Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(31): E2925-33, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23818640

RESUMO

The human gamma herpesviruses, Kaposi sarcoma-associated virus (KSHV) and EBV, are associated with multiple cancers. Recent evidence suggests that EBV and possibly other viruses can manipulate the tumor microenvironment through the secretion of specific viral and cellular components into exosomes, small endocytically derived vesicles that are released from cells. Exosomes produced by EBV-infected nasopharyngeal carcinoma cells contain high levels of the viral oncogene latent membrane protein 1 and viral microRNAs that activate critical signaling pathways in recipient cells. In this study, to determine the effects of EBV and KSHV on exosome content, quantitative proteomics techniques were performed on exosomes purified from 11 B-cell lines that are uninfected, infected with EBV or with KSHV, or infected with both viruses. Using mass spectrometry, 871 proteins were identified, of which ∼360 were unique to the viral exosomes. Analysis by 2D difference gel electrophoresis and spectral counting identified multiple significant changes compared with the uninfected control cells and between viral groups. These data predict that both EBV and KSHV exosomes likely modulate cell death and survival, ribosome function, protein synthesis, and mammalian target of rapamycin signaling. Distinct viral-specific effects on exosomes suggest that KSHV exosomes would affect cellular metabolism, whereas EBV exosomes would activate cellular signaling mediated through integrins, actin, IFN, and NFκB. The changes in exosome content identified in this study suggest ways that these oncogenic viruses modulate the tumor microenvironment and may provide diagnostic markers specific for EBV and KSHV associated malignancies.


Assuntos
Linfócitos B/metabolismo , Infecções por Vírus Epstein-Barr/metabolismo , Exossomos/metabolismo , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas de Neoplasias/metabolismo , Sarcoma de Kaposi/metabolismo , Linfócitos B/patologia , Morte Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular , Infecções por Vírus Epstein-Barr/diagnóstico , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Exossomos/patologia , Exossomos/virologia , Humanos , Biossíntese de Proteínas , Sarcoma de Kaposi/diagnóstico , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais
2.
FASEB J ; 28(8): 3769-79, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24812087

RESUMO

Impaired adipogenesis renders an adipose tissue unable to expand, leading to lipotoxicity and conditions such as diabetes and cardiovascular disease. While factors important for adipogenesis have been studied extensively, those that set the limits of adipose tissue expansion remain undetermined. Feeding a Western-type diet to apolipoprotein E2 knock-in mice, a model of metabolic syndrome, produced 3 groups of equally obese mice: mice with normal glucose tolerance, hyperinsulinemic yet glucose-tolerant mice, and prediabetic mice with impaired glucose tolerance and reduced circulating insulin. Using proteomics, we compared subcutaneous adipose tissues from mice in these groups and found that the expression of PTRF (polymerase I and transcript release factor) associated selectively with their glucose tolerance status. Lentiviral and pharmacologically overexpressed PTRF, whose function is critical for caveola formation, compromised adipocyte differentiation of cultured 3T3-L1cells. In human adipose tissue, PTRF mRNA levels positively correlated with markers of lipolysis and cellular senescence. Furthermore, a negative relationship between telomere length and PTRF mRNA levels was observed in human subcutaneous fat. PTRF is associated with limited adipose tissue expansion underpinning the key role of caveolae in adipocyte regulation. Furthermore, PTRF may be a suitable adipocyte marker for predicting pathological obesity and inform clinical management.


Assuntos
Adipócitos/patologia , Adipogenia/fisiologia , Cavéolas/fisiologia , Dieta/efeitos adversos , Intolerância à Glucose/etiologia , Hiperinsulinismo/etiologia , Obesidade/etiologia , Estado Pré-Diabético/etiologia , Proteínas de Ligação a RNA/fisiologia , Gordura Subcutânea/metabolismo , Células 3T3-L1 , Adiponectina/sangue , Animais , Aorta/patologia , Apolipoproteína E2/genética , Senescência Celular , Feminino , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Intolerância à Glucose/sangue , Intolerância à Glucose/patologia , Humanos , Hiperinsulinismo/sangue , Hiperinsulinismo/patologia , Resistência à Insulina , Lipólise , Fígado/química , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/classificação , Obesidade/patologia , Estado Pré-Diabético/sangue , Estado Pré-Diabético/patologia , Gravidez , RNA Mensageiro/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Gordura Subcutânea/patologia , Encurtamento do Telômero , Triglicerídeos/metabolismo
3.
Adv Exp Med Biol ; 806: 525-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24952201

RESUMO

Mass spectrometry (MS) has been increasingly used to study central nervous system disorders, including autism spectrum disorders (ASDs). The first studies of ASD using MS focused on the identification of external toxins, but current research is more directed at understanding endogenous protein changes that occur in ASD (ASD proteomics). This chapter focuses on how MS has been used to study ASDs, with particular focus on proteomic analysis. Other neurodevelopmental disorders have been investigated using this technique, including genetic syndromes associated with autism such as fragile X syndrome and Smith-Lemli-Opitz syndrome.


Assuntos
Transtorno Autístico/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Espectrometria de Massas/métodos , Proteômica/métodos , Síndrome de Smith-Lemli-Opitz/metabolismo , Animais , Humanos
4.
Physiol Genomics ; 45(7): 243-55, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23386204

RESUMO

We have previously identified inbred rat strains differing in survival time to a severe controlled hemorrhage (StaH). In efforts to identify cellular mechanisms and ultimately genes that are important contributors to enhanced STaH, we conducted a study to characterize potential differences in cardiac mitochondrial proteins in these rats. Inbred rats from three strains [Brown Norway/Medical College of Wisconsin (BN); Dark Agouti (DA), and Fawn Hooded Hypertensive (FHH)] with different StaH (DA = FHH > BN) were assigned to one of three treatment groups (n = 4/strain): nonoperated controls, surgically catheterized rats, or rats surgically catheterized and hemorrhaged 24 h postsurgery. Rats were euthanized 30 min after handling or 30 min after initiation of a 26 min hemorrhage. After euthanasia, hearts were removed and mitochondria isolated. Differential protein expression was determined using 2D DIGE-based Quantitative Intact Proteomics and proteins identified by MALDI/TOF mass spectrometry. Hundreds of proteins (791) differed among inbred rat strains (P ≤ 0.038), and of these 81 were identified. Thirty-eight were unique proteins and 43 were apparent isoforms. For DA rats (longest STaH), 36 proteins increased and 30 decreased compared with BN (shortest STaH). These 81 proteins were associated with lipid (e.g., acyl CoA dehydrogenase) and carbohydrate (e.g., fumarase) metabolism, oxidative phosphorylation (e.g., ubiquinol-cytochrome C reductase), ATP synthesis (F1 ATPase), and H2S synthesis (3-mercaptopyruvate sulfurtransferase). Although we cannot make associations between these identified mitochondrial proteins and StaH, our data do provide evidence for future candidate proteins with which to consider such associations.


Assuntos
Hemorragia/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteoma/análise , Animais , Masculino , Mitocôndrias Cardíacas/química , Proteínas Mitocondriais/análise , Proteínas Mitocondriais/metabolismo , Proteoma/metabolismo , Proteômica , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos , Fatores de Tempo , Eletroforese em Gel Diferencial Bidimensional
5.
Electrophoresis ; 33(24): 3745-55, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23161654

RESUMO

Apolipoprotein E (ApoE) is a major lipid carrier protein. In humans, ApoE is expressed in three polymorphic isoforms, which are encoded by three different alleles APOE2, APOE3, and APOE4. In the brains of Alzheimer's disease (AD) patients, each one of these three allelic isoforms is found in several "isoelectric" protein isoforms (qPI), i.e. protein isoforms resulting from PTMs altering the net charge (q) of the polypeptide. AD is a complex disease in which multiple causes and several risk factors affect the onset and disease outcome. A major risk factor for AD is ApoE4; therefore, it is important to characterize the different ApoE qPIs. We have implemented a detergent-based method for isolation and quantitation of protein isoforms, and we found differences in the solubility of protein isoforms depending on the type of solvent used. In this manuscript, we describe these methods and applied them to young human-ApoE targeted replacement mice. Our results indicate that there are no significant differences in the hippocampus proteome of these mice as a function of the APOE genotype.


Assuntos
Apolipoproteína E3/biossíntese , Apolipoproteína E4/biossíntese , Proteoma/análise , Análise de Variância , Animais , Apolipoproteína E3/análise , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/análise , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Creatina Quinase/análise , Creatina Quinase/química , Creatina Quinase/metabolismo , Eletroforese em Gel Bidimensional , Genótipo , Hipocampo/química , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Fosfopiruvato Hidratase/análise , Fosfopiruvato Hidratase/química , Fosfopiruvato Hidratase/metabolismo , Isoformas de Proteínas , Proteoma/química , Proteoma/genética , Proteoma/metabolismo , Proteômica/métodos , Solubilidade
6.
J Proteome Res ; 10(4): 1632-44, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21210719

RESUMO

To better elucidate temporal changes in protein oxidation resulting from aging and the Alzheimer's disease-associated Apolipoprotein E (ApoE), we developed a 2D-DIGE-based method for simultaneously detecting differential expression and carbonyl oxidation of proteins. Specifically, we examined changes in the levels of oxidation and total protein expression in hippocampi from young-adult (25-30 weeks) and old (76-97 weeks) mice transgenic for the human Apolipoprotein E gene (APOE, APOE3, APOE4) isoforms, APOE3 or APOE4. Protein samples were labeled with either a fluorescent aminooxyacetamide (Alexa Fluor 488) to detect carbonyl modifications or with NHS-Cy3 to detect total protein expression. A protein sample used as an internal control was labeled with NHS-Cy5 and run on each gel. DIGE analysis revealed 38 differentially oxidized and 100 differentially expressed protein spots with significantly different levels (P < 0.05). For oxidized proteins, principal component analysis revealed two distinct clusters: one in which oxidation increased with age independent of APOE genotype, and the second in which oxidation was dependent on APOE genotype. For total protein expression, principal component analysis revealed a large overlap between changes with overall aging and between APOE genotypes. The use of a fluorescent tag to label oxidized proteins, in combination with a NHS-Cy3 to label total protein, makes it possible to determine changes in both protein oxidation and protein expression levels in a single experiment. These studies reveal that the expression levels of peroxiredoxin protein family members Prdx2, 3, and 6 are modified by age, APOE genotype, or both.


Assuntos
Envelhecimento/fisiologia , Apolipoproteína E3 , Apolipoproteína E4 , Genótipo , Oxirredução , Animais , Apolipoproteína E3/química , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/química , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Humanos , Espectrometria de Massas/métodos , Camundongos , Camundongos Transgênicos , Análise de Componente Principal , Eletroforese em Gel Diferencial Bidimensional/métodos
7.
mBio ; 9(3)2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921667

RESUMO

The Epstein-Barr virus (EBV) oncoproteins latent membrane protein 1 (LMP1) and LMP2A constitutively activate multiple signaling pathways, and both have been shown to interact with cellular ubiquitin ligases and affect cellular ubiquitination. To detect the LMP1- and LMP2A-mediated effects on the global cellular proteome, epithelial cell lines expressing LMP1 or LMP2A were analyzed using label-free quantitative proteomics. To identify proteins whose ubiquitination is affected by the viral proteins, the cells were cultured in the presence and absence of deubiquitinase (DUB) and proteasome inhibitors. More than 7,700 proteins were identified with high confidence and considerably more proteins showed significant differences in expression in the presence of inhibitors. Few of the differentially expressed proteins with or without inhibitors were common between LMP1 and LMP2A, confirming that the viral proteins induce unique changes in cell expression and function. However, ingenuity pathway analysis (IPA) of the data indicated that LMP1 and LMP2A modulate many of the same cellular regulatory pathways, including cell death and survival, cell movement, and actin filament dynamics. In addition, various proteasome subunits, ubiquitin-specific peptidases and conjugating enzymes, vesicle trafficking proteins, and NF-κB and mitogen-activated protein kinase signaling proteins were affected by LMP1 or LMP2A. These findings suggest that LMP1 and LMP2A may commonly target critical cell pathways through effects on distinct genes, with many cellular proteins modified by ubiquitination and/or degradation.IMPORTANCE The Epstein-Barr virus proteins latent membrane protein 1 and 2 have potent effects on cell growth and signaling. Both proteins bind to specific ubiquitin ligases and likely modulate the cellular proteome through ubiquitin-mediated effects on stability and intracellular location. In this study, a comprehensive proteomic analysis of the effects of LMP1 and LMP2A revealed that both proteins affected proteasome subunits, ubiquitin-specific conjugases and peptidases, and vesical trafficking proteins. The data suggest that the effects of these proteins on the abundance and ubiquitination of cellular proteins are in part responsible for their effects on cell growth regulation.


Assuntos
Infecções por Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/imunologia , Proteínas Oncogênicas/metabolismo , Proteômica , Proteínas da Matriz Viral/metabolismo , Morte Celular , Linhagem Celular , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Transdução de Sinais , Ubiquitinação , Proteínas de Transporte Vesicular/genética
8.
Alzheimers Res Ther ; 6(4): 43, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25478016

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is the major cause of dementia among the elderly. Finding blood-based biomarkers for disease diagnosis and prognosis is urgently needed. METHODS: We studied protein distributions in brain tissues, cerebrospinal fluid (CSF), and blood of AD patients by using proteomics and a new proteomic method that we call "2D multiplexed Western blot" (2D mxWd). This method allows us to determine in multiple samples the electrophoretic patterns of protein isoforms with different isoelectric points. RESULTS: Apolipoprotein E (ApoE) displays a unique distribution of electrophoretic isoforms in the presence of AD and also a unique pattern specific to the APOE genotype. CONCLUSIONS: The isoelectric distribution of differentially charged ApoE isoforms was used to determine the presence of AD in a small group of samples. Further studies are needed to validate their use as predictors of disease onset and progression, and as biomarkers for determining the efficacy of therapeutic treatments.

9.
Cell Biochem Biophys ; 67(1): 127-38, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23695782

RESUMO

The ubiquitin-proteasome system (UPS) plays a central role in maintaining protein homeostasis, emphasized by a myriad of diseases that are associated with altered UPS function such as cancer, muscle-wasting, and neurodegeneration. Protein ubiquitination plays a central role in both the promotion of proteasomal degradation as well as cellular signaling through regulation of the stability of transcription factors and other signaling molecules. Substrate-specificity is a critical regulatory step of ubiquitination and is mediated by ubiquitin ligases. Recent studies implicate ubiquitin ligases in multiple models of cardiac diseases such as cardiac hypertrophy, atrophy, and ischemia/reperfusion injury, both in a cardioprotective and maladaptive role. Therefore, identifying physiological substrates of cardiac ubiquitin ligases provides both mechanistic insights into heart disease as well as possible therapeutic targets. Current methods identifying substrates for ubiquitin ligases rely heavily upon non-physiologic in vitro methods, impeding the unbiased discovery of physiological substrates in relevant model systems. Here we describe a novel method for identifying ubiquitin ligase substrates utilizing tandem ubiquitin binding entities technology, two-dimensional differential in gel electrophoresis, and mass spectrometry, validated by the identification of both known and novel physiological substrates of the ubiquitin ligase MuRF1 in primary cardiomyocytes. This method can be applied to any ubiquitin ligase, both in normal and disease model systems, in order to identify relevant physiological substrates under various biological conditions, opening the door to a clearer mechanistic understanding of ubiquitin ligase function and broadening their potential as therapeutic targets.


Assuntos
Proteínas Musculares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Células Cultivadas , Chaperonina 60/metabolismo , Eletroforese em Gel Bidimensional , Proteínas Mitocondriais/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Proteínas Musculares/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ligação Proteica , Proteoma/análise , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Especificidade por Substrato , Transfecção , Proteínas com Motivo Tripartido , Tropomiosina/metabolismo , Ubiquitina-Proteína Ligases/genética
10.
Methods Mol Biol ; 854: 129-43, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22311757

RESUMO

Difference gel electrophoresis (DIGE) is most often used to assess relative changes in the expression levels of individual proteins in multiple complex samples, and this information is valuable in making inferences about relative protein activity. However, a protein's activity is not solely dependent upon its expression level. A change in activity may also be influenced by myriad posttranslational modifications (PTMs), including palmitoylation, ubiquitination, oxidation, and phosphorylation. In this chapter, we describe the use of DIGE to determine specific PTMs by introducing specific labels or changes in pI and/or molecular weight.


Assuntos
Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Proteômica/métodos , Eletroforese em Gel Diferencial Bidimensional/métodos , Métodos Analíticos de Preparação de Amostras , Focalização Isoelétrica , Lipoilação , Peso Molecular , Oxirredução , Fosforilação , Proteínas/química , Proteínas/isolamento & purificação , Coloração e Rotulagem , Especificidade por Substrato , Ubiquitinação
11.
Methods Mol Biol ; 854: 47-66, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22311753

RESUMO

Difference gel electrophoresis (DIGE) is a common technique for characterizing differential protein expression in quantitative proteomics. Usually a combination of enzymatic digestion and peptide analysis by mass spectrometry is used to identify differentially expressed proteins following separation and statistical analysis by DIGE. In this chapter, methods for gel spot picking, enzymatic digestion, and matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS) for protein identification of DIGE-analyzed proteins are discussed. Two examples are given: first, a specific protein is used to test the sensitivity of the 2D DIGE/MALDI MS combination for protein quantification and identification, and second, several proteins with and without the labels typically used in DIGE are identified to demonstrate that these labels do not alter MS-based protein identification. Technical variations of protein gel spot preparation, in-gel digestion, and mass spectral protein identification are discussed.


Assuntos
Proteínas/análise , Proteínas/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Eletroforese em Gel Diferencial Bidimensional/métodos , Humanos , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/isolamento & purificação , Fragmentos de Peptídeos/metabolismo , Proteínas/isolamento & purificação , Proteínas/metabolismo , Proteômica , Tripsina/metabolismo
12.
J Lipid Res ; 48(4): 806-15, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17264352

RESUMO

Withdrawing growth factors or serum from endothelial cells leads to the activation of effector caspases 3 and 7, resulting in apoptotic cell death. HDL protects against caspase induction through sphingosine-1-phosphate (S1P) receptors. This anti-caspase activity of HDL is antagonized by VLDL from apolipoprotein E4 (apoE4) (genotype, APOE4/4; apolipoprotein, apoE) targeted replacement (TR) mice, but not by VLDL from TR APOE3/3 mice, and requires the binding of apoE4-VLDL to an LDL receptor family member. In the absence of HDL, apoE4-VLDL and apoE3-VLDL from TR mice have limited antiapoptotic activity. In contrast, we show here that a high-fat/high-cholesterol/cholate diet (HFD) radically alters this biological activity of VLDL. On HFD, both apoE3-VLDL and apoE4-VLDL (HFD VLDL) inhibit caspase 3/7 activation initiated by serum withdrawal. This activity of HFD VLDL is independent of an LDL receptor family member but requires the activation of S1P(3) receptors, as shown by the ability of pharmacological block of S1P receptors by VPC 23019 and by small interfering RNA-mediated downregulation of S1P(3) receptors to inhibit HFD VLDL anticaspase activity.


Assuntos
Apoptose/efeitos dos fármacos , Colesterol/farmacologia , Gorduras na Dieta/farmacologia , Lipoproteínas VLDL/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Apolipoproteína E3 , Apolipoproteína E4 , Inibidores de Caspase , Caspases/metabolismo , Sobrevivência Celular , Dieta , Lipoproteínas HDL/fisiologia , Camundongos , Camundongos Transgênicos
13.
Cell Biol Int ; 26(5): 407-20, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12095226

RESUMO

Apolipoprotein E (apoE) plays a role in the distribution of lipid within many organs and cell types in the human body, including the central nervous system (CNS). The apoE4 isoform is also an established risk factor for late-onset Alzheimer's disease (AD), however its role in the aetiology of the disease remains largely unknown. Therefore, as AD is a late-onset disease, we sort to investigate how conditions hypothesised to model ageing affect apoE metabolism, such as the transport of apoE along the secretory pathway. Two of these models include oxidative stress and calcium deregulation. Using apoE-EGFP-expressing astrocytoma cell lines we established that vesicle number and velocity are up-regulated under oxidative stress conditions, and slowed under KCl induced calcium deregulation. Although these findings apply to cells in general under these two stress conditions, the up-regulation of apoE in particular may be a response to cell injury with implications for neurodegeneration such as that found with late-onset AD.


Assuntos
Apolipoproteínas E/metabolismo , Cálcio/metabolismo , Estresse Oxidativo , Apolipoproteína E2 , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/genética , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cloreto de Cálcio/farmacologia , Vesículas Citoplasmáticas , Retículo Endoplasmático/metabolismo , Expressão Gênica/efeitos dos fármacos , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde , Humanos , Peróxido de Hidrogênio/farmacologia , Líquido Intracelular/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microtúbulos/metabolismo , Nocodazol/farmacologia , Oxidantes/farmacologia , Paclitaxel/farmacologia , Cloreto de Potássio/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Fatores de Tempo , Células Tumorais Cultivadas
14.
Cell Biol Int ; 26(9): 761-70, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12377208

RESUMO

Apolipoprotein E (apoE), a well characterized protein, forms lipoprotein complexes with cholesterol. Such complexes formed are endocytosed via the LDL receptor family by many cell types in particular within the human central nervous system (CNS). The apoE-endocytic pathway leads to apoE degradation. However, it has recently been indirectly shown that apoE can be retained intracellularly and then re-secreted. To investigate the fate of endocytosed apoE isoforms E2 and E3 within human CNS cells in real-time, we added the CNS form of these apoE isoforms, linked to a green fluorescent protein (EGFP), to cultured human foetal brain tissue. There was bi-directional trafficking of apoE-EGFP in neuron and astrocyte processes and 'stationary' perinuclear vesicles in type-I astrocytes. Thus, active apoE recycling in cells with defined processes suggests a role for apoE in mediating signalling through receptor-mediated endocytosis.


Assuntos
Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Encéfalo/citologia , Endocitose/fisiologia , Neurônios/metabolismo , Astrocitoma , Vesículas Citoplasmáticas/metabolismo , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Transporte Proteico/fisiologia , Células Tumorais Cultivadas
15.
J Lipid Res ; 44(8): 1566-73, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12754278

RESUMO

Endothelial cell apoptosis can be initiated by withdrawing growth factors or serum, and is inhibited by HDL. Our results show that the total lipoprotein population from apolipoprotein E 4/4 (APOE4/4) sera is less anti-apoptotic than total lipoproteins from other APOE genotypes, as measured by caspase 3/7 activity. Moreover, APOE4/4 VLDL antagonizes the antiapoptotic activity of HDL by a mechanism requiring binding of apoE4 on VLDL particles to an LDL family receptor. This ability of APOE4/4 VLDL to inhibit the antiapoptotic effects of HDL presents a potential mechanism by which the expression of several diseases, including atherosclerosis, is enhanced by the APOE4 genotype.


Assuntos
Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apoptose , Apolipoproteína E3 , Apolipoproteína E4 , Caspases/metabolismo , Linhagem Celular , HDL-Colesterol/metabolismo , Genótipo , Humanos , Lipoproteínas VLDL/metabolismo , Lisofosfolipídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA