Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
1.
Pharmacol Ther ; 228: 107927, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34171326

RESUMO

Complex regulation of the immune response is necessary to support effective defense of an organism against hostile invaders and to maintain tolerance to harmless microorganisms and autoantigens. Recent studies revealed previously unappreciated roles of CD71+ erythroid cells (CECs) in regulation of the immune response. CECs physiologically reside in the bone marrow where erythropoiesis takes place. Under stress conditions, CECs are enriched in some organs outside of the bone marrow as a result of extramedullary erythropoiesis. However, the role of CECs goes well beyond the production of erythrocytes. In neonates, increased numbers of CECs contribute to their vulnerability to infectious diseases. On the other side, neonatal CECs suppress activation of immune cells in response to abrupt colonization with commensal microorganisms after delivery. CECs are also enriched in the peripheral blood of pregnant women as well as in the placenta and are responsible for the regulation of feto-maternal tolerance. In patients with cancer, anemia leads to increased frequency of CECs in the peripheral blood contributing to diminished antiviral and antibacterial immunity, as well as to accelerated cancer progression. Moreover, recent studies revealed the role of CECs in HIV and SARS-CoV-2 infections. CECs use a full arsenal of mechanisms to regulate immune response. These cells suppress proinflammatory responses of myeloid cells and T-cell proliferation by the depletion of ʟ-arginine by arginase. Moreover, CECs produce reactive oxygen species to decrease T-cell proliferation. CECs also secrete cytokines, including transforming growth factor ß (TGF-ß), which promotes T-cell differentiation into regulatory T-cells. Here, we comprehensively describe the role of CECs in orchestrating immune response and indicate some therapeutic approaches that might be used to regulate their effector functions in the treatment of human conditions.


Assuntos
Antígenos CD , Células Eritroides , Imunidade , Receptores da Transferrina , Antígenos CD/fisiologia , COVID-19 , Células Eritroides/metabolismo , Humanos , Imunidade/fisiologia , Receptores da Transferrina/fisiologia
2.
J Biol Chem ; 296: 100426, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33609526

RESUMO

Birt-Hogg-Dubé (BHD) syndrome is a multiorgan disorder caused by inactivation of the folliculin (FLCN) protein. Previously, we identified FLCN as a binding protein of Rab11A, a key regulator of the endocytic recycling pathway. This finding implies that the abnormal localization of specific proteins whose transport requires the FLCN-Rab11A complex may contribute to BHD. Here, we used human kidney-derived HEK293 cells as a model, and we report that FLCN promotes the binding of Rab11A with transferrin receptor 1 (TfR1), which is required for iron uptake through continuous trafficking between the cell surface and the cytoplasm. Loss of FLCN attenuated the Rab11A-TfR1 interaction, resulting in delayed recycling transport of TfR1. This delay caused an iron deficiency condition that induced hypoxia-inducible factor (HIF) activity, which was reversed by iron supplementation. In a Drosophila model of BHD syndrome, we further demonstrated that the phenotype of BHD mutant larvae was substantially rescued by an iron-rich diet. These findings reveal a conserved function of FLCN in iron metabolism and may help to elucidate the mechanisms driving BHD syndrome.


Assuntos
Antígenos CD/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores da Transferrina/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/fisiologia , Síndrome de Birt-Hogg-Dubé/metabolismo , Síndrome de Birt-Hogg-Dubé/fisiopatologia , Citoplasma/metabolismo , Proteínas de Drosophila , Drosophila melanogaster , Células HEK293 , Homeostase , Humanos , Ferro/metabolismo , Modelos Animais , Proteínas Proto-Oncogênicas/fisiologia , Receptores da Transferrina/genética , Receptores da Transferrina/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo
3.
Life Sci ; 260: 118305, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32827544

RESUMO

AIM: The study aims to investigate the roles of LncRNA and miRNA in ferroptosis in brain ischemia/reperfusion (I/R) in vivo and in vitro. MATERIALS AND METHODS: qPCR assay was used to analyze lncRNA PVT1 and miR-214 expressions in acute ischemic stroke (AIS) patients. Then, we established brain I/R mice models and OGD/R PC12 cell models to analyze the mechanism of ferroptosis. I/R mice were treated by lncRNA PVT silencing or miR-214 overexpressing lentivirus via lateral ventricles. Infarct size was analyzed by TTC staining, accompanied by the detection of ferroptosis indicators through Perls'Prussian blue staining, iron kit, MDA kit, glutathione kit, GPx activities kit and Western blotting (WB). Dual luciferase reporter assay was used to assess whether miR-214 bound to PVT1, TP53 or TFR1. Co-IP analyzed the interplay of p53 with SLC7A11. KEY FINDINGS: We found that the levels of PVT1 were upregulated and miR-214 levels were downregulated in plasma of AIS patients. NIHSS score was positively correlated with PVT1 levels but was negatively with miR-214 levels. PVT1 silencing or miR-214 overexpression significantly reduced infarct size and suppressed ferroptosis in vivo. miR-214 overexpression markedly decreased PVT1 levels. Specifically, miR-214 could bind to 3'untranslated region (3'UTR) of PVT1, TP53 or TFR1. PVT1 overexpression or miR-214 silencing markedly abolished the effects of Ferrostatin-1 on ferroptosis indicators except for TFR1 expression. Besides, miR-214 silencing counteracted the effects of PVT1 knockdown on the ferroptosis-related proteins. CONCLUSION: PVT1 regulated ferroptosis through miR-214-mediated TFR1 and TP53 expression. There was a positive feedback loop of lncRNA PVT1/miR-214/p53 possibly.


Assuntos
Antígenos CD/fisiologia , Ferroptose/fisiologia , MicroRNAs/fisiologia , RNA Longo não Codificante/fisiologia , Receptores da Transferrina/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Isquemia Encefálica/fisiopatologia , Cicloexilaminas/farmacologia , Modelos Animais de Doenças , Feminino , Ferroptose/efeitos dos fármacos , Inativação Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/sangue , MicroRNAs/genética , Pessoa de Meia-Idade , Células PC12 , Fenilenodiaminas/farmacologia , RNA Longo não Codificante/sangue , RNA Longo não Codificante/genética , Ratos , Traumatismo por Reperfusão , Acidente Vascular Cerebral/fisiopatologia
4.
J Nutr ; 149(9): 1660-1666, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31162576

RESUMO

BACKGROUND: Early-life iron deficiency (ID) impairs hippocampal energy production. Whether there are changes in glucose transporter (GLUT) expression is not known. OBJECTIVE: The aim of this study was to investigate whether early-life ID and the treatment iron dose alter brain regional GLUT expression in adult rats and mice. METHODS: In Study 1, ID was induced in male and female Sprague Dawley rat pups by feeding dams a 3-mg/kg iron diet during gestation and the first postnatal week, followed by treatment using low-iron [3-10 mg/kg; formerly iron-deficient (FID)-10 group], standard-iron (40-mg/kg; FID-40 group), or high-iron (400-mg/kg; FID-400 group) diets until weaning. The control group received the 40 mg/kg iron diet. GLUT1, GLUT3, hypoxia-inducible factor (HIF)-1α, and prolyl-hydroxylase-2 (PHD2) mRNA and protein expression in the cerebral cortex, hippocampus, striatum, cerebellum, and hypothalamus were determined at adulthood. In Study 2, the role of hippocampal ID in GLUT expression was examined by comparing the Glut1, Glut3, Hif1α, and Phd2 mRNA expression in adult male and female wild-type (WT) and nonanemic hippocampal iron-deficient and iron-replete dominant negative transferrin receptor 1 (DNTfR1-/-) transgenic mice. RESULTS: In Study 1, Glut1, Glut3, and Hif1α mRNA, and GLUT1 55-kDa protein expression was upregulated 20-33% in the hippocampus of the FID-10 group but not the FID-40 group, relative to the control group. Hippocampal Glut1 mRNA (-39%) and GLUT1 protein (-30%) expression was suppressed in the FID-400 group, relative to the control group. Glut1 and Glut3 mRNA expression was not altered in the other brain regions in the 3 FID groups. In Study 2, hippocampal Glut1 (+14%) and Hif1α (+147%) expression was upregulated in the iron-deficient DNTfR1-/- mice, but not in the iron-replete DNTfR1-/- mice, relative to the WT mice (P < 0.05, all). CONCLUSIONS: Early-life ID is associated with altered hippocampal GLUT1 expression in adult rodents. The mouse study suggests that tissue ID is potentially responsible.


Assuntos
Transportador de Glucose Tipo 1/genética , Hipocampo/metabolismo , Deficiências de Ferro , Animais , Feminino , Transportador de Glucose Tipo 3/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores da Transferrina/fisiologia
5.
J Crohns Colitis ; 13(2): 230-244, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30272151

RESUMO

BACKGROUND AND AIMS: CD71+ erythroid cells are enriched during pregnancy with immuno suppressive properties. We investigated the frequency and functionality of CD71+ erythroid cells in peripheral blood, cord blood, and placenta of inflammatory bowel disease [IBD] patients versus healthy controls [HCs]. We aimed to determine their role in IBD pathogenesis during pregnancy. METHODS: Peripheral blood was collected at preconception, the first, second and third trimesters, and postpartum. Cord blood and placental tissues were collected at the time of birth. Cells from different specimens were subjected to immune-phenotyping and functional assays. CD71+ erythroid cells were purified for quantitative polymerase chain reaction [qPCR] analysis. Using an allogeneic mouse model of pregnancy, the effects of CD71+ erythroid cells depletion on intestinal homeostasis and dysbiosis was studied. RESULTS: IBD patients had lower CD71+ erythroid cells during pregnancy compared with HCs. Placenta and cord blood CD71+ erythroid cells from IBD patients exhibited impaired functionality and expressed lower inhibitory molecules including VISTA, TGF-ß, and reactive oxygen species [ROS]. Lower CD71+ erythroid cells were correlated with reduced regulatory T cells and increased immune-activation in IBD patients. Depletion of CD71+ erythroid cells in an allogeneic pregnancy model resulted in upregulation of TLRs, IL-6, and CXCL-1, and enhanced production of TNF-α, in intestinal tissues. In contrast, TGF-ß gene expression was reduced. Excessive inflammatory response in the gut [e.g. TNF-α] affects intestinal integrity and CD71+ erythroid cells impact on the gut's bacterial composition. CONCLUSIONS: Reduced frequency and/or impaired functionality of CD71+ erythroid cells during pregnancy may predispose IBD patients to a more pro-inflammatory milieu in their gastrointestinal tract, characterised by lower Tregs, higher IL-6, and TNF-α, and dysbiosis.


Assuntos
Antígenos CD/fisiologia , Células Eritroides/patologia , Doenças Inflamatórias Intestinais/complicações , Complicações na Gravidez/fisiopatologia , Receptores da Transferrina/fisiologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Cisplatino , Modelos Animais de Doenças , Feminino , Humanos , Ifosfamida , Doenças Inflamatórias Intestinais/sangue , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/fisiopatologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitomicina , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/patologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma
6.
Leg Med (Tokyo) ; 36: 17-20, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30312834

RESUMO

Iron is bound to mobile transferrin (TF) and ferritin in blood. TF receptors (TFRC and TFR2) regulate intracellular iron by delivering iron from TF into the cytoplasm. In this study, we examined the effects of 10 single nucleotide polymorphisms (SNPs) in each of the genes for TF and TF receptors on blood iron concentrations in Japanese subjects. Blood iron levels were determined by microwave plasma-atomic emission spectrometry and the SNPs were analyzed by polymerase chain reaction followed by restriction fragment length polymorphism analysis. Blood iron levels in males were significantly higher than those in females. Therefore, the analysis was performed only in males. Blood iron concentrations did not correlate with age and postmortem intervals in males. Among the 10 SNPs in TF, TFRC, and TFR2 genes, significant associations were observed between TF genotypes (rs12769) and male iron concentrations. Individuals with genotype GG in rs12769 had significantly higher blood iron concentrations than those with GA. Previous studies have shown the association between high tissue iron concentrations and disease, liver iron levels are higher in infants dying from sudden infant death syndrome and decreased blood iron concentrations were observed in critically ill children. Therefore, rs12769 in TF might be related to diseases and mortality risk.


Assuntos
Estudos de Associação Genética , Ferro/sangue , Polimorfismo de Nucleotídeo Único , Mudanças Depois da Morte , Receptores da Transferrina/genética , Transferrina/genética , Povo Asiático , Feminino , Genótipo , Humanos , Ferro/metabolismo , Masculino , Ligação Proteica , Receptores da Transferrina/fisiologia , Fatores de Tempo , Transferrina/metabolismo
7.
Clin Exp Immunol ; 195(3): 287-301, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30570135

RESUMO

Immunoglobulin (Ig)A is the most abundant immunoglobulin in humans, and in the airway mucosa secretory IgA (sIgA) plays a pivotal role in first-line defense against invading pathogens and antigens. IgA has been reported to also have pathogenic effects, including possible worsening of the prognosis of idiopathic pulmonary fibrosis (IPF). However, the precise effects of IgA on lung fibroblasts remain unclear, and we aimed to elucidate how IgA activates human lung fibroblasts. We found that sIgA, but not monomeric IgA (mIgA), induced interleukin (IL)-6, IL-8, monocyte chemoattractant protein (MCP)-1 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production by normal human lung fibroblasts (NHLFs) at both the protein and mRNA levels. sIgA also promoted proliferation of NHLFs and collagen gel contraction comparable to with transforming growth factor (TGF)-ß, which is involved in fibrogenesis in IPF. Also, Western blot analysis and real-time quantitative polymerase chain reaction (PCR) revealed that sIgA enhanced production of α-smooth muscle actin (α-SMA) and collagen type I (Col I) by NHLFs. Flow cytometry showed that NHLFs bound sIgA, and among the known IgA receptors, NHLFs significantly expressed CD71 (transferrin receptor). Transfection of siRNA targeting CD71 partially but significantly suppressed cytokine production by NHLFs co-cultured with sIgA. Our findings suggest that sIgA may promote human lung inflammation and fibrosis by enhancing production of inflammatory or fibrogenic cytokines as well as extracellular matrix, inducing fibroblast differentiation into myofibroblasts and promoting human lung fibroblast proliferation. sIgA's enhancement of cytokine production may be due partially to its binding to CD71 or the secretory component.


Assuntos
Citocinas/biossíntese , Imunoglobulina A Secretora/farmacologia , Pulmão/imunologia , Actinas/biossíntese , Antígenos CD/fisiologia , Células Cultivadas , Fibroblastos/imunologia , Humanos , Fibrose Pulmonar Idiopática/etiologia , Pulmão/citologia , Receptores da Transferrina/fisiologia
8.
J Biol Chem ; 292(31): 12727-12734, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28615456

RESUMO

The regulation of iron metabolism in biological systems centers on providing adequate iron for cellular function while limiting iron toxicity. Because mammals cannot excrete iron, mechanisms have evolved to control iron acquisition, storage, and distribution at both systemic and cellular levels. Hepcidin, the master regulator of iron homeostasis, controls iron flows into plasma through inhibition of the only known mammalian cellular iron exporter ferroportin. Hepcidin is feedback-regulated by iron status and strongly modulated by inflammation and erythropoietic demand. This review highlights recent advances that have changed our understanding of iron metabolism and its regulation.


Assuntos
Homeostase , Ferro/fisiologia , Modelos Biológicos , Animais , Proteínas de Transporte de Cátions/fisiologia , Eritropoese , Hepcidinas/fisiologia , Humanos , Imunidade Inata , Absorção Intestinal , Ferro/sangue , Ferro da Dieta/efeitos adversos , Ferro da Dieta/metabolismo , Fígado/fisiologia , Macrófagos/imunologia , Macrófagos/fisiologia , Estado Nutricional , Comunicação Parácrina , Receptores da Transferrina/agonistas , Receptores da Transferrina/fisiologia , Transdução de Sinais , Transferrina/fisiologia
9.
Am J Hematol ; 91(8): 812-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27169626

RESUMO

Iron metabolism and erythropoiesis are inherently interlinked physiological processes. Regulation of iron metabolism is mediated by the iron-regulatory hormone hepcidin. Hepcidin limits the amount of iron released into the blood by binding to and causing the internalization of the iron exporter, ferroportin. A number of molecules and physiological stimuli, including erythropoiesis, are known to regulate hepcidin. An increase in erythropoietic demand decreases hepcidin, resulting in increased bioavailable iron in the blood. Transferrin receptor 2 (TFR2) is involved in the systemic regulation of iron metabolism. Patients and mice with mutations in TFR2 develop hemochromatosis due to inappropriate hepcidin levels relative to body iron. Recent studies from our laboratory and others have suggested an additional role for TFR2 in response to iron-restricted erythropoiesis. These studies used mouse models with perturbed systemic iron metabolism: anemic mice lacking matriptase-2 and Tfr2, or bone marrow transplants from iron-loaded Tfr2 null mice. We developed a novel transgenic mouse model which lacks Tfr2 in the hematopoietic compartment, enabling the delineation of the role of Tfr2 in erythroid development without interfering with its role in systemic iron metabolism. We show that in the absence of hematopoietic Tfr2 immature polychromatic erythroblasts accumulate with a concordant reduction in the percentage of mature erythroid cells in the spleen and bone marrow of anemic mice. These results demonstrate that erythroid Tfr2 is essential for an appropriate erythropoietic response in iron-deficient anemia. These findings may be of relevance in clinical situations in which an immediate and efficient erythropoietic response is required. Am. J. Hematol. 91:812-818, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Anemia Ferropriva/patologia , Eritropoese , Deleção de Genes , Receptores da Transferrina/genética , Anemia Ferropriva/etiologia , Animais , Medula Óssea/patologia , Diferenciação Celular , Células Eritroides/patologia , Camundongos , Camundongos Transgênicos , Receptores da Transferrina/fisiologia , Baço/patologia
10.
PLoS One ; 10(3): e0122452, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25803700

RESUMO

The endocytosis of transferrin receptor (TfR) has served as a model to study the receptor-targeted cargo delivery system for cancer therapy for many years. To accurately evaluate and optically measure this TfR targeting delivery in vitro, a CHO cell line with enhanced green fluorescent protein (EGFP)-tagged human TfR was established. A chimera of the hTfR and EGFP was engineered by fusing EGFP to the amino terminus of hTfR. Data were provided to demonstrate that hTfR-EGFP chimera was predominantly localized on the plasma membrane with some intracellular fluorescent structures on CHO cells and the EGFP moiety did not affect the endocytosis property of hTfR. Receptor internalization occurred similarly to that of HepG2 cells expressing wild-type hTfR. The internalization percentage of this chimeric receptor was about 81 ± 3% of wild type. Time-dependent co-localization of hTfR-EGFP and PE-conjugated anti-hTfR mAb in living cells demonstrated the trafficking of mAb-receptor complexes through the endosomes followed by segregation of part of the mAb and receptor at the late stages of endocytosis. The CHO-hTfR cells preferentially took up anti-hTfR mAb conjugated nanoparticles. This CHO-hTfR cell line makes it feasible for accurate evaluation and visualization of intracellular trafficking of therapeutic agents conjugated with transferrin or Abs targeting the hTfRs.


Assuntos
Endocitose/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Receptores da Transferrina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Western Blotting , Células CHO , Cricetinae , Cricetulus , Primers do DNA/genética , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Nanopartículas/metabolismo , Plasmídeos/genética , Receptores da Transferrina/fisiologia , Estatísticas não Paramétricas
12.
Br J Haematol ; 168(6): 891-901, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25403101

RESUMO

Effective erythropoiesis requires an appropriate supply of iron and mechanisms regulating iron homeostasis and erythropoiesis are intrinsically linked. Iron dysregulation, typified by iron-deficiency anaemia and iron overload, is common in many clinical conditions and impacts the health of up to 30% of the world's population. The proteins transmembrane protease, serine 6 (TMPRSS6; also termed matriptase-2), HFE and transferrin receptor 2 (TFR2) play important and opposing roles in systemic iron homeostasis, by regulating expression of the iron regulatory hormone hepcidin. We have performed a systematic analysis of mice deficient in these three proteins and show that TMPRSS6 predominates over HFE and TFR2 in hepcidin regulation. The phenotype of mice lacking TMPRSS6 and TFR2 is characterized by severe anaemia and extramedullary haematopoiesis in the spleen. Stress erythropoiesis in these mice results in increased expression of the newly identified erythroid iron regulator erythroferrone, which does not appear to overcome the hepcidin overproduction mediated by loss of TMPRSS6. Extended analysis reveals that TFR2 plays an important role in erythroid cells, where it is involved in terminal erythroblast differentiation and the regulation of erythropoietin. In conclusion, we have identified an essential role for TFR2 in erythropoiesis that may provide new targets for the treatment of anaemia.


Assuntos
Anemia Ferropriva/sangue , Eritropoese/fisiologia , Receptores da Transferrina/fisiologia , Anemia Ferropriva/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Eritroides/patologia , Eritropoetina/biossíntese , Hematopoese Extramedular/fisiologia , Proteína da Hemocromatose , Hepcidinas/metabolismo , Antígenos de Histocompatibilidade Classe I/sangue , Antígenos de Histocompatibilidade Classe I/fisiologia , Rim/metabolismo , Fígado/metabolismo , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/deficiência , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Receptores da Eritropoetina/metabolismo , Receptores da Transferrina/sangue , Receptores da Transferrina/deficiência , Serina Endopeptidases/sangue , Serina Endopeptidases/deficiência , Serina Endopeptidases/fisiologia , Esplenomegalia/sangue
13.
Ther Deliv ; 5(5): 511-24, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24998271

RESUMO

AIM: PEGylated fluorescent nanodiamond (FND) conjugated with Tf (FND-PEG-Tf) was investigated for targeted drug delivery. MATERIALS & METHODS: Human hepatoma (HepG2) and normal (L-02) cell lines were used to investigate the difference in cellular uptake of FND-PEG-Tf and its loading drug system. Nanoparticle uptake was evaluated by flow cytometry and laser scanning confocal microscopy. RESULTS: FND-PEG-Tf showed highly specific TfR-mediated uptake by HepG2 cells, relative to negative controls (L-02 cell), which was a strong correlation among TfR density on the cell surface. The mechanism of TfR-mediated uptake was attested by free Tf with Fe³âº as a competitive agent. The difference in cell viability between L-02 and HepG2 cells treated with doxorubicin hydrochloride (DOX) nanoparticles (FND-PEG-Tf-DOX) can be explained by FND-PEG-Tf, which can target drug delivery to cancer cells. CONCLUSION: FND-PEG-Tf can potentially be utilized in targeted cancer cell imaging and effective drug delivery for cancer therapy.


Assuntos
Sistemas de Liberação de Medicamentos , Nanodiamantes/administração & dosagem , Neoplasias/tratamento farmacológico , Transferrina/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Humanos , Tamanho da Partícula , Receptores da Transferrina/fisiologia
14.
J Membr Biol ; 247(4): 291-307, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24573305

RESUMO

Current cancer management strategies fail to adequately treat malignancies with multivariable dose-restricting factors such as systemic toxicity and multi-drug resistance limiting therapeutic benefit, quality of life and complete long-term remission rates. The targeted delivery of a therapeutic compound aims to enhance its circulation and cellular uptake, decrease systemic toxicity and improve therapeutic benefit with disease specificity. The transferrin peptide, its receptor and their biological significance, has been widely characterised and vastly relevant when applied to targeting strategies. Utilising knowledge about the physiological function of the transferrin-transferrin receptor complex and the efficiency of its receptor-mediated endocytosis provides rationale to continue the development of transferrin-targeted anticancer modalities. Furthermore, multiple studies report an upregulation in expression of the transferrin receptor on metastatic and drug resistant tumours, highlighting its selectivity to cancer. Due to the increased expression of the transferrin receptor in brain glioma, the successful delivery of anticancer compounds to the tumour site and the ability to cross the blood brain barrier has shown to be an important discovery. Its significance in the development of cancer-specific therapies is shown to be important by direct conjugation and immunotoxin studies which use transferrin and anti-transferrin receptor antibodies as the targeting moiety. Such conjugates have demonstrated enhanced cellular uptake via transferrin-mediated mechanisms and increased selective cytotoxicity in a number of cancer cell lines and tumour xenograft animal models. In addition, incubation of chemotherapy-insensitive cancer cells with transferrin-targeted conjugates in vitro has resulted in a reversal of their drug resistance. Transferrin immunotoxins have also shown similar promise, with a diphtheria toxin mutant covalently bound to transferrin (Tf-CRM107) currently involved in human clinical trials for the treatment of glioblastoma. Despite this, the inability to translate preliminary research into a clinical setting has compelled research into novel targeting strategies including the use of nanoparticulate theory in the design of drug delivery systems. The main objective of this review is to evaluate the importance of the transferrin-transferrin receptor complex as a target for cancer therapy through extensive knowledge of both the physiological and pathological interactions between the complex and different cell types. In addition, this review serves as a summary to date of direct conjugation and immunotoxin studies, with an emphasis on transferrin as an important targeting moiety in the directed delivery of anticancer therapeutic compounds.


Assuntos
Antineoplásicos/uso terapêutico , Portadores de Fármacos/uso terapêutico , Endocitose , Neoplasias/tratamento farmacológico , Transferrina/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/metabolismo , Portadores de Fármacos/metabolismo , Humanos , Transporte Proteico , Receptores da Transferrina/fisiologia , Transferrina/metabolismo , Transferrina/farmacologia
15.
Am J Physiol Gastrointest Liver Physiol ; 306(2): G132-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24284962

RESUMO

Treatment for iron deficiency anemia can involve iron supplementation via dietary or parenteral routes that result in different cellular iron distributions. The effect of the administered iron on the iron regulatory system and hepcidin in the liver has not been well studied. Hepcidin, the liver-expressed central iron-regulatory peptide, is itself regulated through the bone morphogenetic protein (BMP)/SMAD signaling pathway. Specifically, Bmp6 expression is upregulated in response to iron and induces hepcidin through phosphorylation of Smad1/5/8. The hemochromatosis-associated proteins Hfe and transferrin receptor 2 (Tfr2) are known upstream regulators of hepcidin, although their precise roles are still unclear. To investigate the mechanisms of this regulation and the roles of the Hfe and Tfr2, we subjected wild-type, Hfe(-/-), Tfr2(-/-), and Hfe(-/-)/Tfr2(-/-) mice to iron loading via dietary or parenteral routes. Systematic analysis demonstrated that Tfr2 is required for effective upregulation of Bmp6 in response to hepatocyte iron, but not nonparenchymal iron. Hfe is not required for Bmp6 upregulation, regardless of iron localization, but rather, is required for efficient downstream transmission of the regulatory signal. Our results demonstrate that Hfe and Tfr2 play separate roles in the regulatory responses to iron compartmentalized in different cell types and further elucidates the regulatory mechanisms controlling iron homeostasis.


Assuntos
Hepcidinas/fisiologia , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/fisiologia , Ferro/administração & dosagem , Ferro/farmacologia , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Receptores da Transferrina/genética , Receptores da Transferrina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Administração Oral , Animais , Western Blotting , Proteína Morfogenética Óssea 6/metabolismo , Corantes , Proteína da Hemocromatose , Hepcidinas/genética , Infusões Parenterais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Smad/metabolismo
16.
Eur J Drug Metab Pharmacokinet ; 39(4): 301-10, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24363124

RESUMO

The extended use of doxorubicin (DOX) could be limited due to the emergence of drug resistance and cardiotoxicity associated with its treatment. Conjugates of DOX with transferrin (DOX-TRF) can effectively alleviate these side effects, thereby leading to a better treatment. The effectiveness of DOX-TRF could result from the enhancement of transferrin receptor (TfR)-mediated transportation. However, detailed TfR-mediated DOX delivery has not been fully elucidated thus far, which may rely on the quantitative subcellular study of DOX distribution and metabolism. In this study, an immunoisolation assay was developed to isolate the organelles with high purity, yield and integrity. Using this immunoisolation assay together with liquid chromatography-tandem mass spectrometry (LC/MS/MS), the subcellular distribution profiles of DOX and its main metabolite doxorubicinol (DOXol) in human breast cancer cells MCF-7/WT and MCF-7/ADR were determined and compared after the treatment of DOX and DOX-TRF. As expected, DOX-TRF treated cells have a higher drug accumulation compared to DOX treated cells. DOX-TRF was predominantly cytoplasmic. In addition, TfR-mediated transportation had a significant impact on the transformation of DOX to DOXol in the cells. This study provided the evidence that immunoisolation together with LC/MS/MS is an effective technique in subcellular investigations.


Assuntos
Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Receptores da Transferrina/fisiologia , Cromatografia Líquida , Feminino , Humanos , Separação Imunomagnética , Células MCF-7 , Espectrometria de Massas em Tandem , Distribuição Tecidual
17.
Curr Aging Sci ; 6(1): 21-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23895519

RESUMO

Advanced age is associated with an increased incidence of immune and degenerative disorders, mediated by metabolic changes, dysregulation of proinflammatory signals, and apoptosis. Concurrently, there is a progressive decline in self-recognition. Investigations on biologic functions of transferrin (Tf) other than iron transport showed that Tf has a profound cytoprotective (anti-apoptotic) effect on lympho-hematopoietic cells and the thymus, and interferes with stress-induced signals. Tf protects hepatocytes against Fas-induced cell death by reducing BID cleavage, inhibiting caspase-3 and -9 activation and up-regulating survival signals such as Bcl-xL. The involvement in the regulation of alloreactivity and apoptosis suggests that Tf participates in the maintenance of "self-identity" mechanisms, which are tightly linked to the capacity of the immune system to recognize and react against any noxious agent. Some of the disorders associated with aging are thought to be related to thymic involution, reflecting alterations in the interplay of neural, endocrine and immune factors. We established a murine model of thymic involution induced by stereotactically placed electrolytic lesions in the anterior hypothalamic area. The events observed in this model mimic those observed during senescence including thymus involution, i.e. enhanced glucocorticoid reaction to distress, and obesity. The described properties of Tf can be exploited to modify immune responses and provide cytoprotection against pro-apoptotic and cytotoxic signals when neuroimmunomodulatory mechanisms are impaired, as is the case with aging.


Assuntos
Envelhecimento/imunologia , Envelhecimento/fisiologia , Neuroimunomodulação , Transferrina/imunologia , Transferrina/fisiologia , Envelhecimento/patologia , Animais , Apoptose , Citocinas/metabolismo , Humanos , Hipotálamo/imunologia , Hipotálamo/patologia , Hipotálamo/fisiologia , Mediadores da Inflamação/metabolismo , Fígado/imunologia , Fígado/patologia , Fígado/fisiologia , Camundongos , Modelos Imunológicos , Modelos Neurológicos , Receptores da Transferrina/fisiologia , Tolerância a Antígenos Próprios , Transdução de Sinais , Timo/imunologia , Timo/patologia , Timo/fisiologia
18.
Crit Rev Oncog ; 18(5): 463-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23879590

RESUMO

Anemia is a frequent finding of Hodgkin's lymphoma (HL) diagnosis. It is usually mild, with hemoglobin levels between 10 and 12 g/dl; it is rarely (<10% of cases) a result of bone marrow infiltration; and it displays the characteristics of the anemia of chronic disease due to abnormalities in iron metabolism. The inflammatory cytokine IL-6 is frequently up-regulated in Hodgkin's lymphoma, and IL-6 levels are strongly associated with hepcidin, the main regulator of iron metabolism. Elevated hepcidin levels result in iron restriction and signs of anemia of chronic disease. In addition, the abundant microenvironment surrounding the neoplastic Hodgkin's and Reed-Sternberg cells may contribute to alterations in iron metabolism. Tumor-infiltrating macrophages can sequester iron using scavenger receptors. Iron-restricted anemia at HL diagnosis can be aggravated by intensive chemotherapy, and iron overload may become clinically relevant in heavily treated patients with relapsed or refractory disease undergoing high-dose chemotherapy and stem cell transplantation.


Assuntos
Doença de Hodgkin/metabolismo , Ferro/metabolismo , Anemia/etiologia , Anemia/metabolismo , Animais , Microambiente Celular , Ferritinas/sangue , Transplante de Células-Tronco Hematopoéticas/mortalidade , Hepcidinas/fisiologia , Doença de Hodgkin/etiologia , Doença de Hodgkin/terapia , Humanos , Sobrecarga de Ferro/sangue , Receptores da Transferrina/fisiologia
19.
Biochim Biophys Acta ; 1830(8): 4254-64, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23648413

RESUMO

BACKGROUND: Targeting nanoobjects via the iron-acquisition pathway is always reported slower than the transferrin/receptor endocytosis. Is there a remedy? METHODS: Maghemite superparamagnetic and theragnostic nanoparticles (diameter 8.6nm) were synthesized, coated with 3-aminopropyltriethoxysilane (NP) and coupled to four holotransferrin (TFe2) by amide bonds (TFe2-NP). The constructs were characterized by X-ray diffraction, transmission electron microscopy, FTIR, X-ray Electron Spectroscopy, Inductively Coupled Plasma with Atomic Emission Spectrometry. The in-vitro protein/protein interaction of TFe2-NP with transferrin receptor-1 (R1) and endocytosis in HeLa cells were investigated spectrophotometrically, by fast T-jump kinetics and confocal microscopy. RESULTS: In-vitro, R1 interacts with TFe2-NP with an overall dissociation constant KD=11nM. This interaction occurs in two steps: in the first, the C-lobe of the TFe2-NP interacts with R1 in 50µs: second-order rate constant, k1=6×10(10)M(-1)s(-1); first-order rate constant, k-1=9×10(4)s(-1); dissociation constant, K1d=1.5µM. In the second step, the protein/protein adduct undergoes a slow (10,000s) change in conformation to reach equilibrium. This mechanism is identical to that occurring with the free TFe2. In HeLa cells, TFe2-NP is internalized in the cytosol in less than 15min. CONCLUSION: This is the first time that a nanoparticle-transferrin construct is shown to interact with R1 and is internalized in time scales similar to those of the free holotransferrin. GENERAL SIGNIFICANCE: TFe2-NP behaves as free TFe2 and constitutes a model for rapidly targeting theragnostic devices via the main iron-acquisition pathway.


Assuntos
Antígenos CD/fisiologia , Compostos Férricos/metabolismo , Ferro/metabolismo , Nanopartículas , Receptores da Transferrina/fisiologia , Termodinâmica , Transferrina/metabolismo , Compostos Férricos/administração & dosagem , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Microscopia Confocal , Transferrina/administração & dosagem , Difração de Raios X
20.
Cell Death Differ ; 20(8): 1101-15, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23686137

RESUMO

Mismanaged protein trafficking by the proteostasis network contributes to several conformational diseases, including cystic fibrosis, the most frequent lethal inherited disease in Caucasians. Proteostasis regulators, as cystamine, enable the beneficial action of cystic fibrosis transmembrane conductance regulator (CFTR) potentiators in ΔF508-CFTR airways beyond drug washout. Here we tested the hypothesis that functional CFTR protein can sustain its own plasma membrane (PM) stability. Depletion or inhibition of wild-type CFTR present in bronchial epithelial cells reduced the availability of the small GTPase Rab5 by causing Rab5 sequestration within the detergent-insoluble protein fraction together with its accumulation in aggresomes. CFTR depletion decreased the recruitment of the Rab5 effector early endosome antigen 1 to endosomes, thus reducing the local generation of phosphatidylinositol-3-phosphate. This diverts recycling of surface proteins, including transferrin receptor and CFTR itself. Inhibiting CFTR function also resulted in its ubiquitination and interaction with SQSTM1/p62 at the PM, favoring its disposal. Addition of cystamine prevented the recycling defect of CFTR by enhancing BECN1 expression and reducing SQSTM1 accumulation. Our results unravel an unexpected link between CFTR protein and function, the latter regulating the levels of CFTR surface expression in a positive feed-forward loop, and highlight CFTR as a pivot of proteostasis in bronchial epithelial cells.


Assuntos
Brônquios/fisiopatologia , Membrana Celular/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Fibrose Cística/fisiopatologia , Células Epiteliais/fisiologia , Deficiências na Proteostase/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Proteína Beclina-1 , Brônquios/patologia , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/patologia , Humanos , Proteínas de Membrana/fisiologia , Mutação/genética , Monoéster Fosfórico Hidrolases/fisiologia , Receptores da Transferrina/fisiologia , Proteína Sequestossoma-1 , Proteínas rab5 de Ligação ao GTP/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA