Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Nutr ; 8: 744825, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869521

RESUMO

Observational studies indicate that selenium may contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Transcriptomic exploration of the aetiology and progression of NAFLD may offer insight into the role selenium plays in this disease. This study compared gene expression levels of known selenoprotein pathways between individuals with a healthy liver to those with NAFLD. Publicly available gene expression databases were searched for studies that measured global gene expression in liver samples from patients with steatosis and non-alcoholic steatohepatitis (NASH) and healthy controls (with [HOC] or without [HC] obesity). A subset of five selenoprotein-related pathways (164 genes) were assessed in the four datasets included in this analysis. The gene TXNRD3 was less expressed in both disease groups when compared with HOC. SCLY and SELENOO were less expressed in NASH when compared with HC. SELENOM, DIO1, GPX2, and GPX3 were highly expressed in NASH when compared to HOC. Disease groups had lower expression of iron-associated transporters and higher expression of ferritin-encoding sub-units, consistent with dysregulation of iron metabolism often observed in NAFLD. Our bioinformatics analysis suggests that the NAFLD liver may have lower selenium levels than a disease-free liver, which may be associated with a disrupted iron metabolism. Our findings indicate that gene expression variation may be associated with the progressive risk of NAFLD.

2.
Front Nutr ; 8: 696024, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262926

RESUMO

Although literature has been consistently showing an increased risk of type 2 diabetes (T2DM) in populations with high exposure to selenium, there is a lack of information quantifying the association between diabetes-related markers and the nutritional status of selenium. Therefore, we aimed to investigate the association between blood selenium concentration and glucose markers in a representative sample of the US population, which is known to have moderate to high exposure to selenium. This cross-sectional analysis included 4,339 participants ≥18 years from the 2013 to 2018 National Health and Nutrition Examination Survey (NHANES). All participants were assessed for whole blood selenium concentration, fasting plasma insulin and glucose, HbA1c, and HOMA-IR (Homeostatic Model Assessment for Insulin Resistance). In this cohort, all participants presented with adequate selenium status [196.2 (SD: 0.9) µg/L] and 867 (15%) had diabetes mellitus. Selenium was positively associated with insulin, glucose and HOMA-IR in models adjusted for age and sex. When the models were further adjusted for smoking status, physical activity, metabolic syndrome and BMI, the associations with insulin and HOMA-IR remained but the association with glucose was no longer significant. A 10 µg/L increase in selenium was associated with 1.5% (95% CI: 0.4-2.6%) increase in insulin and 1.7% (95% CI: 0.5-2.9%) increase in HOMA-IR in fully adjusted models. There was no evidence of an association between selenium and diabetes prevalence. Our findings corroborate the notion that selenium supplementation should not be encouraged in populations with high dietary intake of selenium.

3.
Biochem J ; 478(6): 1227-1239, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33616158

RESUMO

Hepatocytes are essential for maintaining the homeostasis of iron and lipid metabolism in mammals. Dysregulation of either iron or lipids has been linked with serious health consequences, including non-alcoholic fatty liver disease (NAFLD). Considered the hepatic manifestation of metabolic syndrome, NAFLD is characterised by dysregulated lipid metabolism leading to a lipid storage phenotype. Mild to moderate increases in hepatic iron have been observed in ∼30% of individuals with NAFLD; however, direct observation of the mechanism behind this increase has remained elusive. To address this issue, we sought to determine the metabolic consequences of iron loading on cellular metabolism using live cell, time-lapse Fourier transform infrared (FTIR) microscopy utilising a synchrotron radiation source to track biochemical changes. The use of synchrotron FTIR is non-destructive and label-free, and allowed observation of spatially resolved, sub-cellular biochemical changes over a period of 8 h. Using this approach, we have demonstrated that iron loading in AML12 cells induced perturbation of lipid metabolism congruent with steatosis development. Iron-loaded cells had approximately three times higher relative ester carbonyl concentration compared with controls, indicating an accumulation of triglycerides. The methylene/methyl ratio qualitatively suggests the acyl chain length of fatty acids in iron-loaded cells increased over the 8 h period of monitoring compared with a reduction observed in the control cells. Our findings provide direct evidence that mild to moderate iron loading in hepatocytes drives de novo lipid synthesis, consistent with a role for iron in the initial hepatic lipid accumulation that leads to the development of hepatic steatosis.


Assuntos
Rastreamento de Células/métodos , Fígado Gorduroso/patologia , Hepatócitos/metabolismo , Sobrecarga de Ferro/fisiopatologia , Ferro/metabolismo , Síncrotrons/instrumentação , Imagem com Lapso de Tempo/métodos , Animais , Fígado Gorduroso/metabolismo , Hepatócitos/citologia , Metabolismo dos Lipídeos , Camundongos , Microscopia
4.
Microorganisms ; 10(1)2021 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-35056469

RESUMO

Acidihalobacter aeolianus is an acidophilic, halo-tolerant organism isolated from a marine environment near a hydrothermal vent, an ecosystem whereby levels of salinity and total dissolved salts are constantly fluctuating creating ongoing cellular stresses. In order to survive these continuing changes, the synthesis of compatible solutes-also known as organic osmolytes-is suspected to occur, aiding in minimising the overall impact of environmental instability. Previous studies on A. aeolianus identified genes necessary for the accumulation of proline, betaine and ectoine, which are known to act as compatible solutes in other halophilic species. In this study, the impact of increasing the osmotic stress as well as the toxic ion effect was investigated by subjecting A. aeolianus to concentrations of NaCl and MgSO4 up to 1.27 M. Exposure to high concentrations of Cl- resulted in the increase of ectC expression in log-phase cells with a corresponding accumulation of ectoine at stationary phase. Osmotic stress via MgSO4 exposure did not trigger the same up-regulation of ectC or accumulation of ectoine, indicating the transcriptionally regulated response against osmotic stress was induced by chloride toxicity. These findings have highlighted how the adaptive properties of halo-tolerant organisms in acidic environments are likely to differ and are dependent on the initial stressor.

5.
Cell Immunol ; 337: 42-47, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30770093

RESUMO

The resurgence of whooping cough reflects novel genetic variants of Bordetella pertussis and inadequate protection conferred by current acellular vaccines (aP). Biofilm is a source of novel vaccine candidates, including membrane protein assembly factor (BamB) and lipopolysaccharide assembly protein (LptD). Responses of BALB/c mice to candidate vaccines included IFN-γ and IL-17a production by spleen and lymph node cells, and serum IgG1 and IgG2a reactive with whole bacteria or aP. Protection was determined using bacterial cultured from lungs of vaccinated mice challenged with virulent B. pertussis. Mice vaccinated with biofilm produced efficient IFN-γ responses and more IL-17a and IgG2a than mice vaccinated with planktonic cells, aP or adjuvant alone. Vaccination with aP produced abundant IgG1 with little IgG2a. Mice vaccinated with aP plus BamB and LptD retained lower bacterial loads than mice vaccinated with aP alone. Whooping cough vaccines formulated with biofilm antigens, including BamB and LptD, may have clinical value.


Assuntos
Bordetella pertussis/imunologia , Imunogenicidade da Vacina/imunologia , Vacinas Acelulares/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antibacterianos/sangue , Antígenos/imunologia , Biofilmes , Modelos Animais de Doenças , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interferon gama/imunologia , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Vacina contra Coqueluche/imunologia , Baço/imunologia , Vacinação/métodos , Coqueluche/imunologia
6.
Med Microbiol Immunol ; 207(1): 3-26, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29164393

RESUMO

Despite high vaccine coverage, whooping cough caused by Bordetella pertussis remains one of the most common vaccine-preventable diseases worldwide. Introduction of whole-cell pertussis (wP) vaccines in the 1940s and acellular pertussis (aP) vaccines in 1990s reduced the mortality due to pertussis. Despite induction of both antibody and cell-mediated immune (CMI) responses by aP and wP vaccines, there has been resurgence of pertussis in many countries in recent years. Possible reasons hypothesised for resurgence have ranged from incompliance with the recommended vaccination programmes with the currently used aP vaccine to infection with a resurged clinical isolates characterised by mutations in the virulence factors, resulting in antigenic divergence with vaccine strain, and increased production of pertussis toxin, resulting in dampening of immune responses. While use of these vaccines provide varying degrees of protection against whooping cough, protection against infection and transmission appears to be less effective, warranting continuation of efforts in the development of an improved pertussis vaccine formulations capable of achieving this objective. Major approaches currently under evaluation for the development of an improved pertussis vaccine include identification of novel biofilm-associated antigens for incorporation in current aP vaccine formulations, development of live attenuated vaccines and discovery of novel non-toxic adjuvants capable of inducing both antibody and CMI. In this review, the potential roles of different accredited virulence factors, including novel biofilm-associated antigens, of B. pertussis in the evolution, formulation and delivery of improved pertussis vaccines, with potential to block the transmission of whooping cough in the community, are discussed.


Assuntos
Antígenos de Bactérias/imunologia , Bordetella pertussis/imunologia , Transmissão de Doença Infecciosa/prevenção & controle , Vacina contra Coqueluche/imunologia , Fatores de Virulência/imunologia , Coqueluche/prevenção & controle , Bordetella pertussis/patogenicidade , Descoberta de Drogas/tendências , Humanos , Vacina contra Coqueluche/isolamento & purificação , Coqueluche/epidemiologia
7.
Biofouling ; 32(9): 1141-1152, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27669900

RESUMO

Whooping cough caused by Bordetella pertussis is increasing in several countries despite high vaccine coverage. One potential reason for the resurgence is the emergence of genetic variants of the bacterium. Biofilm formation has recently been associated with the pathogenesis of B. pertussis. Biofilm formation of 21 Western Australian B. pertussis clinical isolates was investigated. All isolates formed thicker biofilms than the reference vaccine strain Tohama I while retaining susceptibility to ampicillin, erythromycin, azithromycin and streptomycin. When two biofilm-forming clinical isolates were compared with Tohama I, minimum bactericidal concentrations of antimicrobial agents increased. Isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic analysis revealed significant differences in protein expression in B. pertussis biofilms, providing an opportunity for identification of novel biofilm-associated antigens for incorporation in current pertussis vaccines to improve their protective efficacy. The study also highlights the importance of determining antibiograms for biofilms to formulate improved antimicrobial therapeutic regimens.

8.
Rare Dis ; 4(1): e1198458, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27500074

RESUMO

We previously demonstrated elevated brain iron levels in myelinated structures and associated cells in a hemochromatosis Hfe (-/-) xTfr2 (mut) mouse model. This was accompanied by altered expression of a group of myelin-related genes, including a suite of genes causatively linked to the rare disease family 'neurodegeneration with brain iron accumulation' (NBIA). Expanded data mining and ontological analyses have now identified additional myelin-related transcriptome changes in response to brain iron loading. Concordance between the mouse transcriptome changes and human myelin-related gene expression networks in normal and NBIA basal ganglia testifies to potential clinical relevance. These analyses implicate, among others, genes linked to various rare central hypomyelinating leukodystrophies and peripheral neuropathies including Pelizaeus-Merzbacher-like disease and Charcot-Marie-Tooth disease as well as genes linked to other rare neurological diseases such as Niemann-Pick disease. The findings may help understand interrelationships of iron and myelin in more common conditions such as hemochromatosis, multiple sclerosis and various psychiatric disorders.

9.
Microarrays (Basel) ; 2(2): 131-52, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-27605185

RESUMO

While Illumina microarrays can be used successfully for detecting small gene expression changes due to their high degree of technical replicability, there is little information on how different normalization and differential expression analysis strategies affect outcomes. To evaluate this, we assessed concordance across gene lists generated by applying different combinations of normalization strategy and analytical approach to two Illumina datasets with modest expression changes. In addition to using traditional statistical approaches, we also tested an approach based on combinatorial optimization. We found that the choice of both normalization strategy and analytical approach considerably affected outcomes, in some cases leading to substantial differences in gene lists and subsequent pathway analysis results. Our findings suggest that important biological phenomena may be overlooked when there is a routine practice of using only one approach to investigate all microarray datasets. Analytical artefacts of this kind are likely to be especially relevant for datasets involving small fold changes, where inherent technical variation-if not adequately minimized by effective normalization-may overshadow true biological variation. This report provides some basic guidelines for optimizing outcomes when working with Illumina datasets involving small expression changes.

10.
J Alzheimers Dis ; 30(4): 791-803, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22466002

RESUMO

Iron abnormalities are observed in the brains of Alzheimer's disease (AD) patients, but it is unclear whether common disorders of systemic iron overload such as hemochromatosis alter risks of AD. We used microarrays and real-time reverse transcription-PCR to investigate changes in the brain transcriptome of adult Hfe-/- mice, a model of hemochromatosis, relative to age- and gender-matched wildtype controls. Classification by functional pathway analysis revealed transcript changes for various genes important in AD. There were decreases of up to 9-fold in transcripts for amyloid-ß protein precursor, tau, apolipoprotein E, presenilin 1, and various other γ-secretase components, as well as Notch signaling pathway molecules. This included decreased transcripts for 'hairy and enhancer of split' Hes1 and Hes5, downstream targets of Notch canonical signaling. The reductions in Hes1 and Hes5 transcripts provide evidence that the changes in levels of transcripts for γ-secretase components and Notch signaling genes have functional consequences. The effects appeared relatively specific for AD in that few genes pertaining to other important neurodegenerative diseases, notably Parkinson's disease and Huntington's disease, or to inflammation, oxidative stress, or apoptosis, showed altered transcript levels. The observed effects on AD-related gene transcripts do not appear to be consistent with increased AD risk in HFE hemochromatosis and might, if anything, be predicted to protect against AD to some extent. As Hfe-/- mice did not have higher brain iron levels than wildtype controls, these studies highlight the need for further research in models of more severe hemochromatosis with brain iron loading.


Assuntos
Doença de Alzheimer/genética , Química Encefálica , Hemocromatose/genética , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/genética , Transcrição Gênica/genética , Doença de Alzheimer/metabolismo , Animais , Química Encefálica/genética , Modelos Animais de Doenças , Redes Reguladoras de Genes/genética , Hemocromatose/metabolismo , Proteína da Hemocromatose , Masculino , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Fatores de Risco
11.
Hepatology ; 56(2): 585-93, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22383097

RESUMO

UNLABELLED: Mutations in hemochromatosis protein (HFE) or transferrin receptor 2 (TFR2) cause hereditary hemochromatosis (HH) by impeding production of the liver iron-regulatory hormone, hepcidin (HAMP). This study examined the effects of disruption of Hfe or Tfr2, either alone or together, on liver iron loading and injury in mouse models of HH. Iron status was determined in Hfe knockout (Hfe(-/-)), Tfr2 Y245X mutant (Tfr2(mut)), and double-mutant (Hfe(-/-) ×Tfr2(mut) ) mice by measuring plasma and liver iron levels. Plasma alanine transaminase (ALT) activity, liver histology, and collagen deposition were evaluated to assess liver injury. Hepatic oxidative stress was assessed by measuring superoxide dismutase (SOD) activity and F(2)-isoprostane levels. Gene expression was measured by real-time polymerase chain reaction. Hfe(-/-) ×Tfr2(mut) mice had elevated hepatic iron with a periportal distribution and increased plasma iron, transferrin saturation, and non-transferrin-bound iron, compared with Hfe(-/-), Tfr2(mut), and wild-type (WT) mice. Hamp1 expression was reduced to 40% (Hfe(-/-) and Tfr2(mut) ) and 1% (Hfe(-/-) ×Tfr2(mut)) of WT values. Hfe(-/-) ×Tfr2(mut) mice had elevated plasma ALT activity and mild hepatic inflammation with scattered aggregates of infiltrating inflammatory cluster of differentiation 45 (CD45)-positive cells. Increased hepatic hydoxyproline levels as well as Sirius red and Masson's Trichrome staining demonstrated advanced portal collagen deposition. Hfe(-/-) and Tfr2(mut) mice had less hepatic inflammation and collagen deposition. Liver F(2) -isoprostane levels were elevated, and copper/zinc and manganese SOD activities decreased in Hfe(-/-) ×Tfr2(mut), Tfr2(mut), and Hfe(-/-) mice, compared with WT mice. CONCLUSION: Disruption of both Hfe and Tfr2 caused more severe hepatic iron overload with more advanced lipid peroxidation, inflammation, and portal fibrosis than was observed with the disruption of either gene alone. The Hfe(-/-) ×Tfr2(mut) mouse model of iron-induced liver injury reflects the liver injury phenotype observed in human HH.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Sobrecarga de Ferro , Hepatopatias , Proteínas de Membrana/metabolismo , Receptores da Transferrina/metabolismo , Alanina Transaminase/sangue , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Proteína Morfogenética Óssea 6/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Expressão Gênica/fisiologia , Proteína da Hemocromatose , Hepcidinas , Antígenos de Histocompatibilidade Classe I/genética , Proteína 1 Inibidora de Diferenciação/genética , Ferro/sangue , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Fígado/patologia , Fígado/fisiologia , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Fenótipo , Receptores da Transferrina/genética
12.
Brain Res ; 1448: 144-52, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22370144

RESUMO

Severe disruption of brain iron homeostasis can cause fatal neurodegenerative disease, however debate surrounds the neurologic effects of milder, more common iron loading disorders such as hereditary hemochromatosis, which is usually caused by loss-of-function polymorphisms in the HFE gene. There is evidence from both human and animal studies that HFE gene variants may affect brain function and modify risks of brain disease. To investigate how disruption of HFE influences brain transcript levels, we used microarray and real-time reverse transcription polymerase chain reaction to assess the brain transcriptome in Hfe(-/-) mice relative to wildtype AKR controls (age 10 weeks, n≥4/group). The Hfe(-/-) mouse brain showed numerous significant changes in transcript levels (p<0.05) although few of these related to proteins directly involved in iron homeostasis. There were robust changes of at least 2-fold in levels of transcripts for prominent genes relating to transcriptional regulation (FBJ osteosarcoma oncogene Fos, early growth response genes), neurotransmission (glutamate NMDA receptor Grin1, GABA receptor Gabbr1) and synaptic plasticity and memory (calcium/calmodulin-dependent protein kinase IIα Camk2a). As previously reported for dietary iron-supplemented mice, there were altered levels of transcripts for genes linked to neuronal ceroid lipofuscinosis, a disease characterized by excessive lipofuscin deposition. Labile iron is known to enhance lipofuscin generation which may accelerate brain aging. The findings provide evidence that iron loading disorders can considerably perturb levels of transcripts for genes essential for normal brain function and may help explain some of the neurologic signs and symptoms reported in hemochromatosis patients.


Assuntos
Química Encefálica/genética , Antígenos de Histocompatibilidade Classe I/fisiologia , Sobrecarga de Ferro/genética , Proteínas de Membrana/fisiologia , Transcriptoma/genética , Animais , Suplementos Nutricionais , Hemocromatose/genética , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Ferro/metabolismo , Ferro da Dieta/farmacologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Memória/fisiologia , Camundongos , Camundongos Knockout , Análise em Microsséries , Transtornos dos Movimentos/genética , Plasticidade Neuronal/genética , Ferroproteínas não Heme/sangue , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Elementos Reguladores de Transcrição/genética , Transmissão Sináptica/genética
13.
Hepatology ; 52(2): 462-71, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20683946

RESUMO

UNLABELLED: Iron and cholesterol are both essential metabolites in mammalian systems, and too much or too little of either can have serious clinical consequences. In addition, both have been associated with steatosis and its progression, contributing, inter alia, to an increase in hepatic oxidative stress. The interaction between iron and cholesterol is unclear, with no consistent evidence emerging with respect to changes in plasma cholesterol on the basis of iron status. We sought to clarify the role of iron in lipid metabolism by studying the effects of iron status on hepatic cholesterol synthesis in mice with differing iron status. Transcripts of seven enzymes in the cholesterol biosynthesis pathway were significantly up-regulated with increasing hepatic iron (R(2) between 0.602 and 0.164), including those of the rate-limiting enzyme, 3-hydroxy-3-methylglutarate-coenzyme A reductase (Hmgcr; R(2) = 0.362, P < 0.002). Hepatic cholesterol content correlated positively with hepatic iron (R(2) = 0.255, P < 0.007). There was no significant relationship between plasma cholesterol and either hepatic cholesterol or iron (R(2) = 0.101 and 0.014, respectively). Hepatic iron did not correlate with a number of known regulators of cholesterol synthesis, including sterol-regulatory element binding factor 2 (Srebf2; R(2) = 0.015), suggesting that the increases seen in the cholesterol biosynthesis pathway are independent of Srebf2. Transcripts of genes involved in bile acid synthesis, transport, or regulation did not increase with increasing hepatic iron. CONCLUSION: This study suggests that hepatic iron loading increases liver cholesterol synthesis and provides a new and potentially important additional mechanism by which iron could contribute to the development of fatty liver disease or lipotoxicity.


Assuntos
Colesterol/biossíntese , Ferro/administração & dosagem , Ferro/fisiologia , Animais , Fígado Gorduroso/etiologia , Masculino , Camundongos , Camundongos Endogâmicos AKR
14.
J Hepatol ; 52(3): 425-31, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20133002

RESUMO

BACKGROUND & AIMS: Hereditary haemochromatosis type 3 is caused by mutations in transferrin receptor (TFR) 2. TFR2 has been shown to mediate iron transport in vitro and regulate iron homeostasis. The aim of this study was to determine the role of Tfr2 in iron transport in vivo using a Tfr2 mutant mouse. METHODS: Tfr2 mutant and wild-type mice were injected intravenously with (59)Fe-transferrin and tissue (59)Fe uptake was measured. Tfr1, Tfr2 and ferroportin expression was measured by real-time PCR and Western blot. Cellular localisation of ferroportin was determined by immunohistochemistry. RESULTS: Transferrin-bound iron uptake by the liver and spleen in Tfr2 mutant mice was reduced by 20% and 65%, respectively, whilst duodenal and renal uptake was unchanged compared with iron-loaded wild-type mice. In Tfr2 mutant mice, liver Tfr2 protein was absent, whilst ferroportin protein was increased in non-parenchymal cells and there was a low level of expression in hepatocytes. Tfr1 expression was unchanged compared with iron-loaded wild-type mice. Splenic Tfr2 protein expression was absent whilst Tfr1 and ferroportin protein expression was increased in Tfr2 mutant mice compared with iron-loaded wild-type mice. CONCLUSIONS: A small reduction in hepatic transferrin-bound iron uptake in Tfr2 mutant mice suggests that Tfr2 plays a minor role in liver iron transport and its primary role is to regulate iron metabolism. Increased ferroportin expression due to decreased hepcidin mRNA levels is likely to be responsible for impaired splenic iron uptake in Tfr2 mutant mice.


Assuntos
Hemocromatose/metabolismo , Ferro/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Transporte Biológico/fisiologia , Proteínas de Transporte de Cátions/metabolismo , Modelos Animais de Doenças , Feminino , Hemocromatose/genética , Hepcidinas , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , RNA Mensageiro/metabolismo , Receptores da Transferrina/genética , Baço/metabolismo
15.
Am J Physiol Cell Physiol ; 297(6): C1567-75, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19828835

RESUMO

Transferrin receptor (TFR) 1 and 2 are expressed in the liver; TFR1 levels are regulated by cellular iron levels while TFR2 levels are regulated by transferrin saturation. The aims of this study were to 1) determine the relative importance of TFR1 and TFR2 in transferrin-bound iron (TBI) uptake by HuH7 human hepatoma cells and 2) characterize the role of metal-transferrin complexes in the regulation of these receptors. TFR expression was altered by 1) incubation with metal-transferrin (Tf) complexes, 2) TFR1 and TFR2 small interfering RNA knockdown, and 3) transfection with a human TFR2 plasmid. TBI uptake was measured using (59)Fe-(125)I-labeled Tf and mRNA and protein expression by real-time PCR and Western blot analysis, respectively. Fe(2)Tf, Co(2)Tf, and Mn(2)Tf increased TFR2 protein expression, indicating that the upregulation was not specifically regulated by iron-transferrin but also other metal-transferrins. In addition, Co(2)Tf and Mn(2)Tf upregulated TFR1, reduced ferritin, and increased hypoxia-inducible factor-1alpha protein expression, suggesting that TFR1 upregulation was due to a combination of iron deficiency and chemical hypoxia. TBI uptake correlated with changes in TFR1 but not TFR2 expression. TFR1 knockdown reduced iron uptake by 80% while TFR2 knockdown did not affect uptake. At 5 microM transferrin, iron uptake was not affected by combined TFR1 and TFR2 knockdown. Transfection with a hTFR2 plasmid increased TFR2 protein expression, causing a 15-20% increase in iron uptake and ferritin levels. This shows for the first time that TFR-mediated TBI uptake is mediated primarily via TFR1 but not TFR2 and that a high-capacity TFR-independent pathway exists in hepatoma cells.


Assuntos
Antígenos CD/metabolismo , Carcinoma Hepatocelular/metabolismo , Ferro/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Antígenos CD/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Cobalto/metabolismo , Humanos , Ferro/farmacocinética , Neoplasias Hepáticas/patologia , Metais/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores da Transferrina/genética , Transfecção , Regulação para Cima
16.
J Biol Chem ; 284(8): 4796-805, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19068481

RESUMO

The ring contraction process that occurs during cobalamin (vitamin B(12)) biosynthesis is mediated via the action of two enzymes, CobG and CobJ. The first of these generates a tertiary alcohol at the C-20 position of precorrin-3A by functioning as a monooxygenase, a reaction that also forms a gamma lactone with the acetic acid side chain on ring A. The product, precorrin-3B, is then acted upon by CobJ, which methylates at the C-17 position and promotes ring contraction of the macrocycle by catalyzing a masked pinacol rearrangement. Here, we report the characterization of CobG enzymes from Pseudomonas denitrificans and Brucella melitensis. We show that both contain a [4Fe-4S] center as well as a mononuclear non-heme iron. Although both enzymes are active in vivo, the P. denitrificans enzyme was found to be inactive in vitro. Further analysis of this enzyme revealed that the mononuclear non-heme iron was not reducible, and it was concluded that it is rapidly inactivated once it is released from the bacterial cell. In contrast, the B. melitensis enzyme was found to be fully active in vitro and the mononuclear non-heme iron was reducible by dithionite. The reduced mononuclear non-heme was able to react with the oxygen analogue NO, but only in the presence of the substrate precorrin-3A. The cysteine residues responsible for binding the Fe-S center were identified by site-directed mutagenesis. A mechanism for CobG is presented.


Assuntos
Proteínas de Bactérias/química , Brucella melitensis/enzimologia , Cobamidas/química , Oxigenases de Função Mista/química , Oxigenases/química , Pseudomonas/enzimologia , Aerobiose , Proteínas de Bactérias/genética , Brucella melitensis/genética , Domínio Catalítico/fisiologia , Cobamidas/genética , Ferro/química , Oxigenases de Função Mista/genética , Mutagênese Sítio-Dirigida , Óxido Nítrico/química , Oxirredução , Oxigenases/genética , Pseudomonas/genética , Enxofre/química
17.
Hepatology ; 47(5): 1737-44, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18393371

RESUMO

UNLABELLED: HFE-related hereditary hemochromatosis results in hepatic iron overload. Hepatocytes acquire transferrin-bound iron via transferrin receptor (Tfr) 1 and Tfr1-independent pathways (possibly Tfr2-mediated). In this study, the role of Hfe in the regulation of hepatic transferrin-bound iron uptake by these pathways was investigated using Hfe knockout mice. Iron and transferrin uptake by hepatocytes from Hfe knockout, non-iron-loaded and iron-loaded wild-type mice were measured after incubation with 50 nM (125)I-Tf-(59)Fe (Tfr1 pathway) and 5 microM (125)I-Tf-(59)Fe (Tfr1-independent or putative Tfr2 pathway). Tfr1 and Tfr2 messenger RNA (mRNA) and protein expression were measured by real-time polymerase chain reaction and western blotting, respectively. Tfr1-mediated iron and transferrin uptake by Hfe knockout hepatocytes were increased by 40% to 70% compared with iron-loaded wild-type hepatocytes with similar iron levels and Tfr1 expression. Iron and transferrin uptake by the Tfr1-independent pathway was approximately 100-fold greater than by the Tfr1 pathway and was not affected by the absence of Hfe. Diferric transferrin increased hepatocyte Tfr2 protein expression, resulting in a small increase in transferrin but not iron uptake by the Tfr1-independent pathway. CONCLUSION: Tfr1-mediated iron uptake is regulated by Hfe in hepatocytes. The Tfr1-independent pathway exhibited a much greater capacity for iron uptake than the Tfr1 pathway but it was not regulated by Hfe. Diferric transferrin up-regulated hepatocyte Tfr2 protein expression but not iron uptake, suggesting that Tfr2 may have a limited role in the Tfr1-independent pathway.


Assuntos
Hepatócitos/fisiologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Ferro/metabolismo , Proteínas de Membrana/fisiologia , Transferrina/metabolismo , Animais , Transporte Biológico , Técnicas de Cultura de Células , Primers do DNA , Proteína da Hemocromatose , Hepatócitos/citologia , Hepatócitos/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Cinética , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ligação Proteica , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/genética
18.
J Hepatol ; 48(2): 327-34, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18083267

RESUMO

BACKGROUND/AIMS: Transferrin receptor 2 appears to have dual roles in iron metabolism; one is signalling, the other is iron transport. It is sensitive to high levels of diferric transferrin, which is associated with disorders of iron overload. Also present in these disorders are increased levels of plasma non-transferrin-bound iron. This study sought to clarify the role of transferrin receptor 2 in the uptake of transferrin-bound and non-transferrin-bound iron. METHODS: Variant Chinese Hamster Ovary (CHO) cells, transfected with transferrin receptor 2, were incubated with radio-labelled transferrin-bound or non-transferrin-bound iron. Competition studies were performed in the presence of unlabelled dimetallic transferrin; knockdown was performed using specific siRNA. RESULTS: Cells expressing transferrin receptor 2 bound and internalised transferrin and transferrin-bound iron. Transferrin recycling occurred with an average cycling time of 11-15 min. Interestingly, the presence of transferrin receptor 2 was also associated with uptake of non-transferrin-bound iron which was inhibited by unlabelled transferrin-bound metals. Knockdown reduced transferrin-bound and non-transferrin-bound iron uptake by approximately 60%. CONCLUSIONS: Transferrin receptor 2 mediates transferrin-bound iron uptake by receptor-mediated endocytosis. It is also involved in the uptake of non-transferrin-bound iron and the inhibition of non-transferrin-bound iron uptake by diferric transferrin in CHO cells.


Assuntos
Ferro/metabolismo , Receptores da Transferrina/fisiologia , Transferrina/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , RNA Interferente Pequeno/farmacologia
19.
Crit Rev Clin Lab Sci ; 44(5-6): 413-59, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17943492

RESUMO

While iron is an essential trace element required by nearly all living organisms, deficiencies or excesses can lead to pathological conditions such as iron deficiency anemia or hemochromatosis, respectively. A decade has passed since the discovery of the hemochromatosis gene, HFE, and our understanding of hereditary hemochromatosis (HH) and iron metabolism in health and a variety of diseases has progressed considerably. Although HFE-related hemochromatosis is the most widespread, other forms of HH have subsequently been identified. These forms are not attributed to mutations in the HFE gene but rather to mutations in genes involved in the transport, storage, and regulation of iron. This review is an overview of cellular iron metabolism and regulation, describing the function of key proteins involved in these processes, with particular emphasis on the liver's role in iron homeostasis, as it is the main target of iron deposition in pathological iron overload. Current knowledge on their roles in maintaining iron homeostasis and how their dysregulation leads to the pathogenesis of HH are discussed.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Distúrbios do Metabolismo do Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Proteínas de Membrana/metabolismo , Mutação , Anemia Ferropriva/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Transporte Biológico , Proteínas de Transporte de Cátions/metabolismo , Ceruloplasmina/metabolismo , Proteínas Ligadas por GPI , Hemocromatose/genética , Proteína da Hemocromatose , Hepcidinas , Humanos , Absorção Intestinal , Sobrecarga de Ferro/metabolismo , Ferro da Dieta/farmacocinética , Proteínas de Ligação ao Ferro/genética , Mutação/genética , Receptores da Transferrina/deficiência , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Transferrina/deficiência , Transferrina/genética , Transferrina/metabolismo
20.
World J Gastroenterol ; 13(35): 4725-36, 2007 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-17729394

RESUMO

The liver plays a central role in iron metabolism. It is the major storage site for iron and also expresses a complex range of molecules which are involved in iron transport and regulation of iron homeostasis. An increasing number of genes associated with hepatic iron transport or regulation have been identified. These include transferrin receptors (TFR1 and 2), a ferrireductase (STEAP3), the transporters divalent metal transporter-1 (DMT1) and ferroportin (FPN) as well as the haemochromatosis protein, HFE and haemojuvelin (HJV), which are signalling molecules. Many of these genes also participate in iron regulatory pathways which focus on the hepatic peptide hepcidin. However, we are still only beginning to understand the complex interactions between liver iron transport and iron homeostasis. This review outlines our current knowledge of molecules of iron metabolism and their roles in iron transport and regulation of iron homeostasis.


Assuntos
Ferro/metabolismo , Fígado/metabolismo , Transporte Biológico/genética , Transporte Biológico/fisiologia , Homeostase/genética , Homeostase/fisiologia , Humanos , Células de Kupffer/metabolismo , Fígado/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...