Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 421
Filtrar
1.
FEBS Open Bio ; 11(11): 3101-3114, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34551213

RESUMO

Overexpression of ferritin heavy chain (FTH1) often associates with good prognosis in breast cancer (BCa), particularly in the triple-negative subtype (triple-negative breast cancer). However, the mechanism by which FTH1 exerts its possible tumor suppressor effects in BCa is not known. Here, we examined the bearing of FTH1 silencing or overexpression on several aspects of BCa cell growth in vitro. FTH1 silencing promoted cell growth and mammosphere formation, increased c-MYC expression, and reduced cell sensitivity to chemotherapy. In contrast, FTH1 overexpression inhibited cell growth, decreased c-MYC expression, and sensitized cancer cells to chemotherapy; silencing of c-MYC recapitulated the effects of FTH1 overexpression. These findings show for the first time that FTH1 suppresses tumor growth by inhibiting the expression of key oncogenes, such as c-MYC.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ferritinas/metabolismo , Oxirredutases/metabolismo , Apoferritinas/biossíntese , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Ferritinas/biossíntese , Ferritinas/genética , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Oxirredutases/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo
2.
Int J Mol Sci ; 22(3)2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540888

RESUMO

Macrophages are essential immune cells of the innate immune system. They participate in the development and regulation of inflammation. Macrophages play a fundamental role in fighting against bacterial infections by phagocytosis of bacteria, and they also have a specific role in immunomodulation by secreting pro-inflammatory cytokines. In bacterial infection, macrophages decrease the serum iron concentration by removing iron from the blood, acting as one of the most important regulatory cells of iron homeostasis. We examined whether the Gram-positive and Gram-negative cell wall components from various bacterial strains affect the cytokine production and iron transport, storage and utilization of THP-1 monocytes in different ways. We found that S. aureus lipoteichoic acid (LTA) was less effective in activating pro-inflammatory cytokine expression that may related to its effect on fractalkine production. LTA-treated cells increased iron uptake through divalent metal transporter-1, but did not elevate the expression of cytosolic and mitochondrial iron storage proteins, suggesting that the cells maintained iron efflux via the ferroportin iron exporter. E. coli and P. aeruginosa lipopolysaccharides (LPSs) acted similarly on THP-1 cells, but the rates of the alterations of the examined proteins were different. E. coli LPS was more effective in increasing the pro-inflammatory cytokine production, meanwhile it caused less dramatic alterations in iron metabolism. P. aeruginosa LPS-treated cells produced a smaller amount of pro-inflammatory cytokines, but caused remarkable elevation of both cytosolic and mitochondrial iron storage proteins and intracellular iron content compared to E. coli LPS. These results prove that LPS molecules from different bacterial sources alter diverse molecular mechanisms in macrophages that prepossess the outcome of the bacterial infection.


Assuntos
Parede Celular/química , Citocinas/metabolismo , Escherichia coli/química , Ferro/metabolismo , Lipopolissacarídeos/farmacologia , Pseudomonas aeruginosa/química , Staphylococcus aureus/química , Células THP-1/metabolismo , Ácidos Teicoicos/farmacologia , Transporte Biológico , Receptor 1 de Quimiocina CX3C/biossíntese , Receptor 1 de Quimiocina CX3C/genética , Quimiocina CX3CL1/metabolismo , Citocinas/biossíntese , Citosol/metabolismo , Ferritinas/biossíntese , Ferritinas/genética , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Hepcidinas/biossíntese , Hepcidinas/genética , Humanos , Mitocôndrias/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Oxirredutases/biossíntese , Oxirredutases/genética , RNA Mensageiro/biossíntese , RNA Neoplásico/genética , Células THP-1/efeitos dos fármacos
3.
J Biochem Mol Toxicol ; 35(4): e22704, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33393188

RESUMO

In the present study, we demonstrate the coaction of thioredoxin and glutathione (GSH) systems in mouse liver against iron overload-induced oxidative stress (OS). Mice were injected intraperitoneally with an iron dextran solution twice a week for 3 weeks. Iron accumulation in mouse liver was demonstrated spectroscopically. To confirm the iron overload model in the liver, the increased gene expression levels of hepcidin (Hamp), ferroportin (Fpn1), and ferritin (Fth1), which regulate iron trafficking, were observed by a quantitative polymerase chain reaction. In the case of iron overload, the GSH level and the reduced glutathione/oxidized glutathione ratio, which represents a marker of OS, decreased significantly. An increase in the malondialdehyde level, one of the final products of the lipid peroxidation process, was observed. The gene expression of the thioredoxin system, including thioredoxin (Trx1) and thioredoxin reductase (TrxR1), was examined. Though TrxR1 expression decreased, no changes were observed in Trx1. The enzyme activity and semiquantitative protein expression of TRXR1 increased. The activity of GSH reductase and GSH peroxidase increased in the iron overload group. The gene and protein expressions of thioredoxininteracting protein, which is an indicator of the commitment of the cell to apoptosis, were elevated significantly. The increased protein expression of Bcl-2-related X protein and CASPASE-3, which is an indicator of apoptosis, increased significantly. In conclusion, excess iron accumulation in mouse liver tissue causes OS, which affects the redox state of the thioredoxin and GSH systems, inducing cell apoptosis and also ferroptosis due to increased lipid peroxidation and the depletion of GSH level.


Assuntos
Glutationa/metabolismo , Sobrecarga de Ferro/metabolismo , Fígado/metabolismo , Estresse Oxidativo , Tiorredoxinas/biossíntese , Animais , Proteínas de Transporte de Cátions/biossíntese , Ferritinas/biossíntese , Regulação da Expressão Gênica , Hepcidinas/biossíntese , Sobrecarga de Ferro/patologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oxirredutases/biossíntese
4.
Neurotherapeutics ; 17(4): 1796-1812, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32959272

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by degeneration of dopaminergic neurons associated with dysregulation of iron homeostasis in the brain. Ferroptosis is an iron-dependent cell death process that serves as a significant regulatory mechanism in PD. However, its underlying mechanisms are not yet fully understood. By performing RNA sequencing analysis, we found that the main iron storage protein ferritin heavy chain 1 (FTH1) is differentially expressed in the rat 6-hydroyxdopamine (6-OHDA) model of PD compared with control rats. Our present work demonstrates that FTH1 is involved in iron accumulation and the ferroptosis pathway in this model. Knockdown of FTH1 in PC-12 cells significantly inhibited cell viability and caused mitochondrial dysfunction. Moreover, FTH1 was found to be involved in ferritinophagy, a selective form of autophagy involving the degradation of ferritin by ferroptosis. Overexpression of FTH1 in PC-12 cells impaired ferritinophagy and downregulated microtubule-associated protein light chain 3 and nuclear receptor coactivator 4 expression, ultimately suppressing cell death induced by ferroptosis. Consistent with these findings, the ferritinophagy inhibitors chloroquine and bafilomycin A1 inhibited ferritin degradation and ferroptosis in 6-OHDA-treated PC-12 cells. This entire process was mediated by the cyclic regulation of FTH1 and ferritinophagy. Taken together, these results suggest that FTH1 links ferritinophagy and ferroptosis in the 6-OHDA model of PD, and provide a new perspective and potential for a pharmacological target in this disease.


Assuntos
Ferritinas/biossíntese , Ferroptose/fisiologia , Oxidopamina/toxicidade , Oxirredutases/biossíntese , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Ferritinas/genética , Ferroptose/efeitos dos fármacos , Masculino , Oxirredutases/genética , Células PC12 , Transtornos Parkinsonianos/genética , Ratos , Ratos Sprague-Dawley
5.
Am J Physiol Renal Physiol ; 317(6): F1563-F1571, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31608670

RESUMO

Experimental data suggest that iron sucrose (FeS) injection, used either alone or in combination with other prooxidants, can induce "renal preconditioning," in part by upregulating cytoprotective ferritin levels. However, the rapidity, degree, composition (heavy vs. light chain), and renal ferritin changes after FeS administration in humans remain to be defined. To address these issues, healthy human volunteers (n = 9) and patients with stage 3-4 chronic kidney disease(n = 9) were injected once with FeS (120, 240, or 360 mg). Plasma ferritin was measured from 0 to 8 days postinjection as an overall index of ferritin generation. Urinary ferritin served as a "biomarker" of renal ferritin production. FeS induced rapid (≤2 h), dose-dependent, plasma ferritin increases in all study participants, peaking at approximately three to five times baseline within 24-48 h. Significant urinary ferritin increases (~3 times), without dose-dependent increases in albuminuria, neutrophil gelatinase-associated lipocalin, or N-acetyl-ß-d-glucosaminidase excretion, were observed. Western blot analysis with ferritin heavy chain (Fhc)- and light chain (Flc)-specific antibodies demonstrated that FeS raised plasma Flc but not Fhc levels. Conversely, FeS increased both Fhc and Flc in urine. To assess sites of FeS-induced ferritin generation, organs from FeS-treated mice were probed for Fhc, Flc, and their mRNAs. FeS predominantly raised hepatic Flc. Conversely, marked Fhc and Flc elevations developed in the kidney and spleen. No cardiopulmonary ferritin increases occurred. Ferritin mRNAs remained unchanged throughout, implying posttranscriptional ferritin production. We conclude that FeS induces rapid, dramatic, and differential Fhc and Flc upregulation in organs. Renal Fhc and Flc increases, in the absence of nephrotoxicity, suggest potential FeS utility as a clinical renal "preconditioning" agent.


Assuntos
Óxido de Ferro Sacarado/farmacologia , Ferritinas/biossíntese , Precondicionamento Isquêmico , Rim/efeitos dos fármacos , Injúria Renal Aguda/prevenção & controle , Adulto , Idoso , Animais , Biomarcadores/urina , Feminino , Óxido de Ferro Sacarado/administração & dosagem , Óxido de Ferro Sacarado/efeitos adversos , Ferritinas/sangue , Ferritinas/urina , Voluntários Saudáveis , Humanos , Infusões Parenterais , Rim/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , Insuficiência Renal Crônica/metabolismo , Baço/metabolismo
6.
Environ Pollut ; 254(Pt A): 112937, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31401526

RESUMO

PM2.5 is becoming a worldwide environmental problem, which profoundly endangers public health, thus progressively capturing public attention this decade. As a fragile target of PM2.5, the underlying mechanisms of endothelial cell damage are still obscure. According to the previous microarray data and signaling pathway analysis, a new form of cell death termed ferroptosis in the current study is proposed following PM2.5 exposure. In order to verify the vital role of ferroptosis in PM2.5-induced endothelial lesion and further understand the potential mechanism involved, intracellular iron content, ROS release and lipid peroxidation, as well as biomarkers of ferroptosis were detected, respectively. As a result, uptake of particles increases cellular iron content and ROS production. Meanwhile, GSH depletion, and the decrease of GSH-Px and NADPH play significant roles in PM2.5-induced endothelial cell ferroptosis. Moreover, significantly changed expression of TFRC, FTL and FTH1 hinted that dysfunction of iron uptake and storage is a major inducer of ferroptosis. Importantly, index monitored above can be partially rescued by lipid peroxidation inhibitor ferrostatin-1 and iron chelator deferoxamine mesylate, which mediated antiferroptosis activity mainly depends on the restoration of antioxidant activity and iron metabolism. In conclusion, our data basically show that PM2.5 enhances ferroptosis sensitivity with increased ferroptotic events in endothelial cells, in which iron overload, lipid peroxidation and redox imbalance act pivotal roles.


Assuntos
Células Endoteliais/metabolismo , Ferroptose/fisiologia , Sobrecarga de Ferro/patologia , Ferro/toxicidade , Material Particulado/toxicidade , Antígenos CD/biossíntese , Apoferritinas/biossíntese , Apoptose/efeitos dos fármacos , Cicloexilaminas/farmacologia , Desferroxamina/farmacologia , Ferritinas/biossíntese , Glutationa/metabolismo , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Oxirredutases , Fenilenodiaminas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores da Transferrina/biossíntese , Transdução de Sinais/efeitos dos fármacos
7.
Med Sci Monit ; 25: 3700-3715, 2019 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-31104064

RESUMO

BACKGROUND Serum ferritin is a useful tumor marker for renal cell carcinoma (RCC). However, the expression of ferritin heavy chain (FTH1), the main subunit of ferritin, is unclear in primary RCC tissues. In this study, we investigated FTH1 mRNA expression and its diagnostic and prognostic value in RCC. MATERIAL AND METHODS The mRNA expression of FTH1 was analyzed using including Oncomine, Gene Expression Omnibus, and Cancer Genome Atlas datasets, while the protein level of FTH1 was analyzed using the Human Protein Atlas database. The associations between FTH1 and clinicopathologic characteristics and survival time and Cox multivariate survival analysis were analyzed using SPSS 22.0 software. A meta-analysis was performed to assess consistency of FTH1 expression. GO, KEGG, and PPI analyses were used to predict biological functions. RESULTS According to TCGA data, overexpression of FTH1 was detected in 890 RCC tissues (15.2904±0.63157) compared to 129 normal kidney tissues (14.4502±0.51523, p<0.001). Among the clinicopathological characteristics evaluated, patients with increased pathologic T staging, lymph node metastasis, and distant metastasis were significantly associated with higher expression of FTH1. Elevated FTH1 mRNA levels were correlated with worse prognosis of RCC patients. Cox multivariate survival analysis indicated that age, stage, and M stage were predictors of poor prognosis in patients with RCC. CONCLUSIONS Our data suggest that FTH1 expression is an effective prognostic and diagnosis biomarker for RCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Ferritinas/biossíntese , Neoplasias Renais/metabolismo , Adulto , Idoso , Apoferritinas/genética , Apoferritinas/metabolismo , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Feminino , Ferritinas/genética , Ferritinas/metabolismo , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Oxirredutases , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma
8.
Stem Cell Res Ther ; 10(1): 85, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30867050

RESUMO

BACKGROUND: Islets of Langerhans transplantation is a promising therapy for type 1 diabetes mellitus, but this technique is compromised by transplantation stresses including inflammation. In other tissues, co-transplantation with mesenchymal stem cells has been shown to reduce damage by improving anti-inflammatory and anti-oxidant defences. Therefore, we probed the protection afforded by bone marrow mesenchymal stem cells to islets under pro-inflammatory cytokine stress. METHODS: In order to evaluate the cytoprotective potential of mesenchymal stem cells on rat islets, co-cultures were exposed to the interleukin-1, tumour necrosis factor α and interferon γ cocktail for 24 h. Islet viability and functionality tests were performed. Reactive oxygen species and malondialdehyde were measured. Expression of stress-inducible genes acting as anti-oxidants and detoxifiers, such as superoxide dismutases 1 and 2, NAD(P)H quinone oxidoreductase 1, heme oxygenase-1 and ferritin H, was compared to non-stressed cells, and the corresponding proteins were measured. Data were analysed by a two-way ANOVA followed by a Holm-Sidak post hoc analysis. RESULTS: Exposure of rat islets to cytokines induces a reduction in islet viability and functionality concomitant with an oxidative status shift with an increase of cytosolic ROS production. Mesenchymal stem cells did not significantly increase rat islet viability under exposure to cytokines but protected islets from the loss of insulin secretion. A drastic reduction of the antioxidant factors heme oxygenase-1 and ferritin H protein levels was observed in islets exposed to the cytokine cocktail with a prevention of this effect by the presence of mesenchymal stem cells. CONCLUSIONS: Our data evidenced that MSCs are able to preserve islet insulin secretion through a modulation of the oxidative imbalance mediated by heme and iron via heme oxygenase-1 and ferritin in a context of cytokine exposure.


Assuntos
Citocinas/farmacologia , Ferritinas/biossíntese , Heme Oxigenase (Desciclizante)/biossíntese , Ilhotas Pancreáticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Animais , Técnicas de Cocultura , Humanos , Ilhotas Pancreáticas/citologia , Células-Tronco Mesenquimais/citologia , Ratos
9.
Am J Med Sci ; 355(2): 183-190, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29406047

RESUMO

BACKGROUND: Although excess iron induces oxidative stress in the liver, it is unclear whether it directly activates the hepatic stellate cells (HSC). MATERIALS AND METHODS: We evaluated the effects of excess iron on fibrogenesis and transforming growth factor beta (TGF-ß) signaling in murine HSC. Cells were treated with holotransferrin (0.005-5g/L) for 24 hours, with or without the iron chelator deferoxamine (10µM). Gene expressions (α-SMA, Col1-α1, Serpine-1, TGF-ß, Hif1-α, Tfrc and Slc40a1) were analyzed by quantitative real time-polymerase chain reaction, whereas TfR1, ferroportin, ferritin, vimentin, collagen, TGF-ß RII and phospho-Smad2 proteins were evaluated by immunofluorescence, Western blot and enzyme-linked immunosorbent assay. RESULTS: HSC expressed the iron-uptake protein transferrin receptor 1 (TfR1) and the iron-export protein ferroportin. Holotransferrin upregulated TfR1 expression by 1.8-fold (P < 0.03) and ferritin accumulation (iron storage) by 2-fold (P < 0.01), and activated HSC with 2-fold elevations (P < 0.03) in α-SMA messenger RNA and collagen secretion, and a 1.6-fold increase (P < 0.01) in vimentin protein. Moreover, holotransferrin activated the TGF-ß pathway with TGF-ß messenger RNA elevated 1.6-fold (P = 0.05), and protein levels of TGF-ß RII and phospho-Smad2 increased by 1.8-fold (P < 0.01) and 1.6-fold (P < 0.01), respectively. In contrast, iron chelation decreased ferritin levels by 30% (P < 0.03), inhibited collagen secretion by 60% (P < 0.01), repressed fibrogenic genes α-SMA (0.2-fold; P < 0.05) and TGF-ß (0.4-fold; P < 0.01) and reduced levels of TGF-ß RII and phospho-Smad2 proteins. CONCLUSIONS: HSC express iron-transport proteins. Holotransferrin (iron) activates HSC fibrogenesis and the TGF-ß pathway, whereas iron depletion by chelation reverses this, suggesting that this could be a useful adjunct therapy for patients with fibrosis. Further studies in primary human HSC and animal models are necessary to confirm this.


Assuntos
Regulação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Ferro/metabolismo , Cirrose Hepática/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Ferritinas/biossíntese , Células Estreladas do Fígado/patologia , Cirrose Hepática/patologia , Camundongos , Proteínas Serina-Treonina Quinases/biossíntese , RNA Mensageiro/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores da Transferrina/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Proteína Smad2/metabolismo
10.
Eur J Nutr ; 57(2): 669-678, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27942845

RESUMO

PURPOSE: Iron is essential for development and growth in young children; unfortunately, iron deficiency (ID) is a significant public health problem in this population. Young Child Formulae (YCF), milk-derived products fortified with iron and ascorbic acid (AA, an enhancer of iron absorption) may be good sources of iron to help prevent ID. Furthermore, some YCF are supplemented with prebiotics, non-digestible carbohydrates suggested to enhance iron bioavailability. The aim of our study was to evaluate iron bioavailability of YCF relative to prebiotic and AA concentrations. We hypothesised that YCF with the highest levels of prebiotics and AA would have the most bioavailable iron. METHODS: We used the in vitro digestion/Caco-2 cell model to measure iron bioavailability from 4 commercially available YCF with approximately equal amounts of iron, but varying amounts of: AA and the prebiotics fructo- and galacto-oligosaccharides. Caco-2 cell ferritin formation was used as a surrogate marker for iron bioavailability. RESULTS: The YCF with the highest concentration of prebiotics and AA had the highest iron bioavailability; conversely, the YCF with the lowest concentration of prebiotics and AA had the lowest. After the addition of exogenous prebiotics, so that all tested YCF had equivalent amounts, there was no longer a significant difference between YCF iron bioavailability. CONCLUSION: Our results suggest that ascorbic acid and prebiotics in YCF improve iron bioavailability. Ensuring that iron is delivered in a bioavailable form would improve the nutritional benefits of YCF in relation to ID/IDA amongst young children; therefore, further exploration of our findings in vivo is warranted.


Assuntos
Digestão , Enterócitos/metabolismo , Fórmulas Infantis/química , Absorção Intestinal , Ferro da Dieta/metabolismo , Prebióticos/análise , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/análise , Ácido Ascórbico/metabolismo , Biomarcadores/metabolismo , Células CACO-2 , Fenômenos Fisiológicos da Nutrição Infantil , Pré-Escolar , Ferritinas/biossíntese , Alimentos Especializados/análise , Temperatura Alta , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Ferro da Dieta/administração & dosagem , Ferro da Dieta/análise , Valor Nutritivo , Oligossacarídeos/administração & dosagem , Oligossacarídeos/análise , Oligossacarídeos/metabolismo , Prebióticos/administração & dosagem , Trissacarídeos/administração & dosagem , Trissacarídeos/análise , Trissacarídeos/metabolismo
11.
Genet Mol Res ; 15(3)2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27706659

RESUMO

The complementary DNA (cDNA) of the giant panda (Ailuropoda melanoleuca) ferritin light polypeptide (FTL) gene was successfully cloned using reverse transcription-polymerase chain reaction technology. We constructed a recombinant expression vector containing FTL cDNA and overexpressed it in Escherichia coli using pET28a plasmids. The expressed protein was then purified by nickel chelate affinity chromatography. The cloned cDNA fragment was 580 bp long and contained an open reading frame of 525 bp. The deduced protein sequence was composed of 175 amino acids and had an estimated molecular weight of 19.90 kDa, with an isoelectric point of 5.53. Topology prediction revealed one N-glycosylation site, two casein kinase II phosphorylation sites, one N-myristoylation site, two protein kinase C phosphorylation sites, and one cell attachment sequence. Alignment indicated that the nucleotide and deduced amino acid sequences are highly conserved across several mammals, including Homo sapiens, Cavia porcellus, Equus caballus, and Felis catus, among others. The FTL gene was readily expressed in E. coli, which gave rise to the accumulation of a polypeptide of the expected size (25.50 kDa, including an N-terminal polyhistidine tag).


Assuntos
Ferritinas/genética , Ursidae/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Sequência Conservada , Escherichia coli , Ferritinas/biossíntese , Ferritinas/isolamento & purificação , Expressão Gênica , Glicosilação , Ponto Isoelétrico , Peso Molecular , Processamento de Proteína Pós-Traducional , Análise de Sequência de DNA , Análise de Sequência de Proteína
12.
Oncotarget ; 7(48): 78591-78604, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27732930

RESUMO

As a promising magnetic resonance imaging (MRI) reporter, ferritin has been used to track cells in vivo; however, its continuous overexpression can be cytotoxic, which restricts its application. In this study, we aimed to develop a switch to turn this genetic reporter "on" or "off" while monitoring cell grafts via MRI. To accomplish this, we genetically modified the ferritin heavy chain (FTH1) with a Tet-On switch and assessed the expression of FTH1 in transduced neuroblastoma cells (SK-N-SH) in vitro and in xenografted tumors in vivo. We found that FTH1 expression induced by doxycycline (Dox) in SK-N-SH-FTH1 cells depended on treatment dose and duration. We successfully detected T2-weighted MRI contrast in cell grafts after switching "on" the reporter gene using Dox, and this contrast disappeared when we switched it "off". The genetic reporter FTH1 can thus be switched "on" or "off" throughout longitudinal monitoring of cell grafts, limiting expression to when MRI contrast is needed. The controllable imaging system we have developed minimizes risks from constitutive reporter gene overexpression and facilitates tumor cell monitoring in vitro and in vivo.


Assuntos
Biomarcadores Tumorais/genética , Doxiciclina/farmacologia , Ferritinas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Imageamento por Ressonância Magnética , Imagem Molecular/métodos , Neuroblastoma/diagnóstico por imagem , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Relação Dose-Resposta a Droga , Ferritinas/biossíntese , Xenoenxertos , Humanos , Ferro/metabolismo , Masculino , Camundongos Nus , Transplante de Neoplasias , Neuroblastoma/genética , Neuroblastoma/metabolismo , Oxirredutases , Fatores de Tempo , Transdução Genética
13.
Med Hypotheses ; 94: 148-50, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27515221

RESUMO

Metastatic renal cell carcinoma (mRCC) develops in approximately 33% of all renal cancer patients. First line treatment of mRCC includes drugs such as sunitinib, temsirolimus and pazopanib, with overall survival now reaching up to 43,6months in patients with favorable-risk metastatic disease. Several side-effects in mRCC treatment, such as hypothyroidism, can be used as positive prognostic factors and indicate good response to therapy. Hypercholesterolemia and hypertriglyceridemia independent of hypothyroidism are reported as side-effects in temsirolimus treatment and recently in sunitinib treatment, but the exact mechanism and significance of the changes remains elusive. Most likely, metabolic changes are caused by inhibition of mechanistic target of rapamycin (mTOR), a positive target of tumor growth suppression, but also a regulator of iron homeostasis. There are no clinical studies reporting changes in iron and ferritin levels during mRCC biotherapy, but we hypothesize that inhibition of mTOR will also affect iron and ferritin levels. If both lipid and iron changes correlate, there is a high possibility that both changes are primarily caused by mTOR inhibition and the level of change should correlate with the inhibition of mTOR pathway and hence the efficacy of targeted treatment. We lastly hypothesize that mRCC biotherapy causes hypercholesterolemia with a possibly improved cholesterol profile due to increase HDL/LDL ratio, so statins might not have a role as supplementary treatment, whereas a sharp rise in triglyceride levels seems to be the primary target for additional therapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/metabolismo , Ferritinas/sangue , Ferro/sangue , Neoplasias Renais/metabolismo , Metaboloma , Animais , Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/terapia , Colesterol/metabolismo , Ferritinas/biossíntese , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Hipercolesterolemia/metabolismo , Hipertrigliceridemia/metabolismo , Hipotireoidismo/metabolismo , Indazóis , Indóis/uso terapêutico , Neoplasias Renais/patologia , Neoplasias Renais/terapia , Lipídeos/química , Modelos Teóricos , Metástase Neoplásica , Prognóstico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , Sirolimo/análogos & derivados , Sirolimo/metabolismo , Sulfonamidas/uso terapêutico , Sunitinibe , Serina-Treonina Quinases TOR/metabolismo , Triglicerídeos/metabolismo
14.
Biochim Biophys Acta ; 1862(9): 1640-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27287253

RESUMO

Osteogenic differentiation of multipotent mesenchymal stem cells (MSCs) plays a crucial role in bone remodeling. Numerous studies have described the deleterious effect of iron overload on bone density and microarchitecture. Excess iron decreases osteoblast activity, leading to impaired extracellular matrix (ECM) mineralization. Additionally, iron overload facilitates osteoclast differentiation and bone resorption. These processes contribute to iron overload-associated bone loss. In this study we investigated the effect of iron on osteogenic differentiation of human bone marrow MSCs (BMSCs), the third player in bone remodeling. We induced osteogenic differentiation of BMSCs in the presence or absence of iron (0-50µmol/L) and examined ECM mineralization, Ca content of the ECM, mRNA and protein expressions of the osteogenic transcription factor runt-related transcription factor 2 (Runx2), and its targets osteocalcin (OCN) and alkaline phosphatase (ALP). Iron dose-dependently attenuated ECM mineralization and decreased the expressions of Runx2 and OCN. Iron accomplished complete inhibition of osteogenic differentiation of BMSCs at 50µmol/L concentration. We demonstrated that in response to iron BMSCs upregulated the expression of ferritin. Administration of exogenous ferritin mimicked the anti-osteogenic effect of iron, and blocked the upregulation of Runx2, OCN and ALP. Iron overload in mice was associated with elevated ferritin and decreased Runx2 mRNA levels in compact bone osteoprogenitor cells. The inhibitory effect of iron is specific toward osteogenic differentiation of MSCs as neither chondrogenesis nor adipogenesis were influenced by excess iron. We concluded that iron and ferritin specifically inhibit osteogenic commitment and differentiation of BMSCs both in vitro and in vivo.


Assuntos
Ferritinas/biossíntese , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Osteogênese/fisiologia , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/fisiologia , Cálcio/metabolismo , Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ferritinas/farmacologia , Humanos , Ferro/administração & dosagem , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Fosfatos/metabolismo , Fosfatos/farmacologia
15.
PLoS One ; 11(2): e0148802, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26866603

RESUMO

OBJECTIVE: Hepcidin reduces iron absorption by binding to the intestinal iron transporter ferroportin, thereby causing its degradation. Although short-term administration of testosterone or growth hormone (GH) has been reported to decrease circulating hepcidin levels, little is known about how hepcidin is influenced in human endocrine conditions associated with anemia. RESEARCH DESIGN AND METHODS: We used a sensitive and specific dual-monoclonal antibody sandwich immunoassay to measure hepcidin-25 in patients (a) during initiation of in vitro fertilization when endogenous estrogens were elevated vs. suppressed, (b) with GH deficiency before and after 12 months substitution treatment, (c) with hyperthyroidism before and after normalization, and (d) with hyperprolactinemia before and after six months of treatment with a dopamine agonist. RESULTS: In response to a marked stimulation of endogenous estrogen production, median hepcidin levels decreased from 4.85 to 1.43 ng/mL (p < 0.01). Hyperthyroidism, hyperprolactinemia, or GH substitution to GH-deficient patients did not influence serum hepcidin-25 levels. CONCLUSIONS: In humans, gonadotropin-stimulated endogenous estrogen markedly decreases circulating hepcidin-25 levels. No clear and stable correlation between iron biomarkers and hepcidin-25 was seen before or after treatment of hyperthyroidism, hyperprolactinemia or growth hormone deficiency.


Assuntos
Anemia/sangue , Estrogênios/fisiologia , Hepcidinas/sangue , Adolescente , Adulto , Idoso , Anemia/metabolismo , Proteína C-Reativa/biossíntese , Proteína C-Reativa/metabolismo , Agonistas de Dopamina/química , Feminino , Ferritinas/biossíntese , Ferritinas/metabolismo , Fertilização in vitro , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/metabolismo , Humanos , Hiperprolactinemia/complicações , Hipertireoidismo/complicações , Imunoensaio , Ferro/metabolismo , Masculino , Pessoa de Meia-Idade , Prolactinoma/sangue , Transferrina/biossíntese , Transferrina/metabolismo , Adulto Jovem
16.
Neurochem Int ; 91: 72-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26522688

RESUMO

In the light of recent studies, we hypothesized that aspirin might have the functions to regulate the expression of iron transport proteins and then affect cellular iron levels. To test this hypothesis, we investigated the effects of aspirin on expression of iron uptake protein transferrin receptor 1 (TfR1), iron release protein ferroportin 1 (Fpn1) and iron storage protein ferritin using Western blot analysis and on tumor necrosis factor (TNF)-αlpha, interleukin (IL)-6, interleukin (IL)-10 and hepcidin using quantitative real-time PCR in BV-2 microglial cells treated with lipopolysaccharides (LPS). We found that aspirin significantly down-regulated TfR1, while also up-regulated Fpn1 and ferritin expressions in BV-2 microglial cells in vitro. We also showed that TfR1 and Fpn1 expressions were significantly higher, while ferritin contents, IL-6, TNF-alpha and hepcidin mRNA levels were lower in cells treated with aspirin plus LPS than those in cells treated with LPS only. We concluded that aspirin has a negative effect on cell iron contents under 'normal' conditions and could partly reverse LPS-induced-disruption in cell iron balance under in vitro inflammatory conditions. Our findings also suggested that hepcidin might play a dominant role in the control of TfR1 expression by aspirin in the cells treated with LPS.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Ferro/metabolismo , Microglia/metabolismo , Receptores da Transferrina/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Citocinas/biossíntese , Ferritinas/biossíntese , Hepcidinas/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
17.
PLoS One ; 10(8): e0134144, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26270345

RESUMO

Earlier observations indicate that free heme is selectively toxic to cells lacking heme oxygenase-1 (HO-1) but how this enzyme prevents heme toxicity remains unexplained. Here, using A549 (human lung cancer) and immortalized human bronchial epithelial cells incubated with exogenous heme, we find knock-down of HO-1 using siRNA does promote the accumulation of cell-associated heme and heme-induced cell death. However, it appears that the toxic effects of heme are exerted by "loose" (probably intralysosomal) iron because cytotoxic effects of heme are lessened by pre-incubation of HO-1 deficient cells with desferrioxamine (which localizes preferentially in the lysosomal compartment). Desferrioxamine also decreases lysosomal rupture promoted by intracellularly generated hydrogen peroxide. Supporting the importance of endogenous oxidant production, both chemical and siRNA inhibition of catalase activity predisposes HO-1 deficient cells to heme-mediated killing. Importantly, it appears that HO-1 deficiency somehow blocks the induction of ferritin; control cells exposed to heme show ~10-fold increases in ferritin heavy chain expression whereas in heme-exposed HO-1 deficient cells ferritin expression is unchanged. Finally, overexpression of ferritin H chain in HO-1 deficient cells completely prevents heme-induced cytotoxicity. Although two other products of HO-1 activity--CO and bilirubin--have been invoked to explain HO-1-mediated cytoprotection, we conclude that, at least in this experimental system, HO-1 activity triggers the induction of ferritin and the latter is actually responsible for the cytoprotective effects of HO-1 activity.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Heme/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desferroxamina/farmacologia , Ferritinas/biossíntese , Técnicas de Silenciamento de Genes , Heme Oxigenase-1/genética , Humanos , Lisossomos/metabolismo , Lisossomos/patologia
18.
Exp Cell Res ; 336(2): 298-307, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26071936

RESUMO

Wound healing is a complex process that involves the well-coordinated interactions of different cell types. Topical application of high doses of curcumin, a plant-derived polyphenol, enhances both normal and diabetic cutaneous wound healing in rodents. For optimal tissue repair interactions between epidermal keratinocytes and dermal fibroblasts are essential. We previously demonstrated that curcumin increased reactive oxygen species (ROS) formation and apoptosis in dermal fibroblasts, which could be prevented by pre-induction of the cytoprotective enzyme heme oxygenase (HO)-1. To better understand the effects of curcumin on wound repair, we now assessed the effects of high doses of curcumin on the survival of HaCaT keratinocytes and the role of the HO system. We exposed HaCaT keratinocytes to curcumin in the presence or absence of the HO-1 inducers heme (FePP) and cobalt protoporphyrin (CoPP). We then assessed cell survival, ROS formation, and caspase activation. Curcumin induced caspase-dependent apoptosis in HaCaT keratinocytes via a ROS-dependent mechanism. Both FePP and CoPP induced HO-1 expression, but only FePP protected against curcumin-induced ROS formation and caspase-mediated apoptosis. In the presence of curcumin, FePP but not CoPP induced the expression of the iron scavenger ferritin. Together, our data show that the induction of ferritin, but not HO, protects HaCaT keratinocytes against cytotoxic doses of curcumin. The differential response of fibroblasts and keratinocytes to high curcumin doses may provide the basis for improving curcumin-based wound healing therapies.


Assuntos
Antioxidantes/farmacologia , Curcumina/farmacologia , Fibroblastos/metabolismo , Queratinócitos/metabolismo , Cicatrização/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ferritinas/biossíntese , Heme/farmacologia , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/metabolismo , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Protoporfirinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo
19.
Cell Death Dis ; 6: e1690, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25766329

RESUMO

Inflammation and fibrosis are well-defined mechanisms involved in the pathogenesis of the incurable Laminin α2-deficient congenital muscular dystrophy (MDC1A), while apoptosis mechanism is barely discussed. Our previous study showed treatment with Losartan, an angiotensin II type I receptor antagonist, improved muscle strength and reduced fibrosis through transforming growth factor beta (TGF-ß) and mitogen-activated protein kinases (MAPK) signaling inhibition in the dy(2J)/dy(2J) mouse model of MDC1A. Here we show for the first time that Losartan treatment up-regulates and shifts the nuclear factor kappa B (NFκB) signaling pathway to favor survival versus apoptosis/damage in this animal model. Losartan treatment was associated with significantly increased serum tumor necrosis factor alpha (TNF-α) level, p65 nuclei accumulation, and decreased muscle IκB-ß protein level, indicating NFκB activation. Moreover, NFκB anti-apoptotic target genes TNF receptor-associated factor 1 (TRAF1), TNF receptor-associated factor 2 (TRAF2), cellular inhibitor of apoptosis (cIAP2), and Ferritin heavy chain (FTH1) were increased following Losartan treatment. Losartan induced protein expression toward a pro-survival profile as BCL-2 expression levels were increased and Caspase-3 expression levels were decreased. Muscle apoptosis reduction was further confirmed using terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL) assay. Thus, along with TGF-ß and MAPK signaling, NFκB serves as an important regulatory pathway which following Losartan treatment promotes survival in the dy(2J)/dy(2J) mouse model of MDC1A.


Assuntos
Distrofias Musculares/genética , NF-kappa B/genética , Fator 1 Associado a Receptor de TNF/biossíntese , Fator 2 Associado a Receptor de TNF/biossíntese , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Modelos Animais de Doenças , Ferritinas/biossíntese , Humanos , Proteínas Inibidoras de Apoptose/biossíntese , Losartan/administração & dosagem , Camundongos , Força Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/patologia , Transdução de Sinais , Fator 1 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/genética , Fator de Transcrição RelA/metabolismo
20.
Parasitology ; 142(7): 879-89, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25711466

RESUMO

Cystic echinococcosis is a chronic infectious disease that results from a host/parasite interaction. Vaccination with ferritin derived from Echinococcus granulosus is a potential preventative treatment. To understand whether ferritin is capable of inducing a host immune response, we investigated the response of dendritic cells (DCs) to both recombinant ferritin protein and the hydatid fluid (HF) of E. granulosus. We evaluated the immunomodulatory potential of these antigens by performing, immunocytochemistry, electron microscopy and in vivo imaging of monocyte-derived murine DCs. During antigen stimulation of DCs, ferritin cause DCs maturation and induced higher levels of surface marker expression and activated T-cell proliferation and migration. On contrary, HF failed to induce surface marker expression and to stimulate T-cell proliferation. In response to HF, DCs produced interleukin-6 (IL-6), but no IL-12 and IL-10. DCs stimulated with ferritin produced high levels of cytokines. Overall, HF appears to induce host immunosuppression in order to ensure parasite survival via inhibits DC maturation and promotes Th2-dependent secretion of cytokines. Although ferritin also promoted DC maturation and cytokine release, it also activates CD4+T-cell proliferation, but regard of the mechanism of the Eg.ferritin induce host to eradicate E. granulosus were not clear.


Assuntos
Antígenos de Helmintos/farmacologia , Células Dendríticas/efeitos dos fármacos , Echinococcus granulosus/química , Ferritinas/farmacologia , Proteínas de Helminto/farmacologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Misturas Complexas/química , Misturas Complexas/farmacologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Equinococose/parasitologia , Echinococcus granulosus/crescimento & desenvolvimento , Echinococcus granulosus/imunologia , Ferritinas/biossíntese , Expressão Gênica , Interleucina-10/biossíntese , Interleucina-10/imunologia , Interleucina-12/biossíntese , Interleucina-12/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA