomal) activity and promote transport of the autophagic marker (Atg8) to the vacuole. Using an in vitro green fluorescent protein (GFP) tagged Aß yeast expression system, we investigated whether latrepirdine-enhanced autophagy was associated with a reduction in levels of intracellular GFP-Aß42. GFP-Aß42 was localized into punctate patterns compared to the diffuse cytosolic pattern of GFP and the GFP-Aß42 (19:34), which does not aggregate. In the autophagy deficient mutant (Atg8Δ), GFP-Aß42 showed a more diffuse cytosolic localization, reflecting the inability of this mutant to sequester GFP-Aß42. Similar to rapamycin, we observed that latrepirdine significantly reduced GFP-Aß42 in wild-type compared to the Atg8Δ mutant. Further, latrepirdine treatment attenuated Aß42-induced toxicity in wild-type cells but not in the Atg8Δ mutant. Together, our findings provide evidence for a novel mechanism of action for latrepirdine in inducing autophagy and reducing intracellular levels of GFP-Aß42.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Autofagia/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Indóis/farmacologia , Líquido Intracelular/metabolismo , Fragmentos de Peptídeos/antagonistas & inibidores , Saccharomyces cerevisiae/metabolismo , Peptídeos beta-Amiloides/metabolismo , Autofagia/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Proteínas de Fluorescência Verde/antagonistas & inibidores , Humanos , Líquido Intracelular/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
7.
Proc Natl Acad Sci U S A ; 108(43): 17797-802, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22006323

RESUMO

A heterologously expressed form of the human Parkinson disease-associated protein α-synuclein with a 10-residue N-terminal extension is shown to form a stable tetramer in the absence of lipid bilayers or micelles. Sequential NMR assignments, intramonomer nuclear Overhauser effects, and circular dichroism spectra are consistent with transient formation of α-helices in the first 100 N-terminal residues of the 140-residue α-synuclein sequence. Total phosphorus analysis indicates that phospholipids are not associated with the tetramer as isolated, and chemical cross-linking experiments confirm that the tetramer is the highest-order oligomer present at NMR sample concentrations. Image reconstruction from electron micrographs indicates that a symmetric oligomer is present, with three- or fourfold symmetry. Thermal unfolding experiments indicate that a hydrophobic core is present in the tetramer. A dynamic model for the tetramer structure is proposed, based on expected close association of the amphipathic central helices observed in the previously described micelle-associated "hairpin" structure of α-synuclein.


Assuntos
Modelos Moleculares , Polímeros/química , Estrutura Secundária de Proteína , alfa-Sinucleína/química , Dicroísmo Circular , Humanos , Microscopia Eletrônica , Ressonância Magnética Nuclear Biomolecular , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
8.
PLoS Biol ; 9(4): e1001052, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21541368

RESUMO

FUS/TLS is a nucleic acid binding protein that, when mutated, can cause a subset of familial amyotrophic lateral sclerosis (fALS). Although FUS/TLS is normally located predominantly in the nucleus, the pathogenic mutant forms of FUS/TLS traffic to, and form inclusions in, the cytoplasm of affected spinal motor neurons or glia. Here we report a yeast model of human FUS/TLS expression that recapitulates multiple salient features of the pathology of the disease-causing mutant proteins, including nuclear to cytoplasmic translocation, inclusion formation, and cytotoxicity. Protein domain analysis indicates that the carboxyl-terminus of FUS/TLS, where most of the ALS-associated mutations are clustered, is required but not sufficient for the toxicity of the protein. A genome-wide genetic screen using a yeast over-expression library identified five yeast DNA/RNA binding proteins, encoded by the yeast genes ECM32, NAM8, SBP1, SKO1, and VHR1, that rescue the toxicity of human FUS/TLS without changing its expression level, cytoplasmic translocation, or inclusion formation. Furthermore, hUPF1, a human homologue of ECM32, also rescues the toxicity of FUS/TLS in this model, validating the yeast model and implicating a possible insufficiency in RNA processing or the RNA quality control machinery in the mechanism of FUS/TLS mediated toxicity. Examination of the effect of FUS/TLS expression on the decay of selected mRNAs in yeast indicates that the nonsense-mediated decay pathway is probably not the major determinant of either toxicity or suppression.


Assuntos
DNA Helicases/metabolismo , Proteína FUS de Ligação a RNA/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Transativadores/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Núcleo Celular/genética , Citoplasma/genética , Citoplasma/metabolismo , DNA Helicases/genética , Regulação da Expressão Gênica , Mutação , Neurônios/metabolismo , RNA Helicases , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
9.
Proc Natl Acad Sci U S A ; 107(39): 16970-5, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20837543

RESUMO

The full complement of molecular pathways contributing to the pathogenesis of Parkinson disease (PD) remains unknown. Here we address this issue by taking a broad approach, beginning by using functional MRI to identify brainstem regions differentially affected and resistant to the disease. Relying on these imaging findings, we then profiled gene expression levels from postmortem brainstem regions, identifying a disease-related decrease in the expression of the catabolic polyamine enzyme spermidine/spermine N1-acetyltransferase 1 (SAT1). Next, a range of studies were completed to support the pathogenicity of this finding. First, to test for a causal link between polyamines and α-synuclein toxicity, we investigated a yeast model expressing α-synuclein. Polyamines were found to enhance the toxicity of α-synuclein, and an unbiased genome-wide screen for modifiers of α-synuclein toxicity identified Tpo4, a member of a family of proteins responsible for polyamine transport. Second, to test for a causal link between SAT1 activity and PD histopathology, we investigated a mouse model expressing α-synuclein. DENSPM (N1, N11-diethylnorspermine), a polyamine analog that increases SAT1 activity, was found to reduce PD histopathology, whereas Berenil (diminazene aceturate), a pharmacological agent that reduces SAT1 activity, worsened the histopathology. Third, to test for a genetic link, we sequenced the SAT1 gene and a rare but unique disease-associated variant was identified. Taken together, the findings from human patients, yeast, and a mouse model implicate the polyamine pathway in PD pathogenesis.


Assuntos
Acetiltransferases/metabolismo , Tronco Encefálico/metabolismo , Doença de Parkinson/metabolismo , Poliaminas/metabolismo , alfa-Sinucleína/metabolismo , Acetiltransferases/genética , Animais , Tronco Encefálico/patologia , Diminazena/análogos & derivados , Diminazena/farmacologia , Variação Genética , Humanos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Transgênicos , Doença de Parkinson/genética , Doença de Parkinson/patologia , Pemolina/análogos & derivados , Pemolina/farmacologia
10.
J Comput Aided Mol Des ; 23(8): 491-500, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19521672

RESUMO

The identification of hot spots, i.e., binding regions that contribute substantially to the free energy of ligand binding, is a critical step for structure-based drug design. Here we present the application of two fragment-based methods to the detection of hot spots for DJ-1 and glucocerebrosidase (GCase), targets for the development of therapeutics for Parkinson's and Gaucher's diseases, respectively. While the structures of these two proteins are known, binding information is lacking. In this study we employ the experimental multiple solvent crystal structures (MSCS) method and computational fragment mapping (FTMap) to identify regions suitable for the development of pharmacological chaperones for DJ-1 and GCase. Comparison of data derived via MSCS and FTMap also shows that FTMap, a computational method for the identification of fragment binding hot spots, is an accurate and robust alternative to the performance of expensive and difficult crystallographic experiments.


Assuntos
Descoberta de Drogas , Glucosilceramidase/química , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas Oncogênicas/química , Bibliotecas de Moléculas Pequenas/química , Sítios de Ligação , Cristalografia por Raios X , Doença de Gaucher/tratamento farmacológico , Humanos , Ligantes , Proteínas de Membrana/química , Terapia de Alvo Molecular , Doença de Parkinson/tratamento farmacológico , Ligação Proteica , Conformação Proteica , Proteína Desglicase DJ-1 , Bibliotecas de Moléculas Pequenas/uso terapêutico , Solventes/química , Propriedades de Superfície
11.
Mol Microbiol ; 63(4): 1248-58, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17257308

RESUMO

Studies have shown that the inositol biosynthetic pathway and the enzyme glycogen synthase kinase-3 (GSK-3) are targets of the mood-stabilizing drugs lithium and valproate. However, a relationship between these targets has not been previously described. We hypothesized that GSK-3 may play a role in inositol synthesis, and that loss of GSK-3 may lead to inositol depletion, thus providing a mechanistic link between the two drug targets. Utilizing a yeast Saccharomyces cerevisiae gsk-3Delta quadruple-null mutant, in which all four genes encoding homologues of mammalian GSK-3 are disrupted, we tested the hypothesis that GSK-3 is required for de novo inositol biosynthesis. The gsk-3Delta mutant exhibited multiple features of inositol depletion, including defective growth in inositol-lacking medium, decreased intracellular inositol, increased INO1 and ITR1 expression, and decreased levels of phosphatidylinositol. Treatment of wild-type cells with a highly specific GSK-3 inhibitor led to a significant increase in INO1 expression. Supplementation with inositol alleviated the temperature sensitivity of gsk-3Delta. Activity of myo-inositol-3 phosphate synthase, the rate-limiting enzyme in inositol de novo biosynthesis, was decreased in gsk-3Delta. These results demonstrate for the first time that GSK-3 is required for optimal myo-inositol-3 phosphate synthase activity and de novo inositol biosynthesis, and that loss of GSK-3 activity causes inositol depletion.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Inositol/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação Fúngica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Inositol/farmacologia , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Transporte de Monossacarídeos , Mutação , Mio-Inositol-1-Fosfato Sintase/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Temperatura , Tiadiazóis/farmacologia
12.
J Biol Chem ; 280(51): 41805-10, 2005 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-16221686

RESUMO

In a genetic screen for Saccharomyces cerevisiae mutants hypersensitive to the inositol-depleting drugs lithium and valproate, a loss of function allele of TPI1 was identified. The TPI1 gene encodes triose phosphate isomerase, which catalyzes the interconversion of dihydroxyacetone phosphate (DHAP) and glyceraldehyde 3-phosphate. A single mutation (N65K) in tpi1 completely abolished Tpi1p enzyme activity and led to a 30-fold increase in the intracellular DHAP concentration. The tpi1 mutant was unable to grow in the absence of inositol and exhibited the "inositol-less death" phenotype. Similarly, the pgk1 mutant, which accumulates DHAP as a result of defective conversion of 3-phosphoglyceroyl phosphate to 3-phosphoglycerate, exhibited inositol auxotrophy. DHAP as well as glyceraldehyde 3-phosphate and oxaloacetate inhibited activity of both yeast and human myo-inositol-3 phosphate synthase, the rate-limiting enzyme in de novo inositol biosynthesis. Implications for the pathology associated with TPI deficiency and responsiveness to inositol-depleting anti-bipolar drugs are discussed. This study is the first to establish a connection between perturbation of glycolysis and inhibition of de novo inositol biosynthesis.


Assuntos
Inositol/biossíntese , Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Glicólise , Lítio/farmacologia , Dados de Sequência Molecular , Mutação , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Homologia de Sequência de Aminoácidos , Triose-Fosfato Isomerase/química , Triose-Fosfato Isomerase/genética , Triose-Fosfato Isomerase/metabolismo , Ácido Valproico/farmacologia
13.
J Biol Chem ; 279(21): 21759-65, 2004 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-15024000

RESUMO

We have cloned, sequenced, and expressed a human cDNA encoding 1-d-myo-inositol-3-phosphate (MIP) synthase (hINO1). The encoded 62-kDa human enzyme converted d-glucose 6-phosphate to 1-d-myo-inositol 3-phosphate, the rate-limiting step for de novo inositol biosynthesis. Activity of the recombinant human MIP synthase purified from Escherichia coli was optimal at pH 8.0 at 37 degrees C and exhibited K(m) values of 0.57 mm and 8 microm for glucose 6-phosphate and NAD(+), respectively. NH(4)(+) and K(+) were better activators than other cations tested (Na(+), Li(+), Mg(2+), Mn(2+)), and Zn(2+) strongly inhibited activity. Expression of the protein in the yeast ino1Delta mutant lacking MIP synthase (ino1Delta/hINO1) complemented the inositol auxotrophy of the mutant and led to inositol excretion. MIP synthase activity and intracellular inositol were decreased about 35 and 25%, respectively, when ino1Delta/hINO1 was grown in the presence of a therapeutically relevant concentration of the anti-bipolar drug valproate (0.6 mm). However, in vitro activity of purified MIP synthase was not inhibited by valproate at this concentration, suggesting that inhibition by the drug is indirect. Because inositol metabolism may play a key role in the etiology and treatment of bipolar illness, functional conservation of the key enzyme in inositol biosynthesis underscores the power of the yeast model in studies of this disorder.


Assuntos
Liases Intramoleculares/fisiologia , Sequência de Aminoácidos , Western Blotting , Cátions , Clonagem Molecular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Escherichia coli/enzimologia , Humanos , Concentração de Íons de Hidrogênio , Inositol/química , Liases Intramoleculares/química , Cinética , Dados de Sequência Molecular , Mutação , Proteínas Recombinantes/química , Saccharomyces cerevisiae/enzimologia , Homologia de Sequência de Aminoácidos , Temperatura , Fatores de Tempo , Ácido Valproico/farmacologia
14.
Mol Microbiol ; 49(6): 1595-603, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12950923

RESUMO

Valproate (VPA) is one of the two drugs approved by the Food and Drug Administration (FDA) for the treatment of bipolar disorder. The therapeutic mechanism of VPA has not been established. We have shown previously that growth of the yeast Saccharomyces cerevisiae in the presence of VPA causes a decrease in intracellular inositol and inositol-1-P, and a dramatic increase in expression of INO1, which encodes the rate limiting enzyme for de novo inositol biosynthesis. To understand the underlying mechanism of action of VPA, INO1, CHO1 and INO2 expression, intracellular inositol and phospholipid biosynthesis were studied as a function of acute and chronic exposure of growing cells to the drug. A decrease in intracellular inositol was apparent immediately after addition of VPA. Surprisingly, expression of genes that are usually derepressed during inositol depletion, including INO1, CHO1 and INO2 (that contain inositol-responsive UASINO sequences) decreased several fold during the first hour, after which expression began to increase. Incorporation of 32Pi into total phospholipids was significantly decreased. Pulse labelling of CDP-DG and PG, shown previously to increase during inositol depletion, increased within 30 min. However, pulse labelling of PS, which normally increases during inositol depletion, was decreased within 30 min. PS synthase activity in cell extracts decreased with time, although VPA did not directly inhibit PS synthase enzyme activity. Thus, in contrast to the effect of chronic VPA treatment, short-term exposure to VPA abrogated the normal response to inositol depletion of inositol responsive genes and led to aberrant synthesis of phospholipids.


Assuntos
Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Inositol/metabolismo , Fosfolipídeos/biossíntese , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ácido Valproico/farmacologia , Regiões 5' não Traduzidas/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Northern Blotting , CDPdiacilglicerol-Serina O-Fosfatidiltransferase/metabolismo , Divisão Celular/efeitos dos fármacos , Eletroforese em Gel de Ágar , Mio-Inositol-1-Fosfato Sintase/genética , Mio-Inositol-1-Fosfato Sintase/metabolismo , Hibridização de Ácido Nucleico , Fosfolipídeos/genética , Proteínas Repressoras/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...