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.
Hum Gene Ther ; 34(17-18): 927-946, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37597209

RESUMO

Lipoprotein lipase deficiency (LPLD) results from mutations within the lipoprotein lipase (LPL) gene that lead to a complete lack of catalytically active LPL protein. Glybera was one of the first adeno-associated virus (AAV) gene replacement therapy to receive European Medicines Agency regulatory approval for the treatment of LPLD. However, Glybera is no longer marketed potentially due to a combination of economical, manufacturing, and vector-related issues. The aim of this study was to develop a more efficacious AAV gene therapy vector for LPLD. Following preclinical biodistribution, efficacy and non-Good Laboratory Practice toxicity studies with novel AAV1 and AAV8-based vectors in mice, we identified AAV8 pVR59. AAV8 pVR59 delivered a codon-optimized, human gain-of-function hLPLS447X transgene driven by a CAG promoter in an AAV8 capsid. AAV8 pVR59 was significantly more efficacious, at 10- to 100-fold lower doses, compared with an AAV1 vector based on Glybera, when delivered intramuscularly or intravenously, respectively, in mice with LPLD. Efficient gene transfer was observed within the injected skeletal muscle and liver following delivery of AAV8 pVR59, with long-term correction of LPLD phenotypes, including normalization of plasma triglycerides and lipid tolerance, for up to 6 months post-treatment. While intramuscular delivery of AAV8 pVR59 was well tolerated, intravenous administration augmented liver pathology. These results highlight the feasibility of developing a superior AAV vector for the treatment of LPLD and provide critical insight for initiating studies in larger animal models. The identification of an AAV gene therapy vector that is more efficacious at lower doses, when paired with recent advances in production and manufacturing technologies, will ultimately translate to increased safety and accessibility for patients.


Assuntos
Hiperlipoproteinemia Tipo I , Humanos , Animais , Camundongos , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/terapia , Distribuição Tecidual , Transgenes , Administração Intravenosa
2.
Medicine (Baltimore) ; 99(20): e20180, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32443337

RESUMO

BACKGROUND: Previous clinical studies have reported that clinical value of high-frequency ultrasound combined computed tomography (HFUCT) is used for diagnosis of thyroid tumor (TT). However, no study has investigated this topic systematically. Therefore, this study will evaluate the clinical value of HFUCT for the diagnosis of TT. METHODS: We will search the databases of Cochrane Library, EMBASE, PUBMED, SCOPUS, Web of Science, OpenGrey, Cumulative Index to Nursing and Allied Health Literature, Allied and Complementary Medicine Database, and China National Knowledge Infrastructure from any time period published to the present. We will consider all case-controlled studies that assessed the clinical value of HFUCT for diagnosis of TT. Two authors will independently scan titles and abstracts to check eligible studies, followed by full-text read. We will extract data and assess study quality using Quality Assessment of Diagnostic Accuracy Studies tool. RevMan 5.3 software will be utilized for data pooling and statistical analysis. RESULTS: This study will be performed to assess the clinical value of HFUCT for the diagnosis of TT, and will provide an evidence-based synthesis for clinical application and further study. CONCLUSION: Summary of this study will provide the latest evidence to determine whether HFUCT can be used for TT diagnosis accurately. STUDY REGISTRATION: INPLASY202040022.


Assuntos
Imagem Multimodal/métodos , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Ultrassonografia/métodos , Estudos de Casos e Controles , China/epidemiologia , Humanos , Imagem Multimodal/estatística & dados numéricos , Garantia da Qualidade dos Cuidados de Saúde/métodos , Projetos de Pesquisa , Sensibilidade e Especificidade , Software , Neoplasias da Glândula Tireoide/patologia , Ultrassonografia/tendências , Metanálise como Assunto
3.
Arterioscler Thromb Vasc Biol ; 37(11): 2147-2155, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28882873

RESUMO

OBJECTIVE: High-density lipoproteins (HDL) are considered to protect against atherosclerosis in part by facilitating the removal of cholesterol from peripheral tissues. However, factors regulating lipid efflux are incompletely understood. We previously identified a variant in adenosine triphosphate-binding cassette transporter A8 (ABCA8) in an individual with low HDL cholesterol (HDLc). Here, we investigate the role of ABCA8 in cholesterol efflux and in regulating HDLc levels. APPROACH AND RESULTS: We sequenced ABCA8 in individuals with low and high HDLc and identified, exclusively in low HDLc probands, 3 predicted deleterious heterozygous ABCA8 mutations (p.Pro609Arg [P609R], IVS17-2 A>G and p.Thr741Stop [T741X]). HDLc levels were lower in heterozygous mutation carriers compared with first-degree family controls (0.86±0.34 versus 1.17±0.26 mmol/L; P=0.005). HDLc levels were significantly decreased by 29% (P=0.01) in Abca8b-/- mice on a high-cholesterol diet compared with wild-type mice, whereas hepatic overexpression of human ABCA8 in mice resulted in significant increases in plasma HDLc and the first steps of macrophage-to-feces reverse cholesterol transport. Overexpression of wild-type but not mutant ABCA8 resulted in a significant increase (1.8-fold; P=0.01) of cholesterol efflux to apolipoprotein AI in vitro. ABCA8 colocalizes and interacts with adenosine triphosphate-binding cassette transporter A1 and further potentiates adenosine triphosphate-binding cassette transporter A1-mediated cholesterol efflux. CONCLUSIONS: ABCA8 facilitates cholesterol efflux and modulates HDLc levels in humans and mice.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol na Dieta/sangue , HDL-Colesterol/sangue , Transportadores de Cassetes de Ligação de ATP/deficiência , Transportadores de Cassetes de Ligação de ATP/genética , Adulto , Idoso , Animais , Apolipoproteína A-I/sangue , Apolipoproteína B-100/sangue , Transporte Biológico , Biomarcadores/sangue , Células COS , Estudos de Casos e Controles , Chlorocebus aethiops , Análise Mutacional de DNA , Dieta Hiperlipídica , Fezes/química , Feminino , Células HEK293 , Hereditariedade , Heterozigoto , Humanos , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Transfecção
4.
Mol Genet Metab ; 119(1-2): 44-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27477828

RESUMO

Primary 5-oxoprolinuria (pyroglutamic aciduria) is caused by a genetic defect in the γ-glutamyl cycle, affecting either glutathione synthetase or 5-oxoprolinase. While several dozens of patients with glutathione synthetase deficiency have been reported, with hemolytic anemia representing the clinical key feature, 5-oxoprolinase deficiency due to OPLAH mutations is less frequent and so far has not attracted much attention. This has prompted us to investigate the clinical phenotype as well as the underlying genotype in patients from 14 families of various ethnic backgrounds who underwent diagnostic mutation analysis following the detection of 5-oxoprolinuria. In all patients with 5-oxoprolinuria studied, bi-allelic mutations in OPLAH were indicated. An autosomal recessive mode of inheritance for 5-oxoprolinase deficiency is further supported by the identification of a single mutation in all 9/14 parent sample sets investigated (except for the father of one patient whose result suggests homozygosity), and the absence of 5-oxoprolinuria in all tested heterozygotes. It is remarkable, that all 20 mutations identified were novel and private to the respective families. Clinical features were highly variable and in several sib pairs, did not segregate with 5-oxoprolinuria. Although a pathogenic role of 5-oxoprolinase deficiency remains possible, this is not supported by our findings. Additional patient ascertainment and long-term follow-up is needed to establish the benign nature of this inborn error of metabolism. It is important that all symptomatic patients with persistently elevated levels of 5-oxoproline and no obvious explanation are investigated for the genetic etiology.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Glutationa Sintase/deficiência , Piroglutamato Hidrolase/deficiência , Piroglutamato Hidrolase/genética , Ácido Pirrolidonocarboxílico/metabolismo , Adolescente , Alelos , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/fisiopatologia , Criança , Pré-Escolar , Feminino , Glutationa/metabolismo , Glutationa Sintase/genética , Heterozigoto , Homozigoto , Humanos , Lactente , Masculino , Mutação
5.
Brain ; 139(Pt 3): 674-91, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26700687

RESUMO

Congenital myopathies are a clinically and genetically heterogeneous group of muscle disorders characterized by congenital or early-onset hypotonia and muscle weakness, and specific pathological features on muscle biopsy. The phenotype ranges from foetal akinesia resulting in in utero or neonatal mortality, to milder disorders that are not life-limiting. Over the past decade, more than 20 new congenital myopathy genes have been identified. Most encode proteins involved in muscle contraction; however, mutations in ion channel-encoding genes are increasingly being recognized as a cause of this group of disorders. SCN4A encodes the α-subunit of the skeletal muscle voltage-gated sodium channel (Nav1.4). This channel is essential for the generation and propagation of the muscle action potential crucial to muscle contraction. Dominant SCN4A gain-of-function mutations are a well-established cause of myotonia and periodic paralysis. Using whole exome sequencing, we identified homozygous or compound heterozygous SCN4A mutations in a cohort of 11 individuals from six unrelated kindreds with congenital myopathy. Affected members developed in utero- or neonatal-onset muscle weakness of variable severity. In seven cases, severe muscle weakness resulted in death during the third trimester or shortly after birth. The remaining four cases had marked congenital or neonatal-onset hypotonia and weakness associated with mild-to-moderate facial and neck weakness, significant neonatal-onset respiratory and swallowing difficulties and childhood-onset spinal deformities. All four surviving cohort members experienced clinical improvement in the first decade of life. Muscle biopsies showed myopathic features including fibre size variability, presence of fibrofatty tissue of varying severity, without specific structural abnormalities. Electrophysiology suggested a myopathic process, without myotonia. In vitro functional assessment in HEK293 cells of the impact of the identified SCN4A mutations showed loss-of-function of the mutant Nav1.4 channels. All, apart from one, of the mutations either caused fully non-functional channels, or resulted in a reduced channel activity. Each of the affected cases carried at least one full loss-of-function mutation. In five out of six families, a second loss-of-function mutation was present on the trans allele. These functional results provide convincing evidence for the pathogenicity of the identified mutations and suggest that different degrees of loss-of-function in mutant Nav1.4 channels are associated with attenuation of the skeletal muscle action potential amplitude to a level insufficient to support normal muscle function. The results demonstrate that recessive loss-of-function SCN4A mutations should be considered in patients with a congenital myopathy.


Assuntos
Hipocinesia/diagnóstico , Hipocinesia/genética , Mutação/genética , Miopatias Congênitas Estruturais/diagnóstico , Miopatias Congênitas Estruturais/genética , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Células HEK293 , Humanos , Recém-Nascido , Masculino , Linhagem , Índice de Gravidade de Doença , Xenopus laevis
6.
Arterioscler Thromb Vasc Biol ; 33(12): 2724-32, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24135019

RESUMO

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) protein maintains cellular cholesterol homeostasis in several different tissues. In the liver, ABCA1 is crucial for high-density lipoprotein biogenesis, and in the pancreas ABCA1 can regulate insulin secretion. In this study, our aim was to identify novel microRNAs that regulate ABCA1 expression in these tissues. APPROACH AND RESULTS: We combined multiple microRNA prediction programs to identify 8 microRNAs that potentially regulate ABCA1. A luciferase reporter assay demonstrated that 5 of these microRNAs (miR-148, miR-27, miR-144, miR-145, and miR-33a/33b) significantly repressed ABCA1 3'-untranslated region activity with miR-145 resulting in one of the larger decreases. In hepatic HepG2 cells, miR-145 can regulate both ABCA1 protein expression levels and cholesterol efflux function. In murine islets, an increase in miR-145 expression decreased ABCA1 protein expression, increased total islet cholesterol levels, and decreased glucose-stimulated insulin secretion. Inhibiting miR-145 produced the opposite effect of increasing ABCA1 protein levels and improving glucose-stimulated insulin secretion. Finally, increased glucose levels in media significantly decreased miR-145 levels in cultured pancreatic beta cells. These findings suggest that miR-145 is involved in glucose homeostasis and is regulated by glucose concentration. CONCLUSIONS: Our studies demonstrate that miR-145 regulates ABCA1 expression and function, and inhibiting this microRNA represents a novel strategy for increasing ABCA1 expression, promoting high-density lipoprotein biogenesis in the liver, and improving glucose-stimulated insulin secretion in islets.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Hepatócitos/metabolismo , Ilhotas Pancreáticas/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Transportador 1 de Cassete de Ligação de ATP/genética , Animais , Sítios de Ligação , Colesterol/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Glucose/metabolismo , Células Hep G2 , Homeostase , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Lipoproteínas HDL/metabolismo , Camundongos , Transfecção
7.
J Lipid Res ; 53(5): 941-950, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22389325

RESUMO

The lipidation of apoA-I in liver greatly influences HDL biogenesis and plasma HDL levels by stabilizing the secreted apoA-I. Niacin is the most effective lipid-regulating agent clinically available to raise HDL. This study was undertaken to identify regulatory mechanisms of niacin action in hepatic lipidation of apoA-I, a critical event involved in HDL biogenesis. In cultured human hepatocytes (HepG2), niacin increased: association of apoA-I with phospholipids and cholesterol by 46% and 23% respectively, formation of lipid-poor single apoA-I molecule-containing particles up to ~2.4-fold, and pre ß 1 and α migrating HDL particles. Niacin dose-dependently stimulated the cell efflux of phospholipid and cholesterol and increased transcription of ABCA1 gene and ABCA1 protein. Mutated DR4, a binding site for nuclear factor liver X receptor alpha (LXR α ) in the ABCA1 promoter, abolished niacin stimulatory effect. Further, knocking down LXR α or ABCA1 by RNA interference eliminated niacin-stimulated apoA-I lipidation. Niacin treatment did not change apoA-I gene expression. The present data indicate that niacin increases apoA-I lipidation by enhancing lipid efflux through a DR4-dependent transcription of ABCA1 gene in HepG2 cells. A stimulatory role of niacin in early hepatic formation of HDL particles suggests a new mechanism that contributes to niacin action to increase the stability of newly synthesized circulating HDL.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Apolipoproteína A-I/metabolismo , HDL-Colesterol/biossíntese , Niacina/farmacologia , Sequências Repetitivas de Ácido Nucleico/genética , Transcrição Gênica/efeitos dos fármacos , Transportador 1 de Cassete de Ligação de ATP , Transporte Biológico/efeitos dos fármacos , Meios de Cultura/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Lipoproteínas de Alta Densidade Pré-beta/metabolismo , Humanos , Movimento/efeitos dos fármacos , Fosfolipídeos/metabolismo , Sequências Repetitivas de Ácido Nucleico/efeitos dos fármacos
8.
Diabetes ; 61(3): 653-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22315319

RESUMO

Changes in cellular cholesterol affect insulin secretion, and ß-cell-specific deletion or loss-of-function mutations in the cholesterol efflux transporter ATP-binding cassette transporter A1 (ABCA1) result in impaired glucose tolerance and ß-cell dysfunction. Upregulation of ABCA1 expression may therefore be beneficial for the maintenance of normal islet function in diabetes. Studies suggest that microRNA-33a (miR-33a) expression inversely correlates with ABCA1 expression in hepatocytes and macrophages. We examined whether miR-33a regulates ABCA1 expression in pancreatic islets, thereby affecting cholesterol accumulation and insulin secretion. Adenoviral miR-33a overexpression in human or mouse islets reduced ABCA1 expression, decreased glucose-stimulated insulin secretion, and increased cholesterol levels. The miR-33a-induced reduction in insulin secretion was rescued by cholesterol depletion by methyl-ß-cyclodextrin or mevastatin. Inhibition of miR-33a expression in apolipoprotein E knockout islets and ABCA1 overexpression in ß-cell-specific ABCA1 knockout islets rescued normal insulin secretion and reduced islet cholesterol. These findings confirm the critical role of ß-cell ABCA1 in islet cholesterol homeostasis and ß-cell function and highlight modulation of ß-cell miR-33a expression as a means to influence insulin secretion.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Colesterol/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , MicroRNAs/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/análise , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Glucose/farmacologia , Humanos , Secreção de Insulina , Camundongos , beta-Ciclodextrinas/farmacologia
9.
J Lipid Res ; 51(8): 2211-22, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20371549

RESUMO

Pioglitazone, a hypoglycemic agent, has been shown to increase plasma HDL cholesterol, but the mechanism is incompletely understood. We further investigated effects of pioglitazone on transcriptional regulation of apolipoprotein (apo)A-I gene and functional properties of pioglitazone-induced apoA-I-containing particles. Pioglitazone dose-dependently stimulated apoA-I promoter activities in HepG2 cells. A peroxisome proliferator-activated receptor (PPAR)-response element located in site A (-214 to -192 bp, upstream of the transcription start site) of the promoter is required for pioglitazone-induced apoA-I gene transcription. Deletion of site A (-214 to -192 bp), B (-169 to -146 bp), or C (-134 to -119 bp), which clusters a number of cis-acting elements for binding of different transcription factors, reduced the basal apoA-I promoter activities, and no additional pioglitazone-sensitive elements were found within this region. Overexpression or knock-down of liver receptor homolog-1, a newly identified nuclear factor with strong stimulatory effect on apoA-I transcription, did not alter pioglitazone-induced apoA-I transcription. Pioglitazone-induced apoA-I transcription is mainly mediated through PPARalpha but not PPARgamma in hepatocytes. Pioglitazone induced production of HDL enriched in its subfraction containing apoA-I without apoA-II, which inhibited monocyte adhesion to endothelial cells in vitro. In conclusion, pioglitazone increases apoA-I production by directly enhancing PPAR-response element-dependent transcription, resulting in generation of apoA-I-containing HDL particles with increased anti-inflammatory property.


Assuntos
Apolipoproteína A-I/biossíntese , Apolipoproteína A-I/genética , Hipoglicemiantes/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/genética , Elementos de Resposta , Tiazolidinedionas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Aorta/citologia , Sequência de Bases , Adesão Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Células Hep G2 , Humanos , Lipoproteínas HDL/química , Monócitos/citologia , Monócitos/efeitos dos fármacos , PPAR alfa/genética , PPAR alfa/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Pioglitazona
10.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 28(5): 682-5, 2006 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-17121231

RESUMO

OBJECTIVE: To explore the feasibility of utilizing two implantable devices made from modified polyurethane films with antibody tethered replication-defective adenoviruses encoding for green fluorescent protein (AdGFP) as gene delivery platforms. METHODS: Intra-aortic button implants of collagen-coated polyurethane films with antibody tethered AdGFP were sutured into the infrarenal aorta of adult pigs and pulmonary valve leaflet in juvenile sheep was replaced by polyurethane pulmonary valve cusp replacement with antibody-tethered AdGFP. After seven days, the buttons, prosthetic leaflets, and their surrounding tissues were explanted and evaluated for biocompatibility and AdGFP-mediated gene transfer by fluorescent microscopy and PCR analysis. RESULTS: In vivo analysis of gene transfer from collagen-coated polyurethane films in pig infrarenal aorta implants, one week explants of the collagen-coated polyurethane films demonstrated (14.2 +/- 2.5)% of neointimal cells on the surface of the implant. In sheep pulmonary valve leaflet replacement studies, polyurethane films with antibody tethered AdGFP vector demonstrated (25.1 +/- 5.7)% of cells attached to polyurethane valve leaflets were transduced in one week. PCR analyses showed that GFP DNA was not detectable in blood or distal tissues. CONCLUSION: Site-specific intravascular delivery of adenoviral vectors for gene therapy can be achieved with these two kinds of polyurethane implants utilizing the antivector antibody tethering mechanism.


Assuntos
Adenoviridae/genética , Prótese Vascular , Técnicas de Transferência de Genes , Terapia Genética/métodos , Próteses Valvulares Cardíacas , Poliuretanos , Animais , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Masculino , Poliuretanos/química , Implantação de Prótese , Ovinos , Suínos
11.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 28(3): 350-4, 2006 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-16900631

RESUMO

OBJECTIVE: To study the feasibility of delivering viral gene vector from a collagen-coated polyurethane (PU) film through a mechanism involving monoclonal antiviral antibody tethering. METHODS: Anti-adenoviral monoclonal antibodies were covalently bound to the collagen-coated PU surface. These antibodies enabled tethering of replication defective adenoviruses through highly specific antigen-antibody affinity. The PU film-based gene delivery using antibody-tethered adenovirus encoding green fluorescent protein (GEP) was tested in rat arterial smooth muscle cell (A10 cell) culture in vitro. The virus binding stability was studied by incubating the collagen-coated PU film in PBS solution at 37 degrees C for 20 days, followed with A10 cell cultures with the incubated films and the corresponding buffer solution. RESULTS: PU films with antibody-tethered adenovirus encoding GFP demonstrated efficient and highly localized gene delivery to A10 cells. Virus binding was stable for at least 10 days at physiological conditions, more than 77% of the originally bound virus remained in the film after 15 day's incubation. CONCLUSION: Gene delivery using PU film-based anti-viral antibody tethering of vectors exhibited potentials of applications in a wide array of single or multiple therapeutic gene strategies, and in further stent-based gene delivery therapeutic strategies.


Assuntos
Anticorpos Monoclonais/imunologia , Terapia Genética , Vetores Genéticos , Poliuretanos/química , Adenoviridae/genética , Especificidade de Anticorpos/imunologia , Técnicas de Transferência de Genes , Humanos , Ligação Proteica
12.
J Lipid Res ; 43(11): 1939-49, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12401893

RESUMO

Mutations in ABCA1 uniformly decrease plasma HDL-cholesterol (HDL-C) and reduce cholesterol efflux, yet different mutations in ABCA1 result in different phenotypic effects in heterozygotes. For example, truncation mutations result in significantly lower HDL-C and apoliprotein A-I (apoA-I) levels in heterozygotes compared with nontruncation mutations, suggesting that truncation mutations may negatively affect the wild-type allele. To specifically test this hypothesis, we examined ABCA1 protein expression in response to 9-cis-retinoic acid (9-cis-RA) and 22-R-hydroxycholesterol (22-R-OH-Chol) in a collection of human fibroblasts representing eight different mutations and observed that truncation mutations blunted the response to oxysterol stimulation and dominantly suppressed induction of the remaining full-length allele to 5-10% of wild-type levels. mRNA levels between truncation and nontruncation mutations were comparable, suggesting that ABCA1 expression was suppressed at the protein level. Dominant negative activity of truncated ABCA1 was recapitulated in an in vitro model using transfected Cos-7 cells. Our results suggest that the severe reduction of HDL-C in patients with truncation mutations may be at least partly explained by dominant negative suppression of expression and activity of the remaining full-length ABCA1 allele. These data suggest that ABCA1 requires a physical association with itself or other molecules for normal function and has important pharmacogenetic implications for individuals with truncation mutations.


Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/genética , Hidroxicolesteróis/farmacologia , Mutação/genética , Tretinoína/farmacologia , Regulação para Cima/efeitos dos fármacos , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/química , Alitretinoína , Alelos , Animais , Apolipoproteína A-I/metabolismo , Fibroblastos , Genes Dominantes , Heterozigoto , Humanos , Lipoproteínas HDL/análise , Macrófagos , Camundongos
13.
J Biol Chem ; 277(44): 41835-42, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12196520

RESUMO

ATP-binding cassette A1 (ABCA1) is a key mediator of cholesterol and phospholipid efflux to apolipoprotein particles. We show that ABCA1 is a constitutively phosphorylated protein in both RAW macrophages and in a human embryonic kidney cell line expressing ABCA1. Furthermore, we demonstrate that phosphorylation of ABCA1 is mediated by protein kinase A (PKA) or a PKA-like kinase in vivo. Through site-directed mutagenesis studies of consensus PKA phosphorylation sites and in vitro PKA kinase assays, we show that Ser-1042 and Ser-2054, located in the nucleotide binding domains of ABCA1, are major phosphorylation sites for PKA. ApoA-I-dependent phospholipid efflux was decreased significantly by mutation of Ser-2054 alone and Ser-1042/Ser-2054 but was not significantly impaired with Ser-1042 alone. The mechanism by which ABCA1 phosphorylation affected ApoA-I-dependent phospholipid efflux did not involve either alterations in ApoA-I binding or changes in ABCA1 protein stability. These studies demonstrate a novel serine (Ser-2054) on the ABCA1 protein crucial for PKA phosphorylation and for regulation of ABCA1 transporter activity.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fosfolipídeos/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/química , Sequência de Aminoácidos , Animais , Apolipoproteína A-I/metabolismo , Células Cultivadas , Humanos , Camundongos , Dados de Sequência Molecular , Fosforilação , Serina , Relação Estrutura-Atividade
14.
Nat Genet ; 32(2): 326-30, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12172548

RESUMO

Familial exudative vitreoretinopathy (FEVR) is a hereditary ocular disorder characterized by a failure of peripheral retinal vascularization. Loci associated with FEVR map to 11q13-q23 (EVR1; OMIM 133780, ref. 1), Xp11.4 (EVR2; OMIM 305390, ref. 2) and 11p13-12 (EVR3; OMIM 605750, ref. 3). Here we have confirmed linkage to the 11q13-23 locus for autosomal dominant FEVR in one large multigenerational family and refined the disease locus to a genomic region spanning 1.55 Mb. Mutations in FZD4, encoding the putative Wnt receptor frizzled-4, segregated completely with affected individuals in the family and were detected in affected individuals from an additional unrelated family, but not in normal controls. FZD genes encode Wnt receptors, which are implicated in development and carcinogenesis. Injection of wildtype and mutated FZD4 into Xenopus laevis embryos revealed that wildtype, but not mutant, frizzled-4 activated calcium/calmodulin-dependent protein kinase II (CAMKII) and protein kinase C (PKC), components of the Wnt/Ca(2+) signaling pathway. In one of the mutants, altered subcellular trafficking led to defective signaling. These findings support a function for frizzled-4 in retinal angiogenesis and establish the first association between a Wnt receptor and human disease.


Assuntos
Neovascularização Patológica/genética , Proteínas/genética , Doenças Retinianas/genética , Vasos Retinianos/patologia , Sequência de Aminoácidos , Pré-Escolar , Feminino , Receptores Frizzled , Marcadores Genéticos , Haplótipos , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Polimorfismo Genético , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Retina/patologia , Doenças Retinianas/patologia , Alinhamento de Sequência , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA