Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 349
Filtrar
1.
Hypertension ; 75(2): 413-421, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31838906

RESUMO

Renal fibrosis is the final pathological process common for several types of end-stage renal diseases, including obstructive nephropathy and diabetic kidney disease. Substantial renal iron loads and oxidative stress have been reported to contribute to the development of renal diseases. TfR1 (transferrin receptor 1) plays a crucial role in cellular iron transport. However, there are no studies investigating TfR1 in the pathophysiology of renal fibrosis. Here, we investigate the role of TfR1 in the development of renal fibrosis. Primarily, to ascertain an association of TfR1 in the pathophysiology of renal fibrosis, we induced unilateral ureteral obstruction in wild-type (WT) and heterozygous TfR1 deleted (TfR1+/-) mice. TfR1+/- mice exhibited attenuated renal fibrosis, along with reduced renal expression of ferritin and 4-hydroxynonenal as compared with WT mice after unilateral ureteral obstruction. In addition, renal expression of TGFß-RI (transforming growth factor-ß receptor I) and Smad2, downstream signaling of TGFß-RI was attenuated in TfR1+/- mice compared with WT mice after unilateral ureteral obstruction. Next, we investigated the role of TfR1 in the development of diabetic kidney disease. No difference was observed in blood glucose levels and urinary albumin:creatinine ratios between WT and TfR1+/- diabetic mice after streptozotocin administration. In contrast, TfR1+/- mice showed suppressed renal fibrosis, along with reduced renal expression of ferritin, 4-hydroxynonenal, TGFß-RI, and Smad2 compared with WT mice after streptozotocin administration. These results suggest that TfR1 plays an important role in the development of renal fibrosis.


Assuntos
Regulação da Expressão Gênica , Nefropatias/genética , Rim/patologia , Estresse Oxidativo , RNA/genética , Receptores da Transferrina/genética , Animais , Western Blotting , Modelos Animais de Doenças , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Triagem de Portadores Genéticos/métodos , Rim/metabolismo , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Transgênicos , Receptores da Transferrina/biossíntese , Transdução de Sinais
2.
ACS Chem Neurosci ; 10(12): 4824-4833, 2019 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-31682397

RESUMO

Morphine tolerance is a classic, challenging clinical issue. However, the mechanism underlying this phenomenon remains poorly understood. Recently, studies have shown that ferroptosis correlates with drug resistance. Therefore, this study investigated whether spinal cord ferroptosis contributes to morphine tolerance. C57BL/6 mice were continuously subcutaneously injected with morphine, with or without the ferroptosis inhibitor liproxstatin-1. We found that chronic morphine exposure led to morphine antinociception tolerance, accompanied by loss of spinal cord neurons, increase in the levels of iron, malondialdehyde, and reactive oxygen species, and decreases in the levels of superoxide dismutase. Additionally, inflammatory response and mitochondrial shrinkage, processes that are involved in ferroptosis, were observed. Simultaneously, we found that 10 mg/kg of liproxstatin-1 could alleviate iron overload by balancing transferrin receptor protein 1/ferroportin expression and attenuate morphine tolerance by increasing glutathione peroxidase 4 levels, while reducing the levels of malondialdehyde and reactive oxygen species. It also downregulated the expression of extracellularly regulated protein kinases that had been induced by chronic morphine exposure. Our results indicate that spinal cord ferroptosis contributes to morphine tolerance, while liproxstatin-1 attenuates the development of morphine tolerance. These findings suggest that ferroptosis may be a potential therapeutic target for morphine tolerance.


Assuntos
Ferroptose/efeitos dos fármacos , Morfina/farmacologia , Nociceptividade/efeitos dos fármacos , Quinoxalinas/farmacologia , Medula Espinal/efeitos dos fármacos , Compostos de Espiro/farmacologia , Animais , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Tolerância a Medicamentos/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Inflamação , Ferro/metabolismo , Sobrecarga de Ferro/tratamento farmacológico , Peroxidação de Lipídeos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Morfina/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/biossíntese , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Distribuição Aleatória , Espécies Reativas de Oxigênio/metabolismo , Receptores da Transferrina/biossíntese , Receptores da Transferrina/genética , Medula Espinal/patologia , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética
3.
J Orthop Surg Res ; 14(1): 296, 2019 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-31484533

RESUMO

BACKGROUND: Osteosarcoma is aggressive and prognostic biomarkers are important to predict the outcomes of surgery and chemotherapy. Here, we investigated the potential of transferrin receptor-1 (TfR1) and vascular endothelial growth factor (VEGF) as prognostic markers of osteosarcoma. METHODS: TfR1 and VEGF in osteosarcoma samples from a cohort of 53 osteosarcoma patients were detected by immunohistochemistry analysis. The correlation of TfR1 and VEGF levels with clinicopathological parameters was analyzed by Pearson chi-square and Spearman-rho tests. Overall patient survival was analyzed by the Kaplan-Meier method. RESULTS: We found that TfR1 and VEGF expression levels were low in 20.8% and 18.9%; modest in 35.8% and 35.8%; and high in 43.4% and 45.3% of osteosarcoma patients, respectively. TfR1 and VEGF expression was significantly correlated to histologic grade, Enneking stage, and distant metastasis. TfR1 expression was significantly correlated to VEGF expression and both TfR1 expression and VEGF expression were correlated to shorter overall survival. CONCLUSIONS: TfR1 and VEGF are potential prognostic factors for osteosarcoma.


Assuntos
Antígenos CD/biossíntese , Biomarcadores Tumorais/biossíntese , Neoplasias Ósseas/metabolismo , Osteossarcoma/metabolismo , Receptores da Transferrina/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adolescente , Antígenos CD/genética , Biomarcadores Tumorais/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/mortalidade , Estudos de Coortes , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Osteossarcoma/genética , Osteossarcoma/mortalidade , Prognóstico , Receptores da Transferrina/genética , Fator A de Crescimento do Endotélio Vascular/genética , Adulto Jovem
4.
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
5.
Nanomedicine ; 21: 102064, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31326524

RESUMO

As a magnetic resonance imaging (MRI) reporter gene, MagA has become a powerful tool to monitor dynamic gene expression and allowed concomitant high resolution anatomical and functional imaging of subcellular genetic information. Here we establish a stably expressed MagA method for lung cancer MRI. The results show that MagA can not only enhance both in vitro and in vivo MRI contrast by specifically alternating the transverse relaxation rate of water, but also inhibit the malignant growth of lung tumor. In addition, MagA can regulate magnetic nanoparticle production in grafted tissues and also suppress transferrin receptor expression by acting as an iron transporter, and meanwhile can permit iron biomineralization in the presence of mammalian iron homeostasis. This work provides experimental evidence for the safe preclinical applications of MagA as both a potential inhibitor and an MRI-based tracing tool for iron ion-dependent lung cancer.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte de Cátions , Genes Reporter , Ferro/metabolismo , Neoplasias Pulmonares , Imageamento por Ressonância Magnética , Proteínas de Neoplasias , Neoplasias Experimentais , Receptores da Transferrina , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Receptores da Transferrina/biossíntese , Receptores da Transferrina/genética
6.
Biochem Pharmacol ; 166: 93-107, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31078603

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the most common and aggressive cancers worldwide, especially in China, with poor prognosis due to the lack of effective therapeutic strategies. Here, the anticancer effect and pharmacological mechanism of a newly synthesized Fe(II) phenanthroline complex was studied in ESCC. Our data showed that transferrin receptor 1 (TFR1) was specifically overexpressed in ESCC tissues compared to its expression in normal esophageal tissues, a finding further supported by public datasets. The newly synthesized Fe(II) complex was selectively transported into ESCC cells overexpressing TFR1 through TFR1-mediated endocytosis and exhibited anticancer activity in a dose-dependent manner. The mechanistic study elucidated that the Fe(II) complex caused cell cycle arrest at the G0/G1 phase by blocking the CDK4/6-cyclin D1 complex and induced mitochondria-mediated apoptosis. Furthermore, exposure to the Fe(II) complex led to excessive reactive oxygen species (ROS) accumulation by thioredoxin reductase (TrxR) inhibition and DNA double-strand breaks (DSBs), which in turn sequentially activated ATM, CHK1/2 and p53. Moreover, combination treatment with cisplatin and the Fe(II) complex exhibited a synergistic effect in ESCC cells. Taken together, our results initially suggest the potential application of the Fe(II) complex in ESCC chemotherapy, especially for patients with TFR1 overexpression.


Assuntos
Antígenos CD/biossíntese , Antineoplásicos/uso terapêutico , Dano ao DNA/efeitos dos fármacos , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Fenantrolinas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Receptores da Transferrina/biossíntese , Antígenos CD/genética , Antineoplásicos/química , Antineoplásicos/farmacologia , Dano ao DNA/fisiologia , Relação Dose-Resposta a Droga , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Compostos de Ferro/química , Compostos de Ferro/farmacologia , Compostos de Ferro/uso terapêutico , Fenantrolinas/química , Fenantrolinas/farmacologia , Receptores da Transferrina/genética
7.
Blood Adv ; 2(15): 1914-1922, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30093529

RESUMO

To identify novel regulators of erythropoiesis, we performed independent forward genetic screens using the chemical mutagen ENU in mice. Among progeny displaying microcytic red-cell phenotypes, 7 independent mouse strains harboring mutations within the transferrin receptor gene Tfrc were identified. Six of the mutants, including the previously described red blood cell 6 (RBC6) strain, displayed reduced erythroblast CD71 expression and midgestation lethality of homozygotes (E12.5-E14.5), and 1 novel strain, RBC21, displayed a variable phenotype with sustained CD71 expression and late homozygous lethality (E18.5). Standard iron studies were normal in the RBC21 mutant, but intracellular ferritin was significantly reduced. The microcytic phenotype seen in the RBC21 strain was the result of impaired binding of transferrin to the receptor. Neither RBC6 nor RBC21 responded to iron replacement therapy. These studies describe how point mutations of the transferrin receptor can cause a microcytic anemia that does not respond to iron therapy and would not be detected by routine iron studies, such as serum ferritin.


Assuntos
Anemia , Antígenos CD/biossíntese , Eritrócitos/metabolismo , Ferritinas/sangue , Mutação Puntual , Receptores da Transferrina/biossíntese , Receptores da Transferrina/genética , Anemia/sangue , Anemia/genética , Anemia/patologia , Animais , Antígenos CD/genética , Eritrócitos/patologia , Camundongos , Camundongos Mutantes , Receptores da Transferrina/metabolismo
8.
J Pharm Pharmacol ; 70(10): 1357-1368, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30051477

RESUMO

OBJECTIVES: Cisplatin is one of the widely used antitumour agents with major clinical side effect, nephrotoxicity. We showed the role of iron in cisplatin-induced nephrotoxicity that entrance to the cell via transferrin receptor (TfR) as a gatekeeper for iron uptake. We also examined the effect of iron chelator deferiprone against this toxicity. METHODS: Thirty male Wistar rats were randomly divided into six groups. Group I (saline orally for 10 days); group II (saline orally for 10 days plus single injection of cisplatin 7 mg/kg, intraperitoneally on 5th day); groups III, IV and V (deferiprone 50, 100 and 200 mg/kg orally for 10 days, respectively, plus cisplatin on 5th day). Group VI (deferiprone, orally). RESULTS: Deferiprone provided functional and significant histological-proven protection in group IV. Deferiprone attenuated the increased creatinine, BUN, malondialdehyde and iron concentrations in cisplatin-injected animals. The increased amounts of TfR and decreased levels of HIF-1α and related anti-apoptotic genes expression in cisplatin-treated animals were improved by deferiprone. CONCLUSIONS: The results supported a role for iron in cisplatin-induced nephrotoxicity and TfR may serve as an important source of iron. Based on these findings, deferiprone pretreatment may play a role in preventing cisplatin-induced nephropathy in cancer patient.


Assuntos
Cisplatino/antagonistas & inibidores , Deferiprona/farmacologia , Insuficiência Renal/prevenção & controle , Aconitato Hidratase/metabolismo , Administração Oral , Animais , Nitrogênio da Ureia Sanguínea , Cisplatino/efeitos adversos , Creatinina/sangue , Deferiprona/administração & dosagem , Relação Dose-Resposta a Droga , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Ferro/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído , Substâncias Protetoras/farmacologia , Ratos , Receptores da Transferrina/biossíntese , Insuficiência Renal/induzido quimicamente , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Superóxido Dismutase/metabolismo
9.
Alcohol Clin Exp Res ; 42(6): 1022-1033, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29672865

RESUMO

BACKGROUND: Prenatal alcohol exposure (PAE) causes neurodevelopmental disability. Clinical and animal studies show gestational iron deficiency (ID) exacerbates PAE's behavioral and growth deficits. In rat, PAE manifests an inability to establish iron homeostasis, increasing hepcidin (maternal and fetal), and fetal liver iron while decreasing brain iron and promoting anemia. Here, we hypothesize dietary iron fortification during pregnancy may mitigate alcohol's disruption of fetal iron homeostasis. METHODS: Pregnant Long-Evans rats, fed iron-sufficient (100 ppm iron) or iron-fortified (IF; 500 ppm iron) diets, received either 5 g/kg alcohol (PAE) or isocaloric maltodextrin daily on gestational days (GD) 13.5 through 19.5. Maternal and fetal outcomes were evaluated on GD20.5. RESULTS: PAE reduced mean fetal weight (p < 0.001) regardless of maternal iron status, suggesting iron fortification did not improve fetal growth. Both PAE (p < 0.01) and IF (p = 0.035) increased fetal liver iron. In fetal brain, PAE (p = 0.015) affected total (p < 0.001) and nonheme iron (p < 0.001) such that iron fortification normalized (p = 0.99) the alcohol-mediated reductions in brain iron and nonheme iron. Iron fortification also improved fetal hematologic indices in PAE including hemoglobin, hematocrit, and mean cell volume (ps<0.001). Iron fortification also normalized hepcidin expression in alcohol-exposed maternal and fetal liver. Neither diet nor PAE affected transferrin (Tf) and ferritin (FTN) content in fetal liver, nor Tf or transferrin receptor in fetal brain. However, IF-PAE fetal brains trended to less FTN content (p = 0.074), suggesting greater availability of nonstorage iron. In PAE, hepcidin levels were linearly related to increased liver iron stores and decreased red blood cell count and brain iron. CONCLUSIONS: Maternal oral iron fortification mitigated PAE's disruption of fetal iron homeostasis and improved brain iron content, hematologic indices, and hepcidin production in this rat PAE model. Clinical studies show maternal ID substantially enhances fetal vulnerability to PAE, and our work supports increased maternal dietary iron intake may improve fetal iron status in alcohol-exposed pregnancies.


Assuntos
Feto/irrigação sanguínea , Hepcidinas/biossíntese , Ferro da Dieta/farmacologia , Ferro/metabolismo , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Animais , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Índices de Eritrócitos/efeitos dos fármacos , Feminino , Ferritinas/metabolismo , Desenvolvimento Fetal , Feto/efeitos dos fármacos , Hematócrito , Hemoglobinas/efeitos dos fármacos , Homeostase , Fígado/metabolismo , Masculino , Gravidez , Ratos , Receptores da Transferrina/biossíntese , Transferrina/metabolismo
10.
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
11.
J Cell Biochem ; 119(8): 6452-6460, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29323739

RESUMO

The RhoA/Rho-kinase cascade plays an important role in many aspects of cardiovascular function. This study aims to investigate the protective effects of fasudil, a Rho-kinase inhibitor, on pressure overload induced heart failure in rats. Pressure overload induced heart failure was induced in SD rats by banding the abdominal aorta for 8 weeks. The rats were divided into four groups: Sham, TAC, TAC plus low dose of fasudil, and TAC plus high dose of fasudil group. Low dose and high dose fasudil were 5 and 10 mg/kg/day, respectively. Rats in the Sham and TAC groups were treated with vehicle. Fasudil effectively inhibited TAC-induced heart failure, as evaluated by echocardiography and transmission electron microscopy. Fasudil could significantly promote superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) activity and significantly decrease malondialdehyde (MDA) content in a dose-dependent maner in TAC rats. Consistently, fasudil evoked significant nuclear translocation of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) with increased DNA/promoter binding and transactivation of Nrf2 targets. In addition, fasudil increased the content of iron as well as transferrin receptor 1 (TfR1) in TAC rats. A mild oxidative stress induced by iron may activate the antioxidant enzymes by feedback response. Taken together, these results indicate that the protective effect of fasudil may be due to its strong antioxidative activities which related with the activated Nrf2 and its down-regulated genes. These findings provide a new treatment concept and support the benefit of fasudil treatment in heart failure.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Antioxidantes/metabolismo , Insuficiência Cardíaca/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Modelos Animais de Doenças , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores da Transferrina/biossíntese
12.
RNA ; 24(4): 468-479, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29295890

RESUMO

Intracellular iron is tightly regulated by coordinated expression of iron transport and storage genes, such as transferrin receptor-1 (TfR1) and ferritin. They are primarily regulated by iron through iron-induced dissociation of iron-regulatory proteins (IRPs) from iron-responsive elements (IREs) in the 3'-UTR (untranslated region) of TfR1 or 5'-UTR of ferritin mRNA, resulting in destabilization of TfR1 mRNA and release of ferritin translation block. Thus high iron decreases iron transport via TfR1 mRNA degradation and increases iron storage via ferritin translational up-regulation. However, the molecular mechanism of TfR1 mRNA destabilization in response to iron remains elusive. Here, we demonstrate that miR-7-5p and miR-141-3p target 3'-TfR1 IREs and down-regulate TfR1 mRNA and protein expression. Conversely, miR-7-5p and miR-141-3p antagomiRs partially but significantly blocked iron- or IRP knockdown-induced down-regulation of TfR1 mRNA, suggesting the interplay between these microRNAs and IRPs along with involvement of another uncharacterized mechanism in TfR1 mRNA degradation. Luciferase reporter assays using 3'-UTR TfR1 IRE mutants suggested that the IREs C and E are targets of miR-7-5p and miR-141-3p, respectively. Furthermore, miR-7 expression was inversely correlated with TfR1 mRNA in human pancreatic adenocarcinoma patient samples. These results suggest a role of microRNAs in the TfR1 regulation in the IRP-IRE system.


Assuntos
Antígenos CD/genética , Proteínas Reguladoras de Ferro/genética , MicroRNAs/genética , RNA Mensageiro/biossíntese , Receptores da Transferrina/genética , Células 3T3 , Animais , Antígenos CD/biossíntese , Proliferação de Células/genética , Ferritinas/genética , Humanos , Ferro/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Receptores da Transferrina/biossíntese
13.
PLoS One ; 13(12): e0206332, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30596656

RESUMO

The bloodstream form of the parasite Trypanosoma brucei obtains iron from its mammalian host by receptor-mediated endocytosis of host transferrin through its own unique transferrin receptor (TbTfR). Expression of TbTfR rapidly increases upon iron starvation by post-transcriptional regulation through a currently undefined mechanism that is distinct from the mammalian iron response system. We have created reporter cell lines by fusing the TbTfR 3'UTR or a control Aldolase 3'UTR to reporter genes encoding GFP or firefly Luciferase, and inserted the fusions into a bloodstream form cell line at a tagged ribosomal RNA locus. Fusion of the TbTfR 3'UTR is sufficient to significantly repress the expression of the reporter proteins under normal growth conditions. Under iron starvation conditions we observed upregulation of the mRNA and protein level of the TbTfR 3'UTR fusions only, with a magnitude and timing consistent with that reported for upregulation of the TbTfR. We conclude that the dynamic regulation of the T. brucei transferrin receptor in response to iron starvation is mediated via its 3'UTR, and that the effect is independent of genomic location.


Assuntos
Regiões 3' não Traduzidas , Regulação da Expressão Gênica , Deficiências de Ferro , Proteínas de Protozoários/biossíntese , Receptores da Transferrina/biossíntese , Trypanosoma brucei brucei/metabolismo , Linhagem Celular , Humanos , Proteínas de Protozoários/genética , Receptores da Transferrina/genética , Trypanosoma brucei brucei/genética
14.
Tumour Biol ; 39(7): 1010428317717655, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28671021

RESUMO

Labile iron pool is a cellular source of ions available for Fenton reactions resulting in oxidative stress. Living organisms avoid an excess of free irons by a tight control of iron homeostasis. We investigated the altered expression of iron regulatory proteins and iron discrimination in the development of liver fluke-associated cholangiocarcinoma. Additionally, the levels of labile iron pool and the functions of transferrin receptor-1 on cholangiocarcinoma development were also identified. Iron deposition was determined using the Prussian blue staining method in human cholangiocarcinoma tissues. We investigated the alteration of iron regulatory proteins including transferrin, transferrin receptor-1, ferritin, ferroportin, hepcidin, and divalent metal transporter-1 in cholangiocarcinoma tissues using immunohistochemistry. The clinicopathological data of cholangiocarcinoma patients and the expressions of proteins were analyzed. Moreover, the level of intracellular labile iron pool in cholangiocarcinoma cell lines was identified by the RhoNox-1 staining method. We further demonstrated transferrin receptor-1 functions on cell proliferation and migration upon small interfering RNA for human transferrin receptor 1 transfection. Results show that Iron was strongly stained in tumor tissues, whereas negative staining was observed in normal bile ducts of healthy donors. Interestingly, high iron accumulation was significantly correlated with poor prognosis of cholangiocarcinoma patients. The expressions of iron regulatory proteins in human cholangiocarcinoma tissues and normal liver from cadaveric donors revealed that transferrin receptor-1 expression was increased in the cancer cells of cholangiocarcinoma tissues when compared with the adjacent normal bile ducts and was significantly correlated with cholangiocarcinoma metastasis. Labile iron pool level and transferrin receptor-1 expression were significantly increased in KKU-214 and KKU-213 when compared with cholangiocyte cells (MMNK1). Additionally, the suppression of transferrin receptor-1 expression significantly decreased intracellular labile iron pool, cholangiocarcinoma migration, and cell proliferation when compared with control media and control small interfering RNA. In Conclusion, high expression of transferrin receptor-1 resulting in iron uptake contributes to increase in the labile iron pool which plays roles in cholangiocarcinoma progression with aggressive clinical outcomes.


Assuntos
Antígenos CD/biossíntese , Colangiocarcinoma/genética , Proteínas Reguladoras de Ferro/genética , Ferro/metabolismo , Estresse Oxidativo/genética , Receptores da Transferrina/biossíntese , Adulto , Idoso , Antígenos CD/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patologia , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Reguladoras de Ferro/metabolismo , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Receptores da Transferrina/genética
15.
J Orthop Surg Res ; 12(1): 75, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28521818

RESUMO

BACKGROUND: Central pain (CP) is a common clinical problem in patients with spinal cord injury (SCI). Recent studies found the pathogenesis of CP was related to the remodeling of the brain. We investigate the roles of iron overload and subsequent microglia activate in the remodeling of the brain after SCI. METHODS: An SCI-induced CP model was established in Sprague-Dawley rats that were randomly assigned to SCI, sham operation, deferoxamine (DFX), minocycline, and nitric oxide synthase inhibitor treatment groups. At 12 weeks, pain behavior and thermal pain threshold were evaluated in each group, and iron transferrin receptor (TfR)1 and ferritin (Fn) mRNA, as well as iron-regulatory protein (IRP)1, FN, lactoferrin, and nuclear factor (NF)-κB protein levels in the rat brains were measured. Microglia proliferation and differentiation and IRP1 expression were evaluated by immunohistochemistry. RESULTS: Autophagy was observed in rats after SCI, accompanied by reduced latency of thermal pain, increased iron content and IRP1 and NF-κB levels in the hindlimb sensory area, hippocampus, and thalamus, and decreased Fn levels in the hindlimb sensory area. TfR1 mRNA expression was upregulated in activated microglia. Treatment with an iron-chelating agent, or inhibitors of nitric oxide synthase or microglia suppressed microglia proliferation. CONCLUSIONS: SCI may induce intracranial iron overload, which activates microglia via NF-κB signaling. Microglia secrete inflammatory factors that induce neuronal damage and lead to CP. Treatment with an iron-chelating agent or NF-κB or microglia inhibitors can relieve CP resulting from SCI.


Assuntos
Córtex Cerebral/metabolismo , Sobrecarga de Ferro/metabolismo , Microglia/metabolismo , Dor/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Córtex Cerebral/patologia , Feminino , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/patologia , Microglia/patologia , Dor/etiologia , Dor/patologia , Ratos , Ratos Sprague-Dawley , Receptores da Transferrina/biossíntese , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/patologia , Vértebras Torácicas
16.
Cell Immunol ; 316: 41-52, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28372797

RESUMO

Plasmacytoid dendritic cells (pDC) are of increasing interest in cancer vaccine development, but many functions of these highly specialized, multifaceted cells are poorly understood. The transferrin receptor CD71 has also been suggested to function as an antigen uptake receptor on professional antigen-presenting cells. In this study, we employed multiparameter flow cytometry to investigate CD71 expression on various leukocyte subsets, including DC subsets, granulocytes, macrophages, T and B lymphocytes, γδ T cells, and natural killer cells. Cells from various lymphoid and non-lymphoid murine tissues were analyzed using fluorochrome-conjugated monoclonal antibodies. High CD71 expression (90-100%) was observed, uniquely on pDC amongst the leukocyte populations examined, in both lymphoid and non-lymphoid tissues, including other DC subsets. In contrast, CD71 expression on non-tissue pDC, in the bone marrow and peripheral blood, was reduced. The cause and function of this high tissue pDC-selective CD71 expression remain to be examined.


Assuntos
Antígenos CD/biossíntese , Receptores da Transferrina/biossíntese , Animais , Antígenos CD/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Granulócitos/imunologia , Granulócitos/metabolismo , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Receptores da Transferrina/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
18.
Curr Pharm Des ; 23(3): 454-466, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27784246

RESUMO

A lot of effort has been devoted to achieving active targeting for cancer therapy in order to reach the right cells. Hence, increasingly it is being realized that active-targeted nanocarriers notably reduce off-target effects, mainly because of targeted localization in tumors and active cellular uptake. In this context, by taking advantage of the overexpression of transferrin receptors on the surface of tumor cells, transferrin-conjugated nanodevices have been designed, in hope that the biomarker grafting would help to maximize the therapeutic benefit and to minimize the side effects. Notably, active targeting nanoparticles have shown improved therapeutic performances in different tumor models as compared to their passive targeting counterparts. In this review, current development of nano-based devices conjugated with transferrin for active tumor-targeting drug delivery are highlighted and discussed. The main objective of this review is to provide a summary of the vast types of nanomaterials that have been used to deliver different chemotherapeutics into tumor cells, and to ultimately evaluate the progression on the strategies for cancer therapy in view of the future research.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Receptores da Transferrina/antagonistas & inibidores , Transferrina/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Humanos , Nanotecnologia , Neoplasias/metabolismo , Neoplasias/patologia , Receptores da Transferrina/biossíntese , Transferrina/síntese química , Transferrina/química
19.
Exp Eye Res ; 151: 122-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27565570

RESUMO

Iron accumulation in the retina is associated with the development of age-related macular degeneration (AMD). IV iron is a common method to treat iron deficiency anemia in adults, and its retinal manifestations have not hitherto been identified. To assess whether IV iron formulations can be retina-toxic, we generated a mouse model for iron-induced retinal damage. Male C57BL/6J mice were randomized into groups receiving IV iron-sucrose (+Fe) or 30% sucrose (-Fe). Iron levels in neurosensory retina (NSR), retinal pigment epithelium (RPE), and choroid were assessed using immunofluorescence, quantitative PCR, and the Perls' iron stain. Iron levels were most increased in the RPE and choroid while levels in the NSR did not differ significantly in +Fe mice compared to controls. Eyes from +Fe mice shared histological features with AMD, including Bruch's membrane (BrM) thickening with complement C3 deposition, as well as RPE hypertrophy and vacuolization. This focal degeneration correlated with areas of high choroidal iron levels. Ultrastructural analysis provided further detail of the RPE/photoreceptor outer segment vacuolization and Bruch's membrane thickening. Findings were correlated with a clinical case of a 43-year-old patient who developed numerous retinal drusen, the hallmark of AMD, within 11 months of IV iron therapy. Our results suggest that IV iron therapy may have the potential to induce or exacerbate a form of retinal degeneration. This retinal degeneration shares features with AMD, indicating the need for further study of AMD risk in patients receiving IV iron treatment.


Assuntos
Compostos Férricos/efeitos adversos , Ácido Glucárico/efeitos adversos , Ferro/metabolismo , Degeneração Macular/induzido quimicamente , Estresse Oxidativo/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia , Animais , Apoferritinas/biossíntese , Apoferritinas/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Compostos Férricos/administração & dosagem , Óxido de Ferro Sacarado , Regulação da Expressão Gênica , Ácido Glucárico/administração & dosagem , Injeções Intravenosas , Degeneração Macular/genética , Degeneração Macular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores da Transferrina/biossíntese , Receptores da Transferrina/genética , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo
20.
Neoplasma ; 63(5): 809-15, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27468886

RESUMO

CD71 (transferrin receptor 1, TfR-1) is a type II membrane glycoprotein and associated closely with tumors. It was recognized as an indication for diagnosing acute erythroid leukemia (AEL). High expression level of CD71 has been identified as a negative prognostic marker for many solid tumors. However, whether CD71 should be identified as an adverse marker in acute myeloid leukemia (AML) remained conflicting. We studied 214 AML patients for analysis of clinical and laboratory data. Taking the CD71 expression level of 60% as a standard, we divided our patients into two groups. We discovered that AML with high expression level of CD71 was prone to linked with severe anemia (P=0.004), thrombocytopenia (P<0.001) and complex karyotype (P=0.024) and had increasing expression level of CD117 (P=0.001). No statistically significant correlations in age, gender, WBC counting, molecular markers between the two groups. And moreover, high expression level of CD71 did not alter the pattern of survival time.


Assuntos
Anemia/patologia , Antígenos CD/biossíntese , Leucemia Mieloide Aguda/patologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores da Transferrina/biossíntese , Trombocitopenia/patologia , Anemia/diagnóstico , Antígenos CD/genética , Proliferação de Células/genética , Intervalo Livre de Doença , Feminino , Humanos , Leucemia Mieloide Aguda/diagnóstico , Masculino , Pessoa de Meia-Idade , Receptores da Transferrina/genética , Fatores de Risco , Trombocitopenia/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA