Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(1)2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36680262

RESUMO

Alaska is a unique US state because of its large size, geographically disparate population density, and physical distance from the contiguous United States. Here, we describe a pattern of SARS-CoV-2 variant emergence across Alaska reflective of these differences. Using genomic data, we found that in Alaska, the Omicron sublineage BA.2.3 overtook BA.1.1 by the week of 27 February 2022, reaching 48.5% of sequenced cases. On the contrary, in the contiguous United States, BA.1.1 dominated cases for longer, eventually being displaced by BA.2 sublineages other than BA.2.3. BA.2.3 only reached a prevalence of 10.9% in the contiguous United States. Using phylogenetics, we found evidence of potential origins of the two major clades of BA.2.3 in Alaska and with logistic regression estimated how it emerged and spread throughout the state. The combined evidence is suggestive of founder events in Alaska and is reflective of how Alaska's unique dynamics influence the emergence of SARS-CoV-2 variants.


Assuntos
COVID-19 , Dermatite , Humanos , Alaska/epidemiologia , SARS-CoV-2/genética , COVID-19/epidemiologia
2.
Sci Rep ; 12(1): 20662, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36450812

RESUMO

Alaska has the lowest population density in the United States (US) with a mix of urban centers and isolated rural communities. Alaska's distinct population dynamics compared to the contiguous US may have contributed to unique patterns of SARS-CoV-2 variants observed in early 2021. Here we examined 2323 SARS-CoV-2 genomes from Alaska and 278,635 from the contiguous US collected from December 2020 through June 2021 because of the notable emergence and spread of lineage B.1.1.519 in Alaska. We found that B.1.1.519 was consistently detected from late January through June of 2021 in Alaska with a peak prevalence in April of 77.9% unlike the rest of the US at 4.6%. The earlier emergence of B.1.1.519 coincided with a later peak of Alpha (B.1.1.7) compared to the contiguous US. We also observed differences in variant composition over time between the two most populated regions of Alaska and a modest increase in COVID-19 cases during the peak incidence of B.1.1.519. However, it is difficult to disentangle how social dynamics conflated changes in COVID-19 during this time. We suggest that the viral characteristics, such as amino acid substitutions in the spike protein, likely contributed to the unique spread of B.1.1.519 in Alaska.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Alaska/epidemiologia , COVID-19/epidemiologia , Substituição de Aminoácidos
3.
medRxiv ; 2022 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-35664999

RESUMO

Alaska is a unique US state because of its large size, geographically disparate population density, and physical distance from the contiguous United States. Here, we describe a pattern of SARS-CoV-2 variant emergence across Alaska reflective of these differences. Using genomic data, we found that in Alaska the Omicron sublineage BA.2.3 overtook BA.1.1 by the week of 2022-02-27, reaching 48.5% of sequenced cases. On the contrary in the contiguous United States, BA.1.1 dominated cases for longer, eventually being displaced by BA.2 sublineages other than BA.2.3. BA.2.3 only reached a prevalence of 10.9% in the contiguous United States. Using phylogenetics, we found evidence of potential origins of the two major clades of BA.2.3 in Alaska and with logistic regression estimated how it emerged and spread throughout the state. The combined evidence is suggestive of founder events in Alaska and is reflective of how Alaska’s unique dynamics influence the emergence of SARS-CoV-2 variants.

5.
Biomedicines ; 9(3)2021 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-33804693

RESUMO

Essential metals such as copper (Cu) and zinc (Zn) are important cofactors in diverse cellular processes, while metal imbalance may impact or be altered by disease state. Cu is essential for aerobic life with significant functions in oxidation-reduction catalysis. This redox reactivity requires precise intracellular handling and molecular-to-organismal levels of homeostatic control. As the central organ of Cu homeostasis in vertebrates, the liver has long been associated with Cu storage disorders including Wilson Disease (WD) (heritable human Cu toxicosis), Idiopathic Copper Toxicosis and Endemic Tyrolean Infantile Cirrhosis. Cu imbalance is also associated with chronic liver diseases that arise from hepatitis viral infection or other liver injury. The labile redox characteristic of Cu is often discussed as a primary mechanism of Cu toxicity. However, work emerging largely from the study of WD models suggests that Cu toxicity may have specific biochemical consequences that are not directly attributable to redox activity. This work reviews Cu toxicity with a focus on the liver and proposes that Cu accumulation specifically impacts Zn-dependent processes. The prospect that Cu toxicity has specific biochemical impacts that are not entirely attributable to redox may promote further inquiry into Cu toxicity in WD and other Cu-associated disorders.

6.
J Nutr Biochem ; 78: 108316, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31986483

RESUMO

This study aimed to characterize the impact of dietary copper on the biochemical and hepatic metabolite changes associated with fructose toxicity in a Wistar rat model of fructose-induced liver disease. Twenty-four male and 24 female, 6-week-old, Wister rats were separated into four experimental dietary treatment groups (6 males and 6 females per group), as follows: (1) a control diet: containing no fructose with adequate copper (i.e., CuA/0% Fruct); (2) a diet regimen identical to the control and supplemented with 30% w/v fructose in the animals' drinking water (CuA/30% Fruct); (3) a diet identical to the control diet but deficient in copper content (CuD/0% Fruct) and (4) a diet identical to the control diet but deficient in copper content and supplemented with 30% w/v fructose in the drinking water (CuD/30% Fruct). The animals were fed the four diet regimens for 5 weeks, followed by euthanization and assessment of histology, elemental profiles and identification and quantitation of liver metabolites. Results from 1H nuclear magnetic resonance metabolomics revealed mechanistic insights into copper modulation of fructose hepatotoxicity through identification of distinct metabolic phenotypes that were highly correlated with diet and sex. This study also identified previously unknown sex-specific responses to both fructose supplementation and restricted copper intake, while the presence of adequate dietary copper promoted most pronounced fructose-induced metabolite changes.


Assuntos
Cobre/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Frutose/metabolismo , Fígado/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , ATPases Transportadoras de Cobre/metabolismo , Dieta , Feminino , Frutose/efeitos adversos , Fígado/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Metabolômica , Análise Multivariada , Hepatopatia Gordurosa não Alcoólica/patologia , Fenótipo , Análise de Componente Principal , Ratos , Ratos Wistar , Fatores Sexuais
7.
J Cell Sci ; 132(19)2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31515276

RESUMO

Copper-responsive intracellular ATP7B trafficking is crucial for maintaining the copper balance in mammalian hepatocytes and thus copper levels in organs. The copper metabolism domain-containing protein 1 (COMMD1) binds both the ATP7B copper transporter and phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P2], whereas COMMD1 loss causes hepatocyte copper accumulation. Although it is clear that COMMD1 is localized to endocytic trafficking complexes, a direct function for COMMD1 in ATP7B trafficking has not yet been defined. In this study, experiments using quantitative colocalization analysis reveal that COMMD1 modulates copper-responsive ATP7B trafficking through recruitment to PtdIns(4,5)P2 Decreased COMMD1 abundance results in loss of ATP7B from lysosomes and the trans-Golgi network (TGN) in high copper conditions, although excess expression of COMMD1 also disrupts ATP7B trafficking and TGN structure. Overexpression of COMMD1 mutated to inhibit PtdIns(4,5)P2 binding has little impact on ATP7B trafficking. A mechanistic PtdIns(4,5)P2-mediated function for COMMD1 is proposed that is consistent with decreased cellular copper export as a result of disruption of the ATP7B trafficking itinerary and early endosome accumulation when COMMD1 is depleted. PtdIns(4,5)P2 interaction with COMMD1 as well as COMMD1 abundance could both be important in maintenance of specific membrane protein trafficking pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , ATPases Transportadoras de Cobre/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Cobre/metabolismo , Cobre/farmacologia , ATPases Transportadoras de Cobre/genética , Células Hep G2 , Humanos , Fígado/metabolismo , Lisossomos/metabolismo , Ligação Proteica , Transporte Proteico/genética , Transporte Proteico/fisiologia , Rede trans-Golgi/metabolismo
8.
J Trace Elem Med Biol ; 53: 113-119, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30910193

RESUMO

The independent toxic effects of copper and acetaminophen are among the most studied topics in liver toxicity. Here, in an animal model of Cebus capucinus chronically exposed to high dietary copper, we assessed clinical and global transcriptional adaptations of the liver induced by a single high dose of acetaminophen. The experiment conditions were chosen to resemble a close to human real-life situation of exposure to both toxic stimuli. The clinical parameters and histological analyses indicated that chronic copper administration does not induce liver damage and may have a protective effect in acetaminophen challenge. Acetaminophen administration in previously non-exposed animals induced down-regulation of a complex network of gene regulators, highlighting the putative participation of the families of gene regulators HNF, FOX, PPAR and NRF controlling this process. This gene response was not observed in animals that previously received chronic oral copper, suggesting that this metal induces a transcriptional adaptation that may protect against acetaminophen toxicity, a classical adaptation response termed preconditioning of the liver.


Assuntos
Acetaminofen/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Cobre/administração & dosagem , Cobre/farmacologia , Substâncias Protetoras/farmacologia , Animais , Cebus , Modelos Animais de Doenças , Substâncias Protetoras/administração & dosagem
9.
Metallomics ; 10(11): 1595-1606, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30277246

RESUMO

Wilson disease (WD) is an autosomal recessive disorder caused by mutation in the ATP7B gene that affects copper transport in the body. ATP7B mutation damages copper transporter function, ultimately resulting in excessive copper accumulation and subsequent toxicity in both the liver and brain. Mechanisms of copper toxicity, however, are not well defined. The Atp7b-/- mouse model is well-characterized and presents a hepatic phenotype consistent with WD. In this study, we found that the untreated Atp7b-/- mice accumulate approximately 2-fold excess hepatic zinc compared to the wild type. We used targeted transcriptomics and proteomics to analyze the molecular events associated with zinc and copper accumulation in the Atp7b-/- mouse liver. Altered gene expression of Zip5 and ZnT1 zinc transporters indicated a transcriptional homeostatic response, while increased copper/zinc ratios associated with high levels of metallothioneins 1 and 2, indicated altered Zn availability in cells. These data suggest that copper toxicity in Wilson disease includes effects on zinc-dependent proteins. Transcriptional network analysis of RNA-seq data reveals an interconnected network of transcriptional activators with over-representation of zinc-dependent and zinc-responsive transcription factors. In the context of previous research, these observations support the hypothesis that mechanisms of copper toxicity include disruption of intracellular zinc distribution in liver cells. The translational significance of this work lies in oral zinc supplementation in treatment for WD, which is thought to mediate protective effects through the induction of metallothionein synthesis in the intestine. This work indicates broader impacts of altered zinc-copper balance in WD, including global transcriptional responses and altered zinc balance in the liver.


Assuntos
ATPases Transportadoras de Cobre/fisiologia , Cobre/toxicidade , Modelos Animais de Doenças , Degeneração Hepatolenticular , Fígado/patologia , Zinco/metabolismo , Animais , Redes Reguladoras de Genes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Metalotioneína/metabolismo , Camundongos , Camundongos Knockout
10.
PLoS Biol ; 16(9): e2006519, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30199530

RESUMO

Copper (Cu) has emerged as an important modifier of body lipid metabolism. However, how Cu contributes to the physiology of fat cells remains largely unknown. We found that adipocytes require Cu to establish a balance between main metabolic fuels. Differentiating adipocytes increase their Cu uptake along with the ATP7A-dependent transport of Cu into the secretory pathway to activate a highly up-regulated amino-oxidase copper-containing 3 (AOC3)/semicarbazide-sensitive amine oxidase (SSAO); in vivo, the activity of SSAO depends on the organism's Cu status. Activated SSAO oppositely regulates uptake of glucose and long-chain fatty acids and remodels the cellular proteome to coordinate changes in fuel availability and related downstream processes, such as glycolysis, de novo lipogenesis, and sphingomyelin/ceramide synthesis. The loss of SSAO-dependent regulation due to Cu deficiency, limited Cu transport to the secretory pathway, or SSAO inactivation shifts metabolism towards lipid-dependent pathways and results in adipocyte hypertrophy and fat accumulation. The results establish a role for Cu homeostasis in adipocyte metabolism and identify SSAO as a regulator of energy utilization processes in adipocytes.


Assuntos
Adipócitos/enzimologia , Adipócitos/metabolismo , Amina Oxidase (contendo Cobre)/metabolismo , Cobre/metabolismo , Células 3T3-L1 , Animais , Sequência de Bases , Transporte Biológico , Diferenciação Celular , Forma Celular , Tamanho Celular , Cobre/deficiência , ATPases Transportadoras de Cobre/metabolismo , Metabolismo Energético , Ativação Enzimática , Ácidos Graxos/biossíntese , Glucose/metabolismo , Homeostase , Hipertrofia , Masculino , Camundongos , Proteômica , Ratos Wistar , Via Secretória , Triglicerídeos/metabolismo
11.
IUBMB Life ; 69(4): 263-270, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28271632

RESUMO

The essential transition metal copper is important in lipid metabolism, redox balance, iron mobilization, and many other critical processes in eukaryotic organisms. Genetic diseases where copper homeostasis is disrupted, including Menkes disease and Wilson disease, indicate the importance of copper balance to human health. The severe consequences of insufficient copper supply are illustrated by Menkes disease, caused by mutation in the X-linked ATP7A gene encoding a protein that transports copper from intestinal epithelia into the bloodstream and across the blood-brain barrier. Inadequate copper supply to the body due to poor diet quality or malabsorption can disrupt several molecular level pathways and processes. Though much of the copper distribution machinery has been described and consequences of disrupted copper handling have been characterized in human disease as well as animal models, physiological consequences of sub-optimal copper due to poor nutrition or malabsorption have not been extensively studied. Recent work indicates that insufficient copper may be important in a number of common diseases including obesity, ischemic heart disease, and metabolic syndrome. Specifically, marginal copper deficiency (CuD) has been reported as a potential etiologic factor in diseases characterized by disrupted lipid metabolism such as non-alcoholic fatty-liver disease (NAFLD). In this review, we discuss the available data suggesting that a significant portion of the North American population may consume insufficient copper, the potential mechanisms by which CuD may promote lipid biosynthesis, and the interaction between CuD and dietary fructose in the etiology of NAFLD. © 2016 IUBMB Life, 69(4):263-270, 2017.


Assuntos
Cobre/metabolismo , Metabolismo dos Lipídeos/genética , Lipídeos/biossíntese , Hepatopatia Gordurosa não Alcoólica/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Barreira Hematoencefálica/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , ATPases Transportadoras de Cobre , Dieta , Humanos , Ferro/metabolismo , Lipídeos/genética , Fígado/metabolismo , Fígado/patologia , Síndrome dos Cabelos Torcidos/genética , Síndrome dos Cabelos Torcidos/metabolismo , Hepatopatia Gordurosa não Alcoólica/dietoterapia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia
12.
Virol J ; 14(1): 11, 2017 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-28115001

RESUMO

BACKGROUND: The essential role of copper in eukaryotic cellular physiology is known, but has not been recognized as important in the context of influenza A virus infection. In this study, we investigated the effect of cellular copper on influenza A virus replication. METHODS: Influenza A/WSN/33 (H1N1) virus growth and macromolecule syntheses were assessed in cultured human lung cells (A549) where the copper concentration of the growth medium was modified, or expression of host genes involved in copper homeostasis was targeted by RNA interference. RESULTS: Exogenously increasing copper concentration, or chelating copper, resulted in moderate defects in viral growth. Nucleoprotein (NP) localization, neuraminidase activity assays and transmission electron microscopy did not reveal significant defects in virion assembly, morphology or release under these conditions. However, RNAi knockdown of the high-affinity copper importer CTR1 resulted in significant viral growth defects (7.3-fold reduced titer at 24 hours post-infection, p = 0.04). Knockdown of CTR1 or the trans-Golgi copper transporter ATP7A significantly reduced polymerase activity in a minigenome assay. Both copper transporters were required for authentic viral RNA synthesis and NP and matrix (M1) protein accumulation in the infected cell. CONCLUSIONS: These results demonstrate that intracellular copper regulates the influenza virus life cycle, with potentially distinct mechanisms in specific cellular compartments. These observations provide a new avenue for drug development and studies of influenza virus pathogenesis.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/fisiologia , Replicação Viral , Linhagem Celular , Transportador de Cobre 1 , ATPases Transportadoras de Cobre , Meios de Cultura/química , Células Epiteliais/metabolismo , Humanos
13.
J Nutr Biochem ; 26(10): 996-1006, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26033743

RESUMO

Nonalcoholic fatty liver disease (NAFLD) prevalence is increasing worldwide, with the affected US population estimated near 30%. Diet is a recognized risk factor in the NAFLD spectrum, which includes nonalcoholic steatohepatitis (NASH) and fibrosis. Low hepatic copper (Cu) was recently linked to clinical NAFLD/NASH severity. Simple sugar consumption including sucrose and fructose is implicated in NAFLD, while consumption of these macronutrients also decreases liver Cu levels. Though dietary sugar and low Cu are implicated in NAFLD, transcript-level responses that connect diet and pathology are not established. We have developed a mature rat model of NAFLD induced by dietary Cu deficiency, human-relevant high sucrose intake (30% w/w) or both factors in combination. Compared to the control diet with adequate Cu and 10% (w/w) sucrose, rats fed either high-sucrose or low-Cu diet had increased hepatic expression of genes involved in inflammation and fibrogenesis, including hepatic stellate cell activation, while the combination of diet factors also increased ATP citrate lyase and fatty acid synthase gene transcription (fold change > 2, P < 0.02). Low dietary Cu decreased hepatic and serum Cu (P ≤ 0.05), promoted lipid peroxidation and induced NAFLD-like histopathology, while the combined factors also induced fasting hepatic insulin resistance and liver damage. Neither low Cu nor 30% sucrose in the diet led to enhanced weight gain. Taken together, transcript profiles, histological and biochemical data indicate that low Cu and high sucrose promote hepatic gene expression and physiological responses associated with NAFLD and NASH, even in the absence of obesity or severe steatosis.


Assuntos
Cobre/deficiência , Sacarose Alimentar/efeitos adversos , Inflamação/etiologia , Lipogênese/fisiologia , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Nutrigenômica , Animais , Cobre/administração & dosagem , Dieta , Modelos Animais de Doenças , Expressão Gênica , Inflamação/genética , Resistência à Insulina , Peroxidação de Lipídeos , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/genética , Masculino , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Ratos , Ratos Wistar
14.
Metallomics ; 4(7): 660-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22565294

RESUMO

Wilson's disease (WD) is an inherited disorder of copper metabolism characterized by liver disease and/or neurologic and psychiatric pathology. The disease is a result of mutation in ATP7B, which encodes the ATP7B copper transporting ATPase. Loss of copper transport function by ATP7B results in copper accumulation primarily in the liver, but also in other organs including the brain. Studies in the Atp7b(-/-) mouse model of WD revealed specific transcript and metabolic changes that precede development of liver pathology, most notably downregulation of transcripts in the cholesterol biosynthetic pathway. In order to gain insight into the molecular mechanisms of transcriptomic and metabolic changes, we used a systems approach analysing the pre-symptomatic hepatic nuclear proteome and liver metabolites. We found that ligand-activated nuclear receptors FXR/NR1H4 and GR/NR3C1 and nuclear receptor interacting partners are less abundant in Atp7b(-/-) hepatocyte nuclei, while DNA repair machinery and the nucleus-localized glutathione peroxidase, SelH, are more abundant. Analysis of metabolites revealed an increase in polyol sugar alcohols, indicating a change in osmotic potential that precedes hepatocyte swelling observed later in disease. This work is the first application of quantitative Multidimensional Protein Identification Technology (MuDPIT) to a model of WD to investigate protein-level mechanisms of WD pathology. The systems approach using "shotgun" proteomics and metabolomics in the context of previous transcriptomic data reveals molecular-level mechanisms of WD development and facilitates targeted analysis of hepatocellular copper toxicity.


Assuntos
Adenosina Trifosfatases/deficiência , Proteínas de Transporte de Cátions/deficiência , Degeneração Hepatolenticular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Glucocorticoides/metabolismo , Biologia de Sistemas/métodos , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , ATPases Transportadoras de Cobre , Dano ao DNA/genética , Reparo do DNA/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Redes Reguladoras de Genes/genética , Glucocorticoides/sangue , Degeneração Hepatolenticular/sangue , Degeneração Hepatolenticular/genética , Metabolismo dos Lipídeos/genética , Metaboloma , Camundongos , Pressão Osmótica , Estresse Oxidativo/genética , Proteoma/metabolismo , Processamento Pós-Transcricional do RNA/genética , Transcriptoma/genética
15.
Biometals ; 24(3): 455-66, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21380607

RESUMO

Wilson's disease (WD) is a severe disorder of copper misbalance, which manifests with a wide spectrum of liver pathology and/or neurologic and psychiatric symptoms. WD is caused by mutations in a gene encoding a copper-transporting ATPase ATP7B and is accompanied by accumulation of copper in tissues, especially in the liver. Copper-chelation therapy is available for treatment of WD symptoms and is often successful, however, significant challenges remain with respect to timely diagnostics and treatment of the disease. The lack of genotype-phenotype correlation remains unexplained, the causes of fulminant liver failure are not known, and the treatment of neurologic symptoms is only partially successful, underscoring the need for better understanding of WD mechanisms and factors that influence disease manifestations. Recent gene and protein profiling studies in animal models of WD began to uncover cellular processes that are primarily affected by copper accumulation in the liver. The results of such studies, summarized in this review, revealed new molecular players and pathways (cell cycle and cholesterol metabolism, mRNA splicing and nuclear receptor signaling) linked to copper misbalance. A systems biology approach promises to generate a comprehensive view of WD onset and progression, thus helping with a more fine-tune treatment and monitoring of the disorder.


Assuntos
Cobre/metabolismo , Degeneração Hepatolenticular/fisiopatologia , Biologia de Sistemas , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , ATPases Transportadoras de Cobre , Modelos Animais de Doenças , Redes Reguladoras de Genes , Estudos de Associação Genética , Degeneração Hepatolenticular/genética , Humanos , Mutação , Fenótipo , Transdução de Sinais/fisiologia
16.
J Mol Biol ; 406(1): 44-58, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21146535

RESUMO

Copper is essential to mammalian physiology, and its homeostasis is tightly regulated. In humans, genetic defects in copper excretion result in copper overload and Wilson's disease (WD). Previous studies on the mouse model for WD (Atp7b(-)(/-)) revealed copper accumulation in hepatic nuclei and specific changes in mRNA profile prior to the onset of pathology. To find a molecular link between nuclear copper elevation and changes in hepatic transcriptome, we utilized quantitative ionomic and proteomic approaches. X-ray fluorescence and inductively coupled plasma mass spectrometry analysis indicate that copper in the Atp7b(-/-) nucleus, while highly elevated, does not markedly alter nuclear ion content. Widespread protein oxidation is also not observed, although the glutathione reductase SelH is upregulated, likely to maintain redox balance. We further demonstrate that accumulating copper affects the abundance and/or modification of a distinct subset of nuclear proteins. These proteins populate pathways that are most significantly associated with RNA processing. An alteration in splicing pattern was observed for hnRNP A2/B1, itself the RNA shuttling factor and spliceosome component. Analysis of hnRNP A2/B1 mRNA and protein revealed an increased retention of exon 2 and a selective 2-fold upregulation of a corresponding protein splice variant. Mass spectrometry measurements suggest that the nucleocytoplasmic distribution of RNA binding proteins, including hnRNP A2/B1, is altered in the Atp7b(-/-) liver. We conclude that remodeling of the RNA processing machinery is an important component of cell response to elevated copper that may guide pathology development in the early stages of WD.


Assuntos
Cobre/metabolismo , Degeneração Hepatolenticular/metabolismo , Fígado/metabolismo , Adenosina Trifosfatases/genética , Animais , Proteínas de Transporte de Cátions/genética , Núcleo Celular/química , Núcleo Celular/metabolismo , Cobre/análise , ATPases Transportadoras de Cobre , Modelos Animais de Doenças , Éxons , Feminino , Perfilação da Expressão Gênica , Glutationa Redutase/análise , Glutationa Redutase/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/análise , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Fígado/química , Masculino , Camundongos , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Proteômica , Splicing de RNA , Spliceossomos/química , Spliceossomos/metabolismo , Regulação para Cima
17.
J Biol Chem ; 285(40): 30875-83, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20647314

RESUMO

Wilson disease (WD) is a severe hepato-neurologic disorder that affects primarily children and young adults. WD is caused by mutations in ATP7B and subsequent copper overload. However, copper levels alone do not predict severity of the disease. We demonstrate that temporal and spatial distribution of copper in hepatocytes may play an important role in WD pathology. High resolution synchrotron-based x-ray fluorescence imaging in situ indicates that copper does not continuously accumulate in Atp7b(-/-) hepatocytes, but reaches a limit at 90-300 fmol. The lack of further accumulation is associated with the loss of copper transporter Ctr1 from the plasma membrane and the appearance of copper-loaded lymphocytes and extracellular copper deposits. The WD progression is characterized by changes in subcellular copper localization and transcriptome remodeling. The synchrotron-based x-ray fluorescence imaging and mRNA profiling both point to the key role of nucleus in the initial response to copper overload and suggest time-dependent sequestration of copper in deposits as a protective mechanism. The metabolic pathways, up-regulated in response to copper, show compartmentalization that parallels changes in subcellular copper concentration. In contrast, significant down-regulation of lipid metabolism is observed at all stages of WD irrespective of copper distribution. These observations suggest new stage-specific as well as general biomarkers for WD. The model for the dynamic role of copper in WD is proposed.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/metabolismo , Cobre/metabolismo , Hepatócitos/metabolismo , Degeneração Hepatolenticular/metabolismo , Adenosina Trifosfatases/genética , Adulto , Animais , Biomarcadores/metabolismo , Proteínas de Transporte de Cátions/genética , Membrana Celular/genética , Membrana Celular/patologia , Criança , Pré-Escolar , Transportador de Cobre 1 , ATPases Transportadoras de Cobre , Modelos Animais de Doenças , Hepatócitos/patologia , Degeneração Hepatolenticular/genética , Degeneração Hepatolenticular/patologia , Humanos , Metabolismo dos Lipídeos/genética , Linfócitos/metabolismo , Linfócitos/patologia , Camundongos , Camundongos Knockout
18.
New Phytol ; 182(4): 799-816, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19402880

RESUMO

Copper (Cu) is a cofactor in proteins that are involved in electron transfer reactions and is an essential micronutrient for plants. Copper delivery is accomplished by the concerted action of a set of evolutionarily conserved transporters and metallochaperones. As a result of regulation of transporters in the root and the rarity of natural soils with high Cu levels, very few plants in nature will experience Cu in toxic excess in their tissues. However, low Cu bioavailability can limit plant productivity and plants have an interesting response to impending Cu deficiency, which is regulated by an evolutionarily conserved master switch. When Cu supply is insufficient, systems to increase uptake are activated and the available Cu is utilized with economy. A number of Cu-regulated small RNA molecules, the Cu-microRNAs, are used to downregulate Cu proteins that are seemingly not essential. On low Cu, the Cu-microRNAs are upregulated by the master Cu-responsive transcription factor SPL7, which also activates expression of genes involved in Cu assimilation. This regulation allows the most important proteins, which are required for photo-autotrophic growth, to remain active over a wide range of Cu concentrations and this should broaden the range where plants can thrive.


Assuntos
Cobre/metabolismo , Homeostase , Fotossíntese , Raízes de Plantas/metabolismo , Plantas/metabolismo , Solo/química
19.
J Biol Chem ; 284(1): 696-707, 2009 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18940794

RESUMO

Copper metabolism Murr1 domain 1 (COMMD1) is a 21-kDa protein involved in copper export from the liver, NF-kappaB signaling, HIV infection, and sodium transport. The precise function of COMMD and the mechanism through which COMMD1 performs its multiple roles are not understood. Recombinant COMMD1 is a soluble protein, yet in cells COMMD1 is largely seen as targeted to cellular membranes. Using co-localization with organelle markers and cell fractionation, we determined that COMMD1 is located in the vesicles of the endocytic pathway, whereas little COMMD1 is detected in either the trans-Golgi network or lysosomes. The mechanism of COMMD1 recruitment to cell membranes was investigated using lipid-spotted arrays and liposomes. COMMD1 specifically binds phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) in the absence of other proteins and does not bind structural lipids; the phosphorylation of PtdIns at position 4 is essential for COMMD1 binding. Proteolytic sensitivity and molecular modeling experiments identified two distinct domains in the structure of COMMD1. The C-terminal domain appears sufficient for lipid binding, because both the full-length and C-terminal domain proteins bind to PtdIns(4,5)P2. In native conditions, endogenous COMMD1 forms large oligomeric complexes both in the cytosol and at the membrane; interaction with PtdIns(4,5)P2 increases the stability of oligomers. Altogether, our results suggest that COMMD1 is a scaffold protein in a distinct sub-compartment of endocytic pathway and offer first clues to its role as a regulator of structurally unrelated membrane transporters.


Assuntos
Proteínas de Transporte/metabolismo , Membranas Intracelulares/metabolismo , Fígado/metabolismo , Complexos Multiproteicos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Vesículas Transportadoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Transporte Biológico/fisiologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Cobre/metabolismo , Humanos , Complexos Multiproteicos/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Fosfatidilinositol 4,5-Difosfato/genética , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína/fisiologia , Transdução de Sinais/fisiologia , Vesículas Transportadoras/genética
20.
Arch Biochem Biophys ; 476(1): 22-32, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18534184

RESUMO

The human copper-transporting ATPases (Cu-ATPases) are essential for dietary copper uptake, normal development and function of the CNS, and regulation of copper homeostasis in the body. In a cell, Cu-ATPases maintain the intracellular concentration of copper by transporting copper into intracellular exocytic vesicles. In addition, these P-type ATPases mediate delivery of copper to copper-dependent enzymes in the secretory pathway and in specialized cell compartments such as secretory granules or melanosomes. The multiple functions of human Cu-ATPase necessitate complex regulation of these transporters that is mediated through the presence of regulatory domains in their structure, posttranslational modification and intracellular trafficking, as well as interactions with the copper chaperone Atox1 and other regulatory molecules. In this review, we summarize the current information on the function and regulatory mechanisms acting on human Cu-ATPases ATP7A and ATP7B. Brief comparison with the Cu-ATPase orthologs from other species is included.


Assuntos
Adenosina Trifosfatases/fisiologia , Proteínas de Transporte de Cátions/fisiologia , Cobre/metabolismo , Citoplasma/fisiologia , Adenosina Trifosfatases/química , Animais , Proteínas de Transporte de Cátions/química , ATPases Transportadoras de Cobre , Proteínas Fúngicas/química , Proteínas Fúngicas/fisiologia , Humanos , Espaço Intracelular/metabolismo , Transporte de Íons , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/fisiologia , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...