Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Stem Cell ; 28(6): 1057-1073.e7, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33571444

RESUMO

Skeletal aging is a complex process, characterized by a decrease in bone formation, an increase in marrow fat, and stem cell exhaustion. Loss of H3K9me3, a heterochromatin mark, has been proposed to be associated with aging. Here, we report that loss of KDM4B in mesenchymal stromal cells (MSCs) exacerbated skeletal aging and osteoporosis by reducing bone formation and increasing marrow adiposity via increasing H3K9me3. KDM4B epigenetically coordinated ß-catenin/Smad1-mediated transcription by removing repressive H3K9me3. Importantly, KDM4B ablation impaired MSC self-renewal and promoted MSC exhaustion by inducing senescence-associated heterochromatin foci formation, providing a mechanistic explanation for stem cell exhaustion with aging. Moreover, while KDM4B was required for parathyroid hormone-mediated bone anabolism, KDM4B depletion accelerated bone loss and marrow adiposity induced by a high-fat diet. Our results suggest that the epigenetic rejuvenation and reversing bone-fat imbalance might be new strategies for preventing and treating skeletal aging and osteoporosis by activating KDM4B in MSCs.


Assuntos
Células-Tronco Mesenquimais , Medula Óssea , Células da Medula Óssea , Diferenciação Celular , Osteogênese
2.
J Biol Chem ; 295(50): 17169-17186, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33028635

RESUMO

We have observed overexpression of PACS-1, a cytosolic sorting protein in primary cervical tumors. Absence of exonic mutations and overexpression at the RNA level suggested a transcriptional and/or posttranscriptional regulation. University of California Santa Cruz genome browser analysis of PACS-1 micro RNAs (miR), revealed two 8-base target sequences at the 3' terminus for hsa-miR-34a and hsa-miR-449a. Quantitative RT-PCR and Northern blotting studies showed reduced or loss of expression of the two microRNAs in cervical cancer cell lines and primary tumors, indicating dysregulation of these two microRNAs in cervical cancer. Loss of PACS-1 with siRNA or exogenous expression of hsa-miR-34a or hsa-miR-449a in HeLa and SiHa cervical cancer cell lines resulted in DNA damage response, S-phase cell cycle arrest, and reduction in cell growth. Furthermore, the siRNA studies showed that loss of PACS-1 expression was accompanied by increased nuclear γH2AX expression, Lys382-p53 acetylation, and genomic instability. PACS-1 re-expression through LNA-hsa-anti-miR-34a or -449a or through PACS-1 cDNA transfection led to the reversal of DNA damage response and restoration of cell growth. Release of cells post 24-h serum starvation showed PACS-1 nuclear localization at G1-S phase of the cell cycle. Our results therefore indicate that the loss of hsa-miR-34a and hsa-miR-449a expression in cervical cancer leads to overexpression of PACS-1 and suppression of DNA damage response, resulting in the development of chemo-resistant tumors.


Assuntos
Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , MicroRNAs/metabolismo , RNA Neoplásico/metabolismo , Neoplasias do Colo do Útero/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Feminino , Fase G1 , Células HeLa , Humanos , MicroRNAs/genética , RNA Neoplásico/genética , Pontos de Checagem da Fase S do Ciclo Celular , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Proteínas de Transporte Vesicular/genética
3.
Nature ; 576(7786): 281-286, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31776511

RESUMO

Limited knowledge of the mechanisms that govern the self-renewal of human haematopoietic stem cells (HSCs), and why this fails in culture, have impeded the expansion of HSCs for transplantation1. Here we identify MLLT3 (also known as AF9) as a crucial regulator of HSCs that is highly enriched in human fetal, neonatal and adult HSCs, but downregulated in culture. Depletion of MLLT3 prevented the maintenance of transplantable human haematopoietic stem or progenitor cells (HSPCs) in culture, whereas stabilizing MLLT3 expression in culture enabled more than 12-fold expansion of transplantable HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Similar to endogenous MLLT3, overexpressed MLLT3 localized to active promoters in HSPCs, sustained levels of H3K79me2 and protected the HSC transcriptional program in culture. MLLT3 thus acts as HSC maintenance factor that links histone reader and modifying activities to modulate HSC gene expression, and may provide a promising approach to expand HSCs for transplantation.


Assuntos
Autorrenovação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Proteínas Nucleares/genética , Ligação Proteica
4.
Mol Cell ; 73(2): 250-263.e5, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30527662

RESUMO

Metazoan chromosomes are sequentially partitioned into topologically associating domains (TADs) and then into smaller sub-domains. One class of sub-domains, insulated neighborhoods, are proposed to spatially sequester and insulate the enclosed genes through self-association and chromatin looping. However, it has not been determined functionally whether promoter-enhancer interactions and gene regulation are broadly restricted to within these loops. Here, we employed published datasets from murine embryonic stem cells (mESCs) to identify insulated neighborhoods that confine promoter-enhancer interactions and demarcate gene regulatory regions. To directly address the functionality of these regions, we depleted estrogen-related receptor ß (Esrrb), which binds the Mediator co-activator complex, to impair enhancers of genes within 222 insulated neighborhoods without causing mESC differentiation. Esrrb depletion reduces Mediator binding, promoter-enhancer looping, and expression of both nascent RNA and mRNA within the insulated neighborhoods without significantly affecting the flanking genes. Our data indicate that insulated neighborhoods represent functional regulons in mammalian genomes.


Assuntos
Cromossomos de Mamíferos , Elementos Facilitadores Genéticos , Elementos Isolantes , Células-Tronco Embrionárias Murinas/fisiologia , Regiões Promotoras Genéticas , Transcrição Gênica , Animais , Sítios de Ligação , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Bases de Dados Genéticas , Regulação para Baixo , Camundongos , Ligação Proteica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Coesinas
5.
Cell Rep ; 22(5): 1211-1224, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29386109

RESUMO

Given its role as the source of definitive hematopoietic cells, we sought to determine whether mutations initiated in the hemogenic endothelium would yield hematopoietic abnormalities or malignancies. Here, we find that endothelium-specific transposon mutagenesis in mice promotes hematopoietic pathologies that are both myeloid and lymphoid in nature. Frequently mutated genes included previously recognized cancer drivers and additional candidates, such as Pi4ka, a lipid kinase whose mutation was found to promote myeloid and erythroid dysfunction. Subsequent validation experiments showed that targeted inactivation of the Pi4ka catalytic domain or reduction in mRNA expression inhibited myeloid and erythroid cell differentiation in vitro and promoted anemia in vivo through a mechanism involving deregulation of AKT, MAPK, SRC, and JAK-STAT signaling. Finally, we provide evidence linking PI4KAP2, previously considered a pseudogene, to human myeloid and erythroid leukemia.


Assuntos
Eritropoese/fisiologia , Leucemia/genética , Antígenos de Histocompatibilidade Menor/genética , Mielopoese/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Diferenciação Celular/genética , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor/metabolismo , Mutagênese , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Peixe-Zebra
6.
Mol Cell ; 67(4): 594-607.e4, 2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28735899

RESUMO

Pervasive transcription initiates from cryptic promoters and is observed in eukaryotes ranging from yeast to mammals. The Set2-Rpd3 regulatory system prevents cryptic promoter function within expressed genes. However, conserved systems that control pervasive transcription within intergenic regions have not been well established. Here we show that Mot1, Ino80 chromatin remodeling complex (Ino80C), and NC2 co-localize on chromatin and coordinately suppress pervasive transcription in S. cerevisiae and murine embryonic stem cells (mESCs). In yeast, all three proteins bind subtelomeric heterochromatin through a Sir3-stimulated mechanism and to euchromatin via a TBP-stimulated mechanism. In mESCs, the proteins bind to active and poised TBP-bound promoters along with promoters of polycomb-silenced genes apparently lacking TBP. Depletion of Mot1, Ino80C, or NC2 by anchor away in yeast or RNAi in mESCs leads to near-identical transcriptome phenotypes, with new subtelomeric transcription in yeast, and greatly increased pervasive transcription in both yeast and mESCs.


Assuntos
Adenosina Trifosfatases/metabolismo , Células-Tronco Embrionárias/enzimologia , Fosfoproteínas/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA , Eucromatina/genética , Eucromatina/metabolismo , Regulação Fúngica da Expressão Gênica , Inativação Gênica , Genótipo , Heterocromatina/genética , Heterocromatina/metabolismo , Fenótipo , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Proteínas Repressoras/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Fatores Associados à Proteína de Ligação a TATA/genética , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Fator de Transcrição TFIID , Fatores de Transcrição/genética , Transfecção
7.
Head Neck ; 38(1): 118-25, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25223295

RESUMO

BACKGROUND: The purpose of our study was to identify serum protein biomarkers for node-positive oral squamous cell carcinoma (OSCC). Biomarkers indicating lymph node metastasis provides a valuable classification methodology to optimize treatment plans for patients with OSCC. METHODS: Quantitative serum proteomic analysis of OSCCs with either node-positive or node-negative disease was performed with tandem mass spectrometry and isobaric tagging for relative and absolute quantitation (iTRAQ). Immunoassays were used to validate a panel of candidate protein biomarkers and receiver operating characteristic (ROC) analysis was used to evaluate the performance of the candidate biomarkers. RESULTS: A total of 282 serum proteins were quantified between node-positive and node-negative OSCCs with the proteomic approach. Four candidate biomarkers, gelsolin, fibronectin, angiotensinogen, and haptoglobin, were validated in an independent group of patients with node-positive or node-negative OSCC. The best candidate biomarker, gelsolin, yielded a ROC value of 89% for node-positive OSCC, although the sample size for validation is relatively small. Fibronectin, gelsolin, and angiotensinogen were also found to be differentially expressed between cancer cell lines of node-positive and node-negative cancer origin. CONCLUSION: Our studies suggest that testing of serum protein biomarkers might help detect lymph node metastasis of oral cancer. Because of limited sample size in our studies, long-term longitudinal studies with large populations of individuals with oral cancer are needed to validate these potential biomarkers.


Assuntos
Angiotensinogênio/sangue , Biomarcadores Tumorais/sangue , Carcinoma de Células Escamosas/genética , Fibronectinas/sangue , Gelsolina/sangue , Haptoglobinas/metabolismo , Neoplasias Bucais/genética , Idoso , Carcinoma de Células Escamosas/sangue , Carcinoma de Células Escamosas/diagnóstico , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/sangue , Neoplasias Bucais/diagnóstico , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Inibidores de Serina Proteinase/sangue
8.
Mol Cell ; 61(1): 27-38, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26669263

RESUMO

Gene activation in metazoans is accompanied by the presence of histone variants H2AZ and H3.3 within promoters and enhancers. It is not known, however, what protein deposits H3.3 into chromatin or whether variant chromatin plays a direct role in gene activation. Here we show that chromatin containing acetylated H2AZ and H3.3 stimulates transcription in vitro. Analysis of the Pol II pre-initiation complex on immobilized chromatin templates revealed that the E1A binding protein p400 (EP400) was bound preferentially to and required for transcription stimulation by acetylated double-variant chromatin. EP400 also stimulated H2AZ/H3.3 deposition into promoters and enhancers and influenced transcription in vivo at a step downstream of the Mediator complex. EP400 efficiently exchanged recombinant histones H2A and H3.1 with H2AZ and H3.3, respectively, in a chromatin- and ATP-stimulated manner in vitro. Our data reveal that EP400 deposits H3.3 into chromatin alongside H2AZ and contributes to gene regulation after PIC assembly.


Assuntos
Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Histonas/metabolismo , Regiões Promotoras Genéticas , Ativação Transcricional , Acetilação , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Genes Reporter , Histonas/genética , Humanos , Interferência de RNA , RNA Polimerase II/metabolismo , Fatores de Tempo , Transfecção
9.
Cell Host Microbe ; 16(5): 663-76, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25525796

RESUMO

Oncogenic transformation by adenovirus small e1a depends on simultaneous interactions with the host lysine acetylases p300/CBP and the tumor suppressor RB. How these interactions influence cellular gene expression remains unclear. We find that e1a displaces RBs from E2F transcription factors and promotes p300 acetylation of RB1 K873/K874 to lock it into a repressing conformation that interacts with repressive chromatin-modifying enzymes. These repressing p300-e1a-RB1 complexes specifically interact with host genes that have unusually high p300 association within the gene body. The TGF-β, TNF-, and interleukin-signaling pathway components are enriched among such p300-targeted genes. The p300-e1a-RB1 complex condenses chromatin in a manner dependent on HDAC activity, p300 lysine acetylase activity, the p300 bromodomain, and RB K873/K874 and e1a K239 acetylation to repress host genes that would otherwise inhibit productive virus infection. Thus, adenovirus employs e1a to repress host genes that interfere with viral replication.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/metabolismo , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/genética , Transformação Celular Viral , Células Cultivadas , Quimiocina CXCL1/metabolismo , Cromatina/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Replicação Viral
10.
Epigenetics ; 9(2): 257-67, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24172870

RESUMO

The cellular epigenetic landscape changes as pluripotent stem cells differentiate to somatic cells or when differentiated cells transform to a cancerous state. These epigenetic changes are commonly correlated with differences in gene expression. Whether active DNA replication is also associated with distinct chromatin environments in these developmentally and phenotypically diverse cell types has not been known. Here, we used BrdU-seq to map active DNA replication loci in human embryonic stem cells (hESCs), normal primary fibroblasts and a cancer cell line, and correlated these maps to the epigenome. In all cell lines, the majority of BrdU peaks were enriched in euchromatin and at DNA repetitive elements, especially at microsatellite repeats, and coincided with previously determined replication origins. The most prominent BrdU peaks were shared between all cells but a sizable fraction of the peaks were specific to each cell type and associated with cell type-specific genes. Surprisingly, the BrdU peaks that were common to all cell lines were associated with H3K18ac, H3K56ac, and H4K20me1 histone marks only in hESCs but not in normal fibroblasts or cancer cells. Depletion of the histone acetyltransferases for H3K18 and H3K56 dramatically decreased the number and intensity of BrdU peaks in hESCs. Our data reveal a unique epigenetic signature that distinguishes active replication loci in hESCs from normal somatic or malignant cells.


Assuntos
Replicação do DNA , DNA/genética , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Loci Gênicos , Células-Tronco Pluripotentes/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , DNA/metabolismo , Fibroblastos/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Humanos , Origem de Replicação
11.
Mol Cell ; 49(2): 310-21, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23201122

RESUMO

Differences in global levels of histone acetylation occur in normal and cancer cells, although the reason why cells regulate these levels has been unclear. Here we demonstrate a role for histone acetylation in regulating intracellular pH (pH(i)). As pH(i) decreases, histones are globally deacetylated by histone deacetylases (HDACs), and the released acetate anions are coexported with protons out of the cell by monocarboxylate transporters (MCTs), preventing further reductions in pH(i). Conversely, global histone acetylation increases as pH(i) rises, such as when resting cells are induced to proliferate. Inhibition of HDACs or MCTs decreases acetate export and lowers pH(i), particularly compromising pH(i) maintenance in acidic environments. Global deacetylation at low pH is reflected at a genomic level by decreased abundance and extensive redistribution of acetylation throughout the genome. Thus, acetylation of chromatin functions as a rheostat to regulate pH(i) with important implications for mechanism of action and therapeutic use of HDAC inhibitors.


Assuntos
Histonas/metabolismo , Líquido Intracelular/metabolismo , Processamento de Proteína Pós-Traducional , Acetatos , Acetilação , Metabolismo dos Carboidratos , Cromatina , Regulação da Expressão Gênica , Glucose/fisiologia , Glutamina/fisiologia , Células HeLa , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Histonas/genética , Humanos , Concentração de Íons de Hidrogênio , Ácidos Hidroxâmicos/farmacologia , Transportadores de Ácidos Monocarboxílicos/metabolismo , Niacinamida/farmacologia , Ácido Pirúvico/metabolismo , Análise de Sequência de RNA , Transcriptoma
12.
Genome Res ; 22(7): 1212-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22499665

RESUMO

Adenovirus small e1a oncoprotein causes ~70% reduction in cellular levels of histone H3 lysine 18 acetylation (H3K18ac). It is unclear, however, where this dramatic reduction occurs genome-wide. ChIP-sequencing revealed that by 24 h after expression, e1a erases 95% of H3K18ac peaks in normal, contact-inhibited fibroblasts and replaces them with one-third as many at new genomic locations. The H3K18ac peaks at promoters and intergenic regions of genes with fibroblast-related functions are eliminated after infection, and new H3K18ac peaks are established at promoters of highly induced genes that regulate cell cycling and at new putative enhancers. Strikingly, the regions bound by the retinoblastoma family of proteins in contact-inhibited fibroblasts gain new peaks of H3K18ac in the e1a-expressing cells, including 55% of RB1-bound loci. In contrast, over half of H3K9ac peaks are similarly distributed before and after infection, independently of RB1. The strategic redistribution of H3K18ac by e1a highlights the importance of this modification for transcriptional activation and cellular transformation as well as functional differences between the RB-family member proteins.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/genética , Epigênese Genética , Genoma Humano , Histonas/metabolismo , Acetilação , Proteínas E1A de Adenovirus/genética , Adenovírus Humanos/metabolismo , Adenovírus Humanos/patogenicidade , Ciclo Celular , Transformação Celular Viral , Células Cultivadas , Imunoprecipitação da Cromatina , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Histonas/genética , Humanos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Anotação de Sequência Molecular/métodos , Nucleossomos/genética , Nucleossomos/metabolismo , Regiões Promotoras Genéticas , Fatores de Tempo , Ativação Transcricional
13.
Biometals ; 22(5): 827-34, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19330300

RESUMO

Disorders of iron metabolism are a significant problem primarily in young and old populations. In this study, We compared 1-year-old C57BL6/J mice on iron deficient, iron overload, or iron sufficient diets with two similarly aged genetic models of disturbed iron homeostasis, the sla (sex-linked anemia), and the ceruloplasmin knockout mice (Cp(-/-)) on iron sufficient diet. We found tissue specific changes in sla and nutritional iron deficiency including decreased liver Hamp1 expression and increased protein expression of the enterocyte basolateral iron transport components, hephaestin and ferroportin. In contrast, the Cp(-/-) mice did not show significantly increased Hamp1 expression despite increased liver iron suggesting that regulation is independent of liver iron levels. Together, these results suggest that older mice have a distinct response to alterations in iron metabolism and that age must be considered in future studies of iron metabolism.


Assuntos
Envelhecimento/fisiologia , Ceruloplasmina/genética , Homeostase , Ferro/metabolismo , Mutação/genética , Anemia Ferropriva/genética , Anemia Ferropriva/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Northern Blotting , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Hepcidinas , Homeostase/efeitos dos fármacos , Immunoblotting , Técnicas In Vitro , Sobrecarga de Ferro/metabolismo , Ferro da Dieta/farmacologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
J Nutr ; 136(5): 1236-41, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16614410

RESUMO

Copper and iron metabolism intersect in mammals. Copper deficiency simultaneously leads to decreased iron levels in some tissues and iron deficiency anemia, whereas it results in iron overload in other tissues such as the intestine and liver. The copper requirement of the multicopper ferroxidases hephaestin and ceruloplasmin likely explains this link between copper and iron homeostasis in mammals. We investigated the effect of in vivo and in vitro copper deficiency on hephaestin (Heph) expression and activity. C57BL/6J mice were separated into 2 groups on the day of parturition. One group was fed a copper-deficient diet and another was fed a control diet for 6 wk. Copper-deficient mice had significantly lower hephaestin and ceruloplasmin (approximately 50% of controls) ferroxidase activity. Liver hepcidin expression was significantly downregulated by copper deficiency (approximately 60% of controls), and enterocyte mRNA and protein levels of ferroportin1 were increased to 2.5 and 10 times, respectively, relative to controls, by copper deficiency, indicating a systemic iron deficiency in the copper-deficient mice. Interestingly, hephaestin protein levels were significantly decreased to approximately 40% of control, suggesting that decreased enterocyte copper content leads to decreased hephaestin synthesis and/or stability. We also examined the effect of copper deficiency on hephaestin in vitro in the HT29 cell line and found dramatically decreased hephaestin synthesis and activity. Both in vivo and in vitro studies indicate that copper is required for the proper processing and/or stability of hephaestin.


Assuntos
Anemia Ferropriva/etiologia , Cobre/deficiência , Proteínas de Membrana/deficiência , Animais , Linhagem Celular Tumoral , Neoplasias do Colo , Feminino , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Valores de Referência , Superóxido Dismutase/metabolismo
15.
Blood ; 102(5): 1893-9, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12730111

RESUMO

Hephaestin is a membrane-bound multicopper ferroxidase necessary for iron egress from intestinal enterocytes into the circulation. Mice with sex-linked anemia (sla) have a mutant form of Hephaestin and a defect in intestinal basolateral iron transport, which results in iron deficiency and anemia. Ireg1 (SLC11A3, also known as Ferroportin1 or Mtp1) is the putative intestinal basolateral iron transporter. We compared iron levels and expression of genes involved in iron uptake and storage in sla mice and C57BL/6J mice fed iron-deficient, iron-overload, or control diets. Both iron-deficient wild-type mice and sla mice showed increased expression of Heph and Ireg1 mRNA, compared to controls, whereas only iron-deficient wild-type mice had increased expression of the brush border transporter Dmt1. Unlike iron-deficient mice, sla mouse enterocytes accumulated nonheme iron and ferritin. These results indicate that Dmt1 can be modulated by the enterocyte iron level, whereas Hephaestin and Ireg1 expression respond to systemic rather than local signals of iron status. Thus, the basolateral transport step appears to be the primary site at which the small intestine responds to alterations in body iron requirements.


Assuntos
Anemia Ferropriva/genética , Anemia Ferropriva/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Especificidade de Anticorpos , Dieta , Enterócitos/metabolismo , Ferritinas/sangue , Expressão Gênica , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Ferro da Dieta/sangue , Ferro da Dieta/farmacocinética , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Masculino , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fenômenos Fisiológicos da Nutrição , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA