Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Rapid Commun Mass Spectrom ; 37(2): e9423, 2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36278931

RESUMO

RATIONALE: Febuxostat (FEB) is a xanthine oxidase inhibitor approved by the U.S. Food and Drug Administration for long-term treatment of gout and hyperuricemia. There were no reports on identification and characterization of stress degradation products of the drug. METHODS: FEB was subjected to forced decomposition conditions such as hydrolysis (neutral, acidic, and alkaline), oxidation, photolysis, and thermal stress, per the ICH guideline Q1A(R2). The degradation products formed were subjected to ultra-performance liquid chromatography (UPLC) on a C18 Kinetex column (100 × 4.6 mm, 2.6 µm) using isocratic elution method. Detection wavelength was 317 nm. The developed method was extended to UPLC-mass spectrometry/time of flight (MS/TOF) studies to identify and characterize the degradation products. RESULTS: The drug exhibited significant degradation under alkaline/neutral hydrolytic, alkaline/acidic photolytic, and oxidative conditions, whereas it remained stable under acid hydrolytic, neutral photolytic, and thermal conditions. In total, eight degradation products (I-VIII) were formed, which could be adequately determined from the drug using the developed UPLC method. Of the eight degradation products identified from the liquid chromatography-ultraviolet (LC-UV) chromatogram, five (III and IV and VI-VIII) could be characterized using their MS/TOF spectral data. The degradation pathway leading to the formation of the products was postulated, and this is not reported so far. CONCLUSIONS: Forced degradation studies were conducted on FEB, and the degradation products produced were identified by their mass spectral data obtained using LC-MS studies.


Assuntos
Febuxostat , Cromatografia Líquida/métodos , Espectrometria de Massas/métodos , Hidrólise , Fotólise , Oxirredução , Preparações Farmacêuticas , Estabilidade de Medicamentos , Cromatografia Líquida de Alta Pressão/métodos
2.
Hum Mol Genet ; 28(14): 2365-2377, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31267131

RESUMO

MEGF10 myopathy is a rare inherited muscle disease that is named after the causative gene, MEGF10. The classic phenotype, early onset myopathy, areflexia, respiratory distress and dysphagia, is severe and immediately life-threatening. There are no disease-modifying therapies. We performed a small molecule screen and follow-up studies to seek a novel therapy. A primary in vitro drug screen assessed cellular proliferation patterns in Megf10-deficient myoblasts. Secondary evaluations were performed on primary screen hits using myoblasts derived from Megf10-/- mice, induced pluripotent stem cell-derived myoblasts from MEGF10 myopathy patients, mutant Drosophila that are deficient in the homologue of MEGF10 (Drpr) and megf10 mutant zebrafish. The screen yielded two promising candidates that are both selective serotonin reuptake inhibitors (SSRIs), sertraline and escitalopram. In depth follow-up analyses demonstrated that sertraline was highly effective in alleviating abnormalities across multiple models of the disease including mouse myoblast, human myoblast, Drosophila and zebrafish models. Sertraline also restored deficiencies of Notch1 in disease models. We conclude that SSRIs show promise as potential therapeutic compounds for MEGF10 myopathy, especially sertraline. The mechanism of action may involve the Notch pathway.


Assuntos
Proteínas de Membrana/genética , Doenças Musculares/tratamento farmacológico , Mioblastos/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Sertralina/uso terapêutico , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Citalopram/farmacologia , Citalopram/uso terapêutico , Drosophila/efeitos dos fármacos , Drosophila/genética , Avaliação Pré-Clínica de Medicamentos , Humanos , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Doenças Musculares/genética , Mutação , Mioblastos/metabolismo , Receptor Notch1/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sertralina/farmacologia , Transdução de Sinais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
3.
Muscle Nerve ; 63(6): 928-940, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33651408

RESUMO

INTRODUCTION: RNA-binding proteins (RBPs) play an important role in skeletal muscle development and disease by regulating RNA splicing. In myotonic dystrophy type 1 (DM1), the RBP MBNL1 (muscleblind-like) is sequestered by toxic CUG repeats, leading to missplicing of MBNL1 targets. Mounting evidence from the literature has implicated other factors in the pathogenesis of DM1. Herein we sought to evaluate the functional role of the splicing factor hnRNP L in normal and DM1 muscle cells. METHODS: Co-immunoprecipitation assays using hnRNPL and MBNL1 expression constructs and splicing profiling in normal and DM1 muscle cell lines were performed. Zebrafish morpholinos targeting hnrpl and hnrnpl2 were injected into one-cell zebrafish for developmental and muscle analysis. In human myoblasts downregulation of hnRNP L was achieved with shRNAi. Ascochlorin administration to DM1 myoblasts was performed and expression of the CUG repeats, DM1 splicing biomarkers, and hnRNP L expression levels were evaluated. RESULTS: Using DM1 patient myoblast cell lines we observed the formation of abnormal hnRNP L nuclear foci within and outside the expanded CUG repeats, suggesting a role for this factor in DM1 pathology. We showed that the antiviral and antitumorigenic isoprenoid compound ascochlorin increased MBNL1 and hnRNP L expression levels. Drug treatment of DM1 muscle cells with ascochlorin partially rescued missplicing of established early biomarkers of DM1 and improved the defective myotube formation displayed by DM1 muscle cells. DISCUSSION: Together, these studies revealed that hnRNP L can modulate DM1 pathologies and is a potential therapeutic target.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Desenvolvimento Muscular/genética , Mioblastos/metabolismo , Distrofia Miotônica/genética , Adulto , Animais , Linhagem Celular , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Masculino , Pessoa de Meia-Idade , Mioblastos/patologia , Distrofia Miotônica/metabolismo , Distrofia Miotônica/patologia , Peixe-Zebra
4.
Hum Mol Genet ; 26(15): 2984-3000, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28498977

RESUMO

Mutations in MEGF10 cause early onset myopathy, areflexia, respiratory distress, and dysphagia (EMARDD), a rare congenital muscle disease, but the pathogenic mechanisms remain largely unknown. We demonstrate that short hairpin RNA (shRNA)-mediated knockdown of Megf10, as well as overexpression of the pathogenic human p.C774R mutation, leads to impaired proliferation and migration of C2C12 cells. Myoblasts from Megf10-/- mice and Megf10-/-/mdx double knockout (dko) mice also show impaired proliferation and migration compared to myoblasts from wild type and mdx mice, whereas the dko mice show histological abnormalities that are not observed in either single mutant mouse. Cell proliferation and migration are known to be regulated by the Notch receptor, which plays an essential role in myogenesis. Reciprocal co-immunoprecipitation studies show that Megf10 and Notch1 interact via their respective intracellular domains. These interactions are impaired by the pathogenic p.C774R mutation. Megf10 regulation of myoblast function appears to be mediated at least in part via interactions with key components of the Notch signaling pathway, and defects in these interactions may contribute to the pathogenesis of EMARDD.


Assuntos
Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Receptor Notch1/metabolismo , Animais , Movimento Celular , Proliferação de Células , Camundongos , Camundongos Endogâmicos mdx , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Doenças Musculares/genética , Mioblastos/metabolismo , Mioblastos/fisiologia , Receptor Notch1/genética , Transdução de Sinais
5.
Physiol Genomics ; 50(11): 929-939, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30345904

RESUMO

Next-generation sequencing is commonly used to screen for pathogenic mutations in families with Mendelian disorders, but due to the pace of discoveries, gaps have widened for some diseases between genetic and pathophysiological knowledge. We recruited and analyzed 16 families with limb-girdle muscular dystrophy (LGMD) of Arab descent from Saudi Arabia and Sudan who did not have confirmed genetic diagnoses. The analysis included both traditional and next-generation sequencing approaches. Cellular and metabolic studies were performed on Pyroxd1 siRNA C2C12 myoblasts and controls. Pathogenic mutations were identified in eight of the 16 families. One Sudanese family of Arab descent residing in Saudi Arabia harbored a homozygous c.464A>G, p.Asn155Ser mutation in PYROXD1, a gene recently reported in association with myofibrillar myopathy and whose protein product reduces thiol residues. Pyroxd1 deficiency in murine C2C12 myoblasts yielded evidence for impairments of cellular proliferation, migration, and differentiation, while CG10721 (Pyroxd1 fly homolog) knockdown in Drosophila yielded a lethal phenotype. Further investigations indicated that Pyroxd1 does not localize to mitochondria, yet Pyroxd1 deficiency is associated with decreased cellular respiration. This study identified pathogenic mutations in half of the LGMD families from the cohort, including one in PYROXD1. Developmental impairments were demonstrated in vitro for Pyroxd1 deficiency and in vivo for CG10721 deficiency, with reduced metabolic activity in vitro for Pyroxd1 deficiency.


Assuntos
Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Adulto , Animais , Animais Geneticamente Modificados , Respiração Celular/genética , Células Cultivadas , Drosophila , Proteínas de Drosophila/genética , Feminino , Humanos , Masculino , Camundongos , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Distrofia Muscular do Cíngulo dos Membros/patologia , Mioblastos/patologia , Linhagem , Arábia Saudita , Sudão
7.
J Neurosci ; 33(43): 17182-7, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-24155322

RESUMO

The mechanisms that drive the normal resolution of acute postoperative pain are not completely understood. We hypothesize a pivotal role of a major spinal mitogen-activated protein kinase (MAPKs) regulator, MAPK phosphatase (MKP)-3, in the resolution of postoperative pain. We used wild-type and MKP-3 knock-out (KO) mice, a paw incision model of acute postoperative pain, and behavioral and molecular biology experiments. We observed persistent mechanical allodynia in mice lacking MKP-3 (postoperative day 21), concurrently with persistent phosphorylation of spinal p38 and extracellular signal-regulated kinases (ERK)-1/2 on postoperative day 12, while both MAPK phosphorylation and allodynia resolved on postoperative day 7 in wild-type mice. Spinal p-ERK was expressed mainly in neurons and microglia, while spinal p-p38 was expressed mostly in microglia in MKP-3 KO mice, and their selective pharmacological inhibition reduced the persistent allodynia observed in these mice. Our findings strongly suggest that dysregulation of MKP-3 prevents spontaneous resolution of acute postoperative pain and drives its transition to persistent pain via persistent neuronal and microglial MAPK phosphorylation in the spinal cord.


Assuntos
Dor Aguda/enzimologia , Fosfatase 6 de Especificidade Dupla/metabolismo , Hiperalgesia/enzimologia , Dor Pós-Operatória/enzimologia , Animais , Modelos Animais de Doenças , Fosfatase 6 de Especificidade Dupla/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Microglia/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/enzimologia , Fosforilação , Medula Espinal/enzimologia , Medula Espinal/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Biochem J ; 447(1): 159-66, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22827337

RESUMO

The extent and duration of MAPK (mitogen-activated protein kinase) signalling govern a diversity of normal and aberrant cellular outcomes. Genetic and pharmacological disruption of the MAPK-activated kinase RSK (ribosomal S6 kinase) leads to elevated MAPK activity indicative of a RSK-dependent negative feedback loop. Using biochemical, pharmacological and quantitative MS approaches we show that RSK phosphorylates the Ras activator SOS1 (Son of Sevenless homologue 1) in cultured cells on two C-terminal residues, Ser(1134) and Ser(1161). Furthermore, we find that RSK-dependent SOS1 phosphorylation creates 14-3-3-binding sites. We show that mutating Ser(1134) and Ser(1161) disrupts 14-3-3 binding and modestly increases and extends MAPK activation. Together these data suggest that one mechanism whereby RSK negatively regulates MAPK activation is via site-specific SOS1 phosphorylation.


Assuntos
Proteínas 14-3-3/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteína SOS1/metabolismo , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células COS , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Humanos , Camundongos , Modelos Biológicos , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Fosforilação , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SOS1/química , Proteína SOS1/genética , Serina/química
9.
FEBS Open Bio ; 11(1): 114-123, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33159715

RESUMO

Biallelic loss-of-function MEGF10 mutations lead to MEGF10 myopathy, also known as early onset myopathy with areflexia, respiratory distress, and dysphagia (EMARDD). MEGF10 is expressed in muscle satellite cells, but the contribution of satellite cell dysfunction to MEGF10 myopathy is unclear. Myofibers and satellite cells were isolated and examined from Megf10-/- and wild-type mice. A separate set of mice underwent repeated intramuscular barium chloride injections. Megf10-/- muscle satellite cells showed reduced proliferation and migration, while Megf10-/- mouse skeletal muscles showed impaired regeneration. Megf10 deficiency is associated with impaired muscle regeneration, due in part to defects in satellite cell function. Efforts to rescue Megf10 deficiency will have therapeutic implications for MEGF10 myopathy and other inherited muscle diseases involving impaired muscle regeneration.


Assuntos
Proteínas de Membrana/deficiência , Fibras Musculares Esqueléticas/patologia , Doenças Musculares/genética , Regeneração/genética , Animais , Movimento Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Humanos , Mutação com Perda de Função , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/citologia , Doenças Musculares/patologia , Células Satélites de Músculo Esquelético/patologia
10.
FEBS Lett ; 593(7): 680-696, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30802937

RESUMO

Recessive mutations in multiple epidermal growth factor-like domains 10 (MEGF10) underlie a rare congenital muscle disease known as MEGF10 myopathy. MEGF10 and its Drosophila homolog Draper (Drpr) are transmembrane receptors expressed in muscle and glia. Drpr deficiency is known to result in muscle abnormalities in flies. In the current study, flies that ubiquitously overexpress Drpr, or mouse Megf10, display developmental arrest. The phenotype is reproduced with overexpression in muscle, but not in other tissues, and with overexpression during intermediate stages of myogenesis, but not in myoblasts. We find that tubular muscle subtypes are particularly sensitive to Megf10/Drpr overexpression. Complementary genetic analyses show that Megf10/Drpr and Notch may interact to regulate myogenesis. Our findings provide a basis for investigating MEGF10 in muscle development using Drosophila.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Membrana/genética , Desenvolvimento Muscular/genética , Doenças Musculares/genética , Animais , Proliferação de Células/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Mutação com Ganho de Função/genética , Predisposição Genética para Doença , Humanos , Camundongos , Músculo Esquelético/crescimento & desenvolvimento , Doenças Musculares/patologia , Mioblastos/metabolismo , Transdução de Sinais/genética
11.
Mol Ther Methods Clin Dev ; 13: 167-179, 2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-30788385

RESUMO

Barth syndrome (BTHS) is a rare mitochondrial disease that causes severe cardiomyopathy and has no disease-modifying therapy. It is caused by recessive mutations in the gene tafazzin (TAZ), which encodes tafazzin-an acyltransferase that remodels the inner mitochondrial membrane lipid cardiolipin. To identify novel mechanistic pathways involved in BTHS and evaluate the effects of gene therapy on proteomic profiles, we performed a multiplex tandem mass tagging (TMT) quantitative proteomics analysis to compare protein expression profiles from heart lysates isolated from BTHS, healthy wild-type (WT), and BTHS treated with adeno-associated virus serotype 9 (AAV9)-TAZ gene replacement as neonates or adults. 197 proteins with ≥2 unique peptides were identified. Of these, 91 proteins were significantly differentially expressed in BTHS compared to WT controls. Cause-effect relationships between tafazzin deficiency and altered protein profiles were confirmed through demonstrated significant improvements in expression levels following administration of AAV9-TAZ. The importance of TMEM65 in Cx43 localization to cardiac intercalated discs was revealed as a novel consequence of tafazzin deficiency that was improved following gene therapy. This study identifies novel mechanistic pathways involved in the pathophysiology of BTHS, demonstrates the ability of gene delivery to improve protein expression profiles, and provides support for clinical translation of AAV9-TAZ gene therapy.

12.
Hum Gene Ther ; 30(2): 139-154, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30070157

RESUMO

Barth syndrome (BTHS) is a rare mitochondrial disease that affects heart and skeletal muscle and has no curative treatment. It is caused by recessive mutations in the X-linked gene TAZ, which encodes tafazzin. To develop a clinically relevant gene therapy to restore tafazzin function and treat BTHS, three different adeno-associated virus serotype 9 vectors were tested and compared to identify the optimal promoter-cytomegalovirus (CMV), desmin (Des), or a native tafazzin promoter (Taz)-for TAZ expression following intravenous administration of 1 × 1013 vector genomes/kilogram to a mouse model of BTHS as either neonates (1-2 days of age) or adults (3 months of age). At 5 months of age, evaluations of biodistribution and TAZ expression levels, mouse activity assessments, fatigue in response to exercise, muscle strength, cardiac function, mitochondrial structure, oxygen consumption, and electron transport chain complex activity assays were performed to measure the extent of improvement in treated mice. Each promoter was scored for significant improvement over untreated control mice and significant improvement compared with the other two promoters for every measurement and within each age of administration. All three of the promoters resulted in significant improvements in a majority of the assessments compared with untreated BTHS controls. When scored for overall effectiveness as a gene therapy, the Des promoter was found to provide improvement in the most assessments, followed by the CMV promoter, and finally Taz regardless of injection age. This study provides substantial support for translation of an adeno-associated virus serotype 9-mediated TAZ gene replacement strategy using a Des promoter for human BTHS patients in the clinic.


Assuntos
Síndrome de Barth , Dependovirus , Terapia Genética , Vetores Genéticos , Fatores de Transcrição , Transdução Genética , Aciltransferases , Animais , Síndrome de Barth/genética , Síndrome de Barth/metabolismo , Síndrome de Barth/fisiopatologia , Síndrome de Barth/terapia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Recuperação de Função Fisiológica/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
13.
J Pain Res ; 10: 763-774, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28405172

RESUMO

Mitogen-activated protein kinase (MAPK) phosphatase-3 (MKP-3) and its substrates (extracellular signal-regulated kinase [ERK] and p38) play an important role in pathophysiological mechanisms of acute postoperative and chronic neuropathic pain in the spinal cord. This study aimed to understand the role of MKP-3 and its target MAPKs at the site of surgical incision in nociceptive behavior. Wild-type (WT) and MKP-3 knockout (KO) mice underwent unilateral plantar hind paw incision. Mechanical allodynia was assessed by using von Frey filaments. Peripheral ERK-1/2 and p38 phosphorylation were measured by Western blot. Cell infiltration was determined using hematoxylin and eosin histological staining. Peripheral phosphorylated ERK-1/2 (p-ERK-1/2) inhibition was performed in MKP-3 KO mice. In WT mice, mechanical hypersensitivity was observed on postoperative day 1 (0.69±0.17 g baseline vs 0.13±0.08 g day 1), which resolved normally by postoperative day 12 (0.46±0.08 g, N=6). In MKP-3 KO mice, this hypersensitivity persisted at least 12 days after surgery (0.19±0.06 g; N=6). KO mice displayed higher numbers of infiltrating cells (51.4±6 cells/0.1 mm2) than WT mice (8.7±1.2 cells/0.1 mm2) on postoperative day 1 (vs 5-6 cells/0.1 mm2 at baseline) that returned to baseline 12 days after surgery (10-12 cells/0.1 mm2). In WT mice, peripheral p-p38 and p-ERK-1/2 expression increased (5- and 3-fold, respectively) on postoperative days 1 and 5, and returned to basal levels 7-12 days after surgery (N=3 per group). Peripheral p-p38 levels in MKP-3 KO mice followed a similar expression pattern as WT mice. Peripheral p-ERK-1/2 levels in MKP-3 KO mice remained elevated 12 days after surgery (2.5-fold, N=3 per group). Administration of PD98059 (MEK inhibitor, N=8, vehicle N=9) reduced p-ERK-1/2 expression in the incised tissue and blocked hypersensitivity in MKP-3 KO mice (N=6). The findings of this study suggest that MKP-3 is pivotal for normal resolution of acute postoperative allodynia, through the regulation of peripheral p-ERK-1/2.

14.
Neurosci Lett ; 575: 13-8, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24861519

RESUMO

Mitogen-activated protein kinases (MAPKs) play critical roles in the central nervous system immune responses through glial function, which are regulated with relative selectivity (or preference) by MAPK phosphatases (MKP). Phosphorylated extracellular signal-regulated protein kinase (p-ERK) is preferentially dephosphorylated by MKP-3, which display little activity over p-p38 and p-c-Jun NH2-terminal kinases (p-JNK). It has been proposed that these substrate preferences may vary depending on tissue or functional cellular processes. Since astrocytes display a prominent activity of JNK>ERK under stressed or reactive phenotype, we hypothesize that MKP-3 possess a similar or differential substrate preference in astrocytes for JNK and ERK (ERK=JNK or JNK>ERK). We generated transient expression of MKP-3 by transfecting a specific cDNA in primary rat neonatal brain cortex astrocytes. Cells were stimulated with lipopolysaccharide (LPS), and MAPKs and downstream pro-inflammatory products were measured by Western blot and ELISA analyses. MKP-3 expression in primary astrocytes reduced LPS-induced p-ERK and p-p38 by ∼50%, and p-JNK by ∼75%, and moderately reduced nitrite oxide (NO), while completely blocked Interleukin (IL)-6 and tumor necrosis factor alpha (TNFα). We confirmed MKP-3 specific activity by developing a BV-2 microglia cell line stably overexpressing MKP-3 and using a specific siRNA against MKP-3. Our data demonstrate MKP-3 has differential substrate preference in astrocytes compared to other cells types, since it preferentially dephosphorylated p-JNK over p-ERK. Our results indicate also that astrocytic immune functions can be modulated by MKP-3 induction, a strategy that could be beneficial in neurological conditions in which astrocytes play a pathophysiological role, i.e. persistent pain.


Assuntos
Astrócitos/metabolismo , Fosfatase 6 de Especificidade Dupla/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Fosfatase 6 de Especificidade Dupla/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Fosforilação , Ratos Sprague-Dawley , Especificidade por Substrato
15.
J Proteomics ; 78: 113-22, 2013 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-23146917

RESUMO

Channels, pumps, receptors, cyclases and other membrane proteins modulate the motility and sensory function of cilia, but these proteins are generally under-represented in proteomic analyses of cilia. Studies of these ciliary membrane proteins would benefit from a protocol to greatly enrich for integral and lipidated membrane proteins. We used LC-MS/MS to compare the proteomes of unfractionated cilia (C), the ciliary membrane (CM) and the ciliary membrane in the detergent phase (DP) of Triton X-114 phase separation. 55% of the proteins in DP were membrane proteins (i.e. predicted transmembrane or membrane-associated through lipid modifications) and 31% were transmembrane. This is to be compared to 23% membrane proteins with 9% transmembrane in CM and 9% membrane proteins with 3% transmembrane in C. 78% of the transmembrane proteins in the DP were found uniquely in DP, and not in C or CM. There were ion channels, cyclases, plasma membrane pumps, Ca(2+) dependent protein kinases, and Rab GTPases involved in the signal transduction in DP that were not identified in the other C and CM preparations. Of 267 proteins unique to the DP, 147 were novel, i.e. not found in other proteomic and genomic studies of cilia.


Assuntos
Proteínas de Membrana/metabolismo , Paramecium tetraurellia/metabolismo , Proteoma/metabolismo , Proteômica , Proteínas de Protozoários/metabolismo , Cílios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA