Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Benef Microbes ; 11(4): 391-401, 2020 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-32720833

RESUMO

Salmonella Heidelberg is one of the most common serovar causing foodborne illnesses. To limit the development of digestive bacterial infection, food supplements containing probiotic bacteria can be proposed. Commensal non-toxigenic Bacteroides fragilis has recently been suggested as a next-generation probiotic candidate. By using an original triple co-culture model including Caco-2 cells (representing human enterocytes), HT29-MTX (representing mucus-secreting goblet cells), and M cells differentiated from Caco-2 by addition of Raji B lymphocytes, bacterial translocation was evaluated. The data showed that S. Heidelberg could translocate in the triple co-culture model with high efficiency, whereas for B. fragilis a weak translocation was obtained. When cells were exposed to both bacteria, S. Heidelberg translocation was inhibited. The cell-free supernatant of B. fragilis also inhibited S. Heidelberg translocation without impacting epithelial barrier integrity. This supernatant did not affect the growth of S. Heidelberg. The non-toxigenic B. fragilis confers health benefits to the host by reducting bacterial translocation. These results suggested that the multicellular model provides an efficient in vitro model to evaluate the translocation of pathogens and to screen for probiotics that have a potential inhibitory effect on this translocation.


Assuntos
Translocação Bacteriana , Bacteroides fragilis/fisiologia , Mucosa Intestinal/microbiologia , Salmonella/fisiologia , Translocação Bacteriana/efeitos dos fármacos , Bacteroides fragilis/metabolismo , Células CACO-2 , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células HT29 , Humanos , Mucosa Intestinal/citologia , Interações Microbianas , Modelos Biológicos , Probióticos/metabolismo , Probióticos/farmacologia
2.
Osteoporos Int ; 29(8): 1905-1915, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29721575

RESUMO

We aimed to study the mechanisms involved in bone-related iron impairment by using the osteoblast-like MG-63 cell line. Our results indicate that iron impact the S1P/S1PR signalizing axis and suggest that iron can affect the S1P process and favor the occurrence of osteoporosis during chronic iron overload. INTRODUCTION: Systemic iron excess favors the development of osteoporosis, especially during genetic hemochromatosis. The cellular mechanisms involved are still unclear despite numerous data supporting a direct effect of iron on bone biology. Therefore, the aim of this study was to characterize mechanisms involved in the iron-related osteoblast impairment. METHODS: We studied, by using the MG-63 cell lines, the effect of iron excess on SPNS2 gene expression which was previously identified by us as potentially iron-regulated. Cell-type specificity was investigated with hepatoma HepG2 and enterocyte-like Caco-2 cell lines as well as in iron-overloaded mouse liver. The SPNS2-associated function was also investigated in MG-63 cells by fluxomic strategy which led us to determinate the S1P efflux in iron excess condition. RESULTS: We showed in MG-63 cells that iron exposure strongly increased the mRNA level of the SPNS2 gene. This was not observed in HepG2, in Caco-2 cells, and in mouse livers. Fluxomic study performed concomitantly on MG-63 cells revealed an unexpected decrease in the cellular capacity to export S1P. Iron excess did not modulate SPHK1, SPHK2, SGPL1, or SGPP1 gene expression, but decreased COL1A1 and S1PR1 mRNA levels, suggesting a functional implication of low extracellular S1P concentration on the S1P/S1PR signalizing axis. CONCLUSIONS: Our results indicate that iron impacts the S1P/S1PR signalizing axis in the MG-63 cell line and suggest that iron can affect the bone-associated S1P pathway and favor the occurrence of osteoporosis during chronic iron overload.


Assuntos
Proteínas de Transporte de Ânions/biossíntese , Sobrecarga de Ferro/metabolismo , Lisofosfolipídeos/metabolismo , Osteoblastos/metabolismo , Esfingosina/análogos & derivados , Regulação para Cima/fisiologia , Animais , Proteínas de Transporte de Ânions/genética , Células CACO-2 , Células Cultivadas , Colágeno Tipo I/biossíntese , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Inativação Gênica , Hemocromatose/metabolismo , Células Hep G2 , Humanos , Ferro/metabolismo , Ferro/farmacologia , Fígado/metabolismo , Masculino , Camundongos Knockout , Osteoblastos/efeitos dos fármacos , RNA Mensageiro/genética , Esfingosina/metabolismo
3.
Diabetes Metab ; 42(2): 105-11, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26677772

RESUMO

AIM: The aim of this study was to assess interactions between glucose and iron homoeostasis in the adipose tissue (AT) of obese subjects. METHODS: A total of 46 obese patients eligible for bariatric surgery were recruited into the study. Anthropometric and biochemical characteristics were assessed, and biopsies of subcutaneous (SCAT) and visceral adipose tissue (VAT) performed. The mRNA levels of genes involved in iron and glucose homoeostasis were measured in their AT and compared with a pool of control samples. RESULTS: Gene expression of hepcidin (HAMP) was significantly increased in the SCAT and VAT of obese patients, while transferrin receptor (TFRC) expression was reduced, compared with non-obese controls, suggesting a higher iron load in obese patients. Also, mRNA levels of adiponectin (ADIPOQ) were decreased in both SCAT and VAT in obese patients, and correlated negatively with hepcidin expression, while adiponectin expression was positively correlated with TFRC expression in both SCAT and VAT. Interestingly, TFRC expression in VAT correlated negatively with several metabolic parameters, such as fasting blood glucose and LDL cholesterol. CONCLUSION: Iron content appears to be increased in the SCAT and VAT of obese patients, and negatively correlated with adiponectin expression, which could be contributing to insulin resistance and the metabolic complications of obesity.


Assuntos
Adiponectina/metabolismo , Tecido Adiposo/metabolismo , Ferro/sangue , Obesidade Mórbida/metabolismo , Adiponectina/análise , Adiponectina/genética , Tecido Adiposo/química , Adulto , Feminino , Hepcidinas/análise , Hepcidinas/genética , Hepcidinas/metabolismo , Humanos , Resistência à Insulina , Masculino , Pessoa de Meia-Idade , Obesidade Mórbida/sangue , Obesidade Mórbida/epidemiologia , Estudos Prospectivos
4.
Amino Acids ; 44(3): 869-77, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23053023

RESUMO

An occurrence and a magnitude of alcoholic liver diseases depend on the balance between ethanol-induced injury and liver regeneration. Like ethanol, polyamines including putrescine, spermidine, and spermine modulate cell proliferation. Thus, the purpose of this study was to evaluate the relationship between effect of ethanol on hepatocyte (HC) proliferation and polyamine metabolism using the HepaRG cell model. Results showed that ethanol effect in proliferating HepaRG cells was associated with a decrease in intracellular polyamine levels and ornithine decarboxylase (ODC) activity. Ethanol also induced disorders in expression of genes coding for polyamine-metabolizing enzymes. The α-difluoromethyl ornithine, an irreversible inhibitor of ODC, amplified ethanol toxicity on cell viability, protein level, and DNA synthesis through accentuation of polyamine depletion in proliferating HepaRG cells. Conversely, putrescine reversed ethanol effect on cell proliferation parameters. In conclusion, this study suggested that ethanol effect on HC proliferation was closely related to polyamine metabolism and that manipulation of this metabolism by putrescine could protect against the anti-proliferative activity of ethanol.


Assuntos
Proliferação de Células , Etanol/toxicidade , Hepatócitos/citologia , Hepatócitos/metabolismo , Poliaminas/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Humanos
6.
Osteoporos Int ; 23(10): 2435-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22237814

RESUMO

UNLABELLED: In order to understand mechanisms involved in osteoporosis observed during iron overload diseases, we analyzed the impact of iron on a human osteoblast-like cell line. Iron exposure decreases osteoblast phenotype. HHIPL-2 is an iron-modulated gene which could contribute to these alterations. Our results suggest osteoblast impairment in iron-related osteoporosis. INTRODUCTION: Iron overload may cause osteoporosis. An iron-related decrease in osteoblast activity has been suggested. METHODS: We investigated the effect of iron exposure on human osteoblast cells (MG-63) by analyzing the impact of ferric ammonium citrate (FAC) and iron citrate (FeCi) on the expression of genes involved in iron metabolism or associated with osteoblast phenotype. A transcriptomic analysis was performed to identify iron-modulated genes. RESULTS: FAC and FeCi exposure modulated cellular iron status with a decrease in TFRC mRNA level and an increase in intracellular ferritin level. FAC increased ROS level and caspase 3 activity. Ferroportin, HFE and TFR2 mRNAs were expressed in MG-63 cells under basal conditions. The level of ferroportin mRNA was increased by iron, whereas HFE mRNA level was decreased. The level of mRNA alpha 1 collagen type I chain, osteocalcin and the transcriptional factor RUNX2 were decreased by iron. Transcriptomic analysis revealed that the mRNA level of HedgeHog Interacting Protein Like-2 (HHIPL-2) gene, encoding an inhibitor of the hedgehog signaling pathway, was decreased in the presence of FAC. Specific inhibition of HHIPL-2 expression decreased osteoblast marker mRNA levels. Purmorphamine, hedgehog pathway activator, increased the mRNA level of GLI1, a target gene for the hedgehog pathway, and decreased osteoblast marker levels. GLI1 mRNA level was increased under iron exposure. CONCLUSION: We showed that in human MG-63 cells, iron exposure impacts iron metabolism and osteoblast gene expression. HHIPL-2 gene expression modulation may contribute to these alterations. Our results support a role of osteoblast impairment in iron-related osteoporosis.


Assuntos
Sobrecarga de Ferro/metabolismo , Osteoblastos/metabolismo , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Ácido Cítrico , Compostos Férricos/farmacologia , Compostos Ferrosos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Sobrecarga de Ferro/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Osteoblastos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Compostos de Amônio Quaternário/farmacologia
7.
Osteoporos Int ; 22(8): 2313-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20976594

RESUMO

UNLABELLED: Genetic hemochromatosis is a cause of osteoporosis; mechanisms leading to iron-related bone loss are not fully characterized. We assessed the bone phenotype of HFE (-/-) male mice, a mouse model of hemochromatosis. They had a phenotype of osteoporosis with low bone mass and alteration of the bone microarchitecture. INTRODUCTION: Genetic hemochromatosis is a cause of osteoporosis. However, the mechanisms leading to iron-related bone loss are not fully characterized. Recent human data have not supported the hypothesis of hypogonadism involvement. The direct role of iron on bone metabolism has been suggested. METHODS: Our aim was to assess the bone phenotype of HFE (-/-) male mice, a mouse model of human hemochromatosis, by using microcomputed tomography and histomorphometry. HFE (-/-) animals were sacrificed at 6 and 12 months and compared to controls. RESULTS: There was a significant increase in hepatic iron concentration and bone iron content in HFE (-/-) mice. No detectable Perls' staining was found in the controls' trabeculae. Trabecular bone volume (BV/TV) was significantly lower in HFE (-/-) mice at 6 and 12 months compared to the corresponding wild-type mice: 9.88 ± 0.82% vs 12.82 ± 0.61% (p = 0.009) and 7.18 ± 0.68% vs 10.4 ± 0.86% (p = 0.015), respectively. In addition, there was an impairment of the bone microarchitecture in HFE (-/-) mice. Finally, we found a significant increase in the osteoclast number in HFE (-/-) mice: 382.5 ± 36.75 vs 273.4 ± 20.95 ¢/mm(2) (p = 0.004) at 6 months and 363.6 ± 22.35 vs 230.8 ± 18.7 ¢/mm(2) (p = 0.001) at 12 months in HFE (-/-) mice vs controls. CONCLUSION: Our data show that HFE (-/-) male mice develop a phenotype of osteoporosis with low bone mass and alteration of the microarchitecture. They suggest that there is a relationship between bone iron overload and the increase of the osteoclast number in these mice. These findings are in accordance with clinical observations in humans exhibiting genetic hemochromatosis and support a role of excess iron in relation to genetic hemochromatosis in the development of osteoporosis in humans.


Assuntos
Modelos Animais de Doenças , Hemocromatose/complicações , Hemocromatose/genética , Osteoporose/patologia , Animais , Hemocromatose/metabolismo , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Ferro/metabolismo , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoclastos/patologia , Osteoporose/etiologia , Osteoporose/metabolismo , Fenótipo , Tíbia/metabolismo , Tíbia/patologia , Microtomografia por Raio-X/métodos
8.
Pathol Biol (Paris) ; 58(5): 316-23, 2010 Oct.
Artigo em Francês | MEDLINE | ID: mdl-19942367

RESUMO

The field of hereditary iron overload has known, in the recent period, deep changes mainly related to major advances in molecular biology. It encompasses now a series of genetic entities. The mechanistic understanding of iron overload development and iron toxicity has greatly improved. The diagnostic approach has become essentially noninvasive with a major role for biological tests. From the therapeutic viewpoint, the phlebotomy treatment is now enriched by the possibility of resorting to oral chelation and by innovative perspectives directly linked to our improvement in the molecular understanding of these diseases.


Assuntos
Sobrecarga de Ferro/genética , Peptídeos Catiônicos Antimicrobianos/deficiência , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Ceruloplasmina/deficiência , Ceruloplasmina/genética , Terapia por Quelação , Previsões , Aconselhamento Genético , Hemocromatose/classificação , Hemocromatose/diagnóstico , Hemocromatose/tratamento farmacológico , Hemocromatose/genética , Hemocromatose/terapia , Proteína da Hemocromatose , Hemossiderose/genética , Hemossiderose/metabolismo , Hepcidinas , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/genética , Sobrecarga de Ferro/diagnóstico , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/fisiopatologia , Sobrecarga de Ferro/terapia , Fígado/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Técnicas de Diagnóstico Molecular , Doenças Neurodegenerativas/genética , Flebotomia
10.
Transfus Clin Biol ; 12(2): 77-82, 2005 Jun.
Artigo em Francês | MEDLINE | ID: mdl-15925529

RESUMO

HFE hemochromatosis is the most frequent genetic iron overload disease. It is linked to the C282Y mutation of the HFE protein, protein encoded by the HFE gene, which is located on chromosome 6. The mechanisms accounting for iron excess are not only digestive hyperabsorption of iron but also excessive recycling of macrophagic iron coming from erythrophagocytosis and secreted into the blood. Both mechanisms are linked to an HFE-related hepatic failure in producing hepcidin, a key hormone of body iron regulation. The marked phenotypic variability of C282Y homozygosity expression is likely related to both genetic and environmental factors. The HFE gene discovery has rendered non invasive the positive diagnostic of HFE hemochromatosis, which is now based first on an increased level of plasma transferrin saturation leading to the request of the HFE mutation. Then, hepatic MRI is a reliable method to quantify iron overload. The HFE gene discovery has also paved the road of an enlarged field of differential diagnoses corresponding to novel entities of non-HFE related genetic iron overload syndromes.


Assuntos
Hemocromatose/diagnóstico , Hemocromatose/etiologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Proteínas de Membrana/fisiologia , Substituição de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/deficiência , Peptídeos Catiônicos Antimicrobianos/fisiologia , Cromossomos Humanos Par 6/genética , Análise Mutacional de DNA , Diagnóstico Diferencial , Duodeno/metabolismo , Regulação da Expressão Gênica , Hemocromatose/genética , Proteína da Hemocromatose , Hepatócitos/metabolismo , Hepcidinas , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Absorção Intestinal , Ferro/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Fagocitose , Fenótipo , Mutação Puntual , Transferrina/análise
11.
Acta Gastroenterol Belg ; 68(1): 33-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15832585

RESUMO

Hereditary Hemochromatosis is an autosomal recessive disease, characterized by chronic iron overload. It is mainly due to mutations of the HFE-1 gene. In the large majority of patients, the substitution of tyrosine for cysteine at amino acid 282 (C282Y) is found at the homozygous state. Since the HFE-1 hemochromatosis identification, several other entities of iron overload have been individualized. In the present article, the frequency, penetrance and pathophysiology of HFE-1 hemochromatosis as well as various clinical presentations resulting from different mutations affecting different proteins involved in iron metabolism are described.


Assuntos
Predisposição Genética para Doença , Hemocromatose/genética , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/genética , Mutação , Receptores da Transferrina/genética , Adulto , Feminino , Regulação da Expressão Gênica , Hemocromatose/diagnóstico , Hemocromatose/terapia , Proteína da Hemocromatose , Humanos , Masculino , Pessoa de Meia-Idade , Biologia Molecular , Prognóstico , Medição de Risco , Índice de Gravidade de Doença
12.
J Biomed Opt ; 9(2): 404-7, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15065908

RESUMO

Infrared fingerprints of molecules in biology contain much information on cells metabolism allowing one to distinguish between healthy and altered tissues. Here, to collect infrared signatures, we used evanescent wave spectroscopy based on an original infrared transmitting tapered glass fiber. A strict control of the fiber diameter in the tapered sensing zone allows high sensitivity and wide spectral range exploration from 800 to 3000 cm(-1). Then, merely in depositing the mouse liver biopsies on the fiber, this device has enable us to differentiate between tumorous and healthy tissues.


Assuntos
Tecnologia de Fibra Óptica , Raios Infravermelhos , Fígado/metabolismo , Análise Espectral , Animais , Testes Diagnósticos de Rotina , Camundongos , Fibras Ópticas , Sensibilidade e Especificidade , Análise Espectral/instrumentação
15.
Gut ; 51(5): 648-53, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12377801

RESUMO

BACKGROUND: Although much progress has been made recently in characterising the proteins involved in duodenal iron trafficking, regulation of intestinal iron transport remains poorly understood. It is not known whether the level of mRNA expression of these recently described molecules is genetically regulated. This is of particular interest however as genetic factors are likely to determine differences in iron status among mouse strains and probably also contribute to the phenotypic variability seen with disruption of the haemochromatosis gene. AIMS: To investigate this issue, we examined concomitant variations in duodenal cytochrome b (Dcytb), divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), hephaestin, stimulator of Fe transport (SFT), HFE, and transferrin receptor 1 (TfR1) transcripts in response to different dietary iron contents in the four mouse strains C57BL/6, DBA/2, CBA, and 129/Sv. SUBJECTS: Six mice of each strain were fed normal levels of dietary iron, six were subjected to the same diet supplemented with 2% carbonyl iron, and six were fed an iron deficient diet. METHODS: Quantification of mRNAs isolated from the duodenum was performed using real time reverse transcription-polymerase chain reaction. RESULTS: There was a significant increase in mRNA expression of Dcytb, DMT1, FPN1, and TfR1 when mice were fed an iron deficient diet, and a significant decrease in mRNA expression of these molecules when mice were fed an iron supplemented diet. Strain to strain differences were observed not only in serum transferrin saturations, with C57BL/6 mice having the lowest values, but also in hepatic iron stores and in duodenal mRNA expression of Dcytb, DMT1, FPN1, hephaestin, HFE, and TfR1. CONCLUSIONS: The results favour some degree of genetic control of mRNA levels of these molecules.


Assuntos
Proteínas de Transporte/genética , Duodeno/metabolismo , Mucosa Intestinal/metabolismo , Ferro da Dieta/administração & dosagem , RNA Mensageiro/análise , Enzimas de Conjugação de Ubiquitina , Animais , Proteínas de Transporte de Cátions/genética , Grupo dos Citocromos b/genética , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Deficiências de Ferro , Proteínas de Ligação ao Ferro/genética , Fígado/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos , Receptores da Transferrina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Transferrina/análise
18.
Biochim Biophys Acta ; 1535(3): 275-84, 2001 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-11278167

RESUMO

In humans, hepatic iron overload can lead to hepatocellular carcinoma development. Iron related dysregulation of hepatic genes could play a role in this phenomenon. We previously found that the carbonyl-iron overloaded mouse was a useful model to study the mechanisms involved in the development of hepatic lesions related to iron excess. The aim of the present study was to identify hepatic genes overexpressed in conditions of iron overload by using this model. A suppressive subtractive hybridization was performed between hepatic mRNAs extracted from control and 3% carbonyl-iron overloaded mice during 8 months. This methodology allowed us to identify stearoyl coenzyme A desaturase 1 (SCD1) mRNA overexpression in the liver of iron loaded mice. The corresponding enzymatic activity was also found to be significantly increased. In addition, we demonstrated that both SCD1 mRNA expression and activity were increased in another iron overload model in mice obtained by a single iron-dextran subcutaneous injection. Moreover, we found, in both models, that SCD1 mRNA was not only influenced by the quantity of iron in the liver but also by the duration of iron overload since SCD1 mRNA upregulation was not detected in earlier stages of iron overload. In addition, we found that cellular repartition likely influenced SCD1 mRNA expression. In conclusion, we demonstrated that iron excess in the liver induced both the expression of SCD1 mRNA and its corresponding enzymatic activity. The level and duration of iron overload, as well as cellular repartition of iron excess in the liver likely play a role in this induction. The fact that the expression and activity of SCD1, an enzyme adding a double bound into saturated fatty acids, are induced in two models of iron overload in mice leads to the conclusion that iron excess in the liver may enhance the biosynthesis of unsaturated fatty acids.


Assuntos
Sobrecarga de Ferro/metabolismo , Fígado/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Animais , Modelos Animais de Doenças , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Compostos Carbonílicos de Ferro , Complexo Ferro-Dextran , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Compostos Organometálicos , RNA Mensageiro/análise , Estearoil-CoA Dessaturase/biossíntese , Estearoil-CoA Dessaturase/genética , Regulação para Cima
19.
J Biol Chem ; 276(11): 7811-9, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11113132

RESUMO

Considering that the development of hepatic lesions related to iron overload diseases might be a result of abnormally expressed hepatic genes, we searched for new genes up-regulated under the condition of iron excess. By suppressive subtractive hybridization performed between livers from carbonyl iron-overloaded and control mice, we isolated a 225-base pair cDNA. By Northern blot analysis, the corresponding mRNA was confirmed to be overexpressed in livers of experimentally (carbonyl iron and iron-dextran-treated mice) and spontaneously (beta(2)-microglobulin knockout mice) iron-overloaded mice. In addition, beta(2)-microglobulin knockout mice fed with a low iron content diet exhibited a decrease of hepatic mRNA expression. The murine full-length cDNA was isolated and was found to encode an 83-amino acid protein presenting a strong homology in its C-terminal region to the human antimicrobial peptide hepcidin. In addition, we cloned the corresponding rat and human orthologue cDNAs. Both mouse and human genes named HEPC are constituted of 3 exons and 2 introns and are located on chromosome 7 and 19, respectively, in close proximity to USF2 gene. In mouse and human, HEPC mRNA was predominantly expressed in the liver. During both in vivo and in vitro studies, HEPC mRNA expression was enhanced in mouse hepatocytes under the effect of lipopolysaccharide. Finally, to analyze the intracellular localization of the predicted protein, we used the green fluorescent protein chimera expression vectors. The murine green fluorescent protein-prohepcidin protein was exclusively localized in the nucleus. When the putative nuclear localization signal was deleted, the resulting protein was addressed to the cytoplasm. Taken together, our data strongly suggest that the product of the new liver-specific gene HEPC might play a specific role during iron overload and exhibit additional functions distinct from its antimicrobial activity.


Assuntos
Anti-Infecciosos , Peptídeos Catiônicos Antimicrobianos/genética , Regulação da Expressão Gênica , Sobrecarga de Ferro/metabolismo , Fígado/metabolismo , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/isolamento & purificação , Regulação da Expressão Gênica/efeitos dos fármacos , Hepcidinas , Ferro/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Especificidade de Órgãos , Ratos , Ratos Sprague-Dawley
20.
Transfus Sci ; 23(3): 193-200, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11099895

RESUMO

Hemochromatosis is one of the most frequent genetic diseases among the white populations, affecting one in three hundred persons. Its diagnosis has been radically transformed by the discovery of the HFE gene. In a given individual, the diagnosis can, from now on, be ascertained on the sole association of a plasma transferrin saturation (TS) over 45% and homozygosity for the C282Y mutation. Liver biopsy is only required to search for cirrhosis whenever there is hepatomegaly and/or serum ferritin >1000 ng/ml and/or elevated serum AST. Family screening is mandatory, primarily centered on the siblings. The treatment remains based on venesection therapy which improves many features of the disease (one of the most refractory, however, being the joint signs) and permits normal life expectancy provided the diagnosis is established prior to the development of cirrhosis or of insulin-dependent diabetes. In view of the prevalence, the non-invasive diagnosis, the spontaneous severity and the efficacy of a very simple therapy, hemochromatosis should benefit from population screening. This screening could be based, first, on the assessment of transferrin saturation, followed - when elevated - by the search for the C282Y mutation. The discovery of the HFE gene has also paved the road for the individualization of other types of iron overload syndromes which are not HFE-related.


Assuntos
Hemocromatose , Proteínas de Membrana , Antígenos HLA/genética , Hemocromatose/genética , Hemocromatose/fisiopatologia , Hemocromatose/terapia , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA