Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Signal ; 118: 111148, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38521179

RESUMEN

Hepatocellular carcinoma (HCC) is the major form of liver malignancy with high incidence and mortality. Identifying novel biomarkers and understanding regulatory mechanisms underlying the development and progression of HCC are critical for improving diagnosis, treatment and patient outcomes. Carboxyl terminus of Hsc-70-interacting protein (CHIP) is a well-described U-box-type E3 ubiquitin ligase which promotes the ubiquitination and degradation of numerous tumor-associated proteins. Recent studies have shown that CHIP can play as a tumor-suppressor gene or an oncogene in different kinds of malignancies. To date, the function and mechanism of CHIP in hepatocellular carcinoma remains largely unknown. Based on TCGA data, we found that compared with high CHIP expression, the overall survival of HCC patients with low expression of CHIP was better. In addition, CHIP overexpression markedly enhanced HCC cell proliferation and colony formation. Conversely, knockdown of CHIP restrained the proliferation and colony formation of HCC cells. Meanwhile, knockdown of CHIP decreased mitochondrial cristae or ruptured outer mitochondrial membrane, promoted the accumulation of Fe2+ and ferroptosis of HCC cells. Further research for the first time confirmed that CHIP interacts and degrades transferrin receptor 1 (TfR1) by ubiquitin-proteasome pathway, which leads to the inhibition of ferroptosis and promotes the proliferation of HCC cells. The analysis of proteomics data from CPTAC revealed a negative correlation between CHIP and TfR1 protein expression levels in HCC. These findings indicate that CHIP acts as a negative modulator of ferroptosis and functions as an oncogene in HCC.


Asunto(s)
Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Hepáticas/patología , Receptores de Transferrina , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
Arthritis Res Ther ; 26(1): 71, 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38493104

RESUMEN

OBJECTIVE: Transferrin receptor-1 (TfR1) plays important roles in controlling cellular iron levels, but its role in OA pathology is unknown. Herein we aim to investigate the role of TfR1 in OA progression and its underlying mechanisms. METHODS: TfR1 expression in cartilage during OA development were examined both in vivo and in vitro. Then IL-1ß was used to induce chondrocytes degeneration in vitro and TfR1 siRNA was used for observing the effect of TfR1 in modulating iron homeostasis, mitochondrial function and degrading enzymes expression. Also the inhibitor of TfR1 was exploited to analyze the protective effect of TfR1 inhibition in vivo. RESULTS: TfR1 is elevated in OA cartilage and contributes to OA inflammation condition. Excess iron not only results in oxidative stress damage and sensitizes chondrocytes to ferroptosis, but also triggers c-GAS/STING-mediated inflammation by promoting mitochondrial destruction and the release of mtDNA. Silencing TfR1 using TfR1 siRNA not only reduced iron content in chondrocytes and inhibited oxidative stress, but also facilitated the mitophagy process and suppressed mtDNA/cGAS/STING-mediated inflammation. Importantly, we also found that Ferstatin II, a novel and selective TfR1 inhibitor, could substantially suppress TfR1 activity both in vivo and in vitro and ameliorated cartilage degeneration. CONCLUSION: Our work demonstrates that TfR1 mediated iron influx plays important roles in chondrocytes degeneration and OA pathogenesis, suggesting that maintaining iron homeostasis through the targeting of TfR1 may represent a novel therapeutic strategy for the treatment of OA.


Asunto(s)
Osteoartritis , Humanos , Osteoartritis/metabolismo , Cartílago/metabolismo , Inflamación/patología , Condrocitos/metabolismo , ADN Mitocondrial , ARN Interferente Pequeño/metabolismo
3.
Nat Prod Res ; 38(4): 673-678, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-36855296

RESUMEN

Norcantharidin (NCTD) is a demethylated analogue of cantharidin. It was recently demonstrated that NCTD reduces iron contents in the liver and spleen of mice in vivo, indicating that NCTD can affect iron metabolism via hepcidin. Here, we investigated the effects of NCTD on expression of iron storage protein ferritin-light chain (Ft-L), transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), ferroportin 1 (Fpn1), hepcidin, iron regulatory protein 1 (IRP1), IL-6, p-JAK2 and p-STAT3 in lipopolysaccharides (LPS)-treated RAW264.7 cells in vitro via Real-time PCR and Western blotting analysis. We demonstrate that NCTD down-regulates Ft-L, hepcidin, IL-6, pJAK2, pSTAT3 and up-regulates TfR1, DMT1, Fpn1 and IRP1 expression in LPS treated cells, showing that NCTD can inhibit hepcidin via the IL-6/JAK2/STAT3 signalling pathway and also increase TfR1, DMT1 and Fpn1 expression via down-regulating hepcidin and up-regulating IRP1. Our findings provide further evidence in vitro for the role of NCTD in iron metabolism.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes , Hepcidinas , Interleucina-6 , Ratones , Animales , Hepcidinas/genética , Hepcidinas/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Regulación hacia Abajo , Lipopolisacáridos/farmacología , Hierro/metabolismo , Macrófagos/metabolismo
4.
Pharmaceutics ; 15(3)2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36986599

RESUMEN

Since the delivery of biologic drugs to the brain is greatly hampered by the existence of the blood-brain barrier (BBB), brain shuttles are being developed to enhance therapeutic efficacy. As we have previously shown, efficient and selective brain delivery was achieved with TXB2, a cross-species reactive, anti-TfR1 VNAR antibody. To further explore the limits of brain penetration, we conducted restricted randomization of the CDR3 loop, followed by phage display to identify improved TXB2 variants. The variants were screened for brain penetration in mice using a 25 nmol/kg (1.875 mg/kg) dose and a single 18 h timepoint. A higher kinetic association rate to TfR1 correlated with improved brain penetration in vivo. The most potent variant, TXB4, showed a 3.6-fold improvement over TXB2, which had on average 14-fold higher brain levels when compared to an isotype control. Like TXB2, TXB4 retained brain specificity with parenchymal penetration and no accumulation in other organs. When fused with a neurotensin (NT) payload, it led to a rapid drop in body temperature upon transport across the BBB. We also showed that fusion of TXB4 to four therapeutic antibodies (anti-CD20, anti-EGFRvIII, anti-PD-L1 and anti-BACE1) improved their brain exposure between 14- to 30-fold. In summary, we enhanced the potency of parental TXB2 brain shuttle and gained a critical mechanistic understanding of brain delivery mediated by the VNAR anti-TfR1 antibody.

5.
Brain Sci ; 12(10)2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36291201

RESUMEN

Depression has become one of the severe mental disorders threatening global human health. In this study, we first used the proteomics approach to obtain the differentially expressed proteins in the liver between naive control and chronic social defeat stress (CSDS) induced depressed mice. We have identified the upregulation of iron binding protein transferrin (TF) in the liver, the peripheral blood, and the brain in CSDS-exposed mice. Furthermore, bioinformatics analysis of the Gene Expression Omnibus (GEO) database from various mouse models of depression revealed the significantly upregulated transcripts of TF and its receptor TfR1 in multiple brain regions in depressed mice. We also used the recombinant TF administration via the tail vein to detect its permeability through the blood-brain barrier (BBB). We demonstrated the permeability of peripheral TF into the brain through the BBB. Together, these results identified the elevated expression of TF and its receptor TfR1 in both peripheral liver and the central brain in CSDS-induced depressed mice, and peripheral administration of TF can be transported into the brain through the BBB. Therefore, our data provide a compelling information for understanding the potential role and mechanisms of the cross-talk between the liver and the brain in stress-induced depression.

6.
Am J Transl Res ; 14(7): 4562-4572, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35958455

RESUMEN

OBJECTIVE: To analyze the pathological features of patients with osteosarcoma and the correlation of expressions of transferrin receptor 1 (TfR1), vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP)-9 in tumor tissue with the pathological features. METHODS: The tumor tissue and paracancerous tissue samples from 31 patients with osteosarcoma were collected before treatment to measure the expressions of TfR1, VEGF and MMP-9. Kaplan-meier curve was used to analyze the relationship of TfR1, VEGF and MMP-9 with the survival time of patients. Log-rank test was used to test the difference, and Spearman test was used to test the correlation. RESULTS: Among the 31 osteosarcoma samples, 23 (74.19%), 24 (77.42%) and 17 (54.84%) samples showed medium-high expressions of VEGF, TfR1 and MMP-9, respectively, which were significantly higher than those in the paracancerous samples (P<0.05). Furthemore, the medium-high expression rates of VEGF, TfR1 and MMP-9 were significantly different in patients with different Enneking stages, different histological differentiation degrees, and with or without distant metastasis. Besides, the expression of VEGF was also significantly different in different-size osteosarcoma samples (P<0.05). The survival time of patients with low expressions of TfR1, VEGF and MMP-9 was significantly longer than that of patients with medium-high expressions (P<0.05). Additionally, in osteosarcoma samples, significant positive correlations were shown between the expressions of TfR1 and VEGF, as well as between TfR1 and MMP-9 (P<0.05), but there was no significant correlation between VEGF and MMP-9 (P>0.05). CONCLUSION: TfR1, VEGF and MMP-9 are abnormally expressed in osteosarcoma lesions, suggesting that they all play a role in the occurrence and development of osteosarcoma.

7.
Pharmaceutics ; 14(7)2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35890231

RESUMEN

Single domain shark antibodies that bind to the transferrin receptor 1 (TfR1) on brain endothelial cells have been used to shuttle antibodies and other cargos across the blood brain barrier (BBB) to the brain. For these studies the TXB4 brain shuttle was fused to a TrkB neurotrophin receptor agonist antibody. The TXB4-TrkB fusion retained potent agonist activity at its cognate receptor and after systemic administration showed a 12-fold increase in brain levels over the unmodified antibody. Only the TXB4-TrkB antibody fusion was detected within the brain and localized to TrkB positive cells in the cortex and tyrosine hydroxylase (TH) positive dopaminergic neurons in the substantia nigra pars compacta (SNc), where it was associated with activated ERK1/2 signaling. When tested in the 6-hydroxydopamine (6-OHDA) mouse model of Parkinson's disease (PD), TXB4-TrkB, but not the unmodified antibody, completely prevented the 6-OHDA induced death of TH positive neurons in the SNc. In conclusion, the fusion of the TXB4 brain shuttle allows a TrkB agonist antibody to reach neuroprotective concentrations in the brain parenchyma following systemic administration.

8.
Cancers (Basel) ; 15(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36612268

RESUMEN

PURPOSE: To identify a molecular signature of macrophages exposed to clinically relevant ionizing radiation (IR) doses, mirroring radiotherapy sessions. METHODS: Human monocyte-derived macrophages were exposed to 2 Gy/ fraction/ day for 5 days, mimicking one week of cancer patient's radiotherapy. Protein expression profile by proteomics was performed. RESULTS: A gene ontology analysis revealed that radiation-induced protein changes are associated with metabolic alterations, which were further supported by a reduction of both cellular ATP levels and glucose uptake. Most of the radiation-induced deregulated targets exhibited a decreased expression, as was the case of cathepsin D, a lysosomal protease associated with cell death, which was validated by Western blot. We also found that irradiated macrophages exhibited an increased expression of the transferrin receptor 1 (TfR1), which is responsible for the uptake of transferrin-bound iron. TfR1 upregulation was also found in tumor-associated mouse macrophages upon tumor irradiation. In vitro irradiated macrophages also presented a trend for increased divalent metal transporter 1 (DMT1), which transports iron from the endosome to the cytosol, and a significant increase in iron release. CONCLUSIONS: Irradiated macrophages present lower ATP levels and glucose uptake, and exhibit decreased cathepsin D expression, while increasing TfR1 expression and altering iron metabolism.

9.
Redox Biol ; 40: 101865, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33493903

RESUMEN

Association of both iron/hepcidin and apolipoprotein E (ApoE) with development of Alzheimer disease (AD) and atherosclerosis led us to hypothesize that ApoE might be required for body iron homeostasis. Here, we demonstrated that ApoE knock-out (KO) induced a progressive accumulation of iron with age in the liver and spleen of mice. Subsequent investigations showed that the increased iron in the liver and spleen was due to phosphorylated extracellular regulated protein kinases (pERK) mediated up-regulation of transferrin receptor 1 (TfR1), and nuclear factor erythroid 2-related factor-2 (Nrf2)-dependent down-regulation of ferroportin 1. Furthermore, replenishment of ApoE could partially reverse the iron-related phenotype in ApoE KO mice. The findings imply that ApoE may be essential for body iron homeostasis and also suggest that clinical late-onset diseases with unexplained iron abnormality may partly be related to deficiency or reduced expression of ApoE.


Asunto(s)
Proteínas de Transporte de Catión , Sobrecarga de Hierro , Animales , Apolipoproteínas E/genética , Proteínas de Transporte de Catión/genética , Hepcidinas , Hierro/metabolismo , Ratones , Ratones Noqueados , Receptores de Transferrina/genética
10.
FASEB J ; 35(2): e21172, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33241587

RESUMEN

Transfer across the blood-brain barrier (BBB) remains a significant hurdle for the development of biopharmaceuticals with therapeutic effects within the central nervous system. We established a functional selection method to identify high affinity single domain antibodies to the transferrin receptor 1 (TfR1) with efficient biotherapeutic delivery across the BBB. A synthetic phage display library based on the variable domain of new antigen receptor (VNAR) was used for in vitro selection against recombinant human TfR1 ectodomain (rh-TfR1-ECD) followed by in vivo selection in mouse for brain parenchyma penetrating antibodies. TXB2 VNAR was identified as a high affinity, species cross-reactive VNAR antibody against TfR1-ECD that does not compete with transferrin or ferritin for receptor binding. IV dosing of TXB2 when fused to human Fc domain (TXB2-hFc) at 25 nmol/kg (1.875 mg/kg) in mice resulted in rapid binding to brain capillaries with subsequent transport into the brain parenchyma and specific uptake into TfR1-positive neurons. Likewise, IV dosing of TXB2-hFc fused with neurotensin (TXB2-hFc-NT) at 25 nmol/kg resulted in a rapid and reversible pharmacological response as measured by body temperature reduction. TXB2-hFc did not elicit any acute adverse reactions, bind, or deplete circulating reticulocytes or reduce BBB-expressed endogenous TfR1 in mice. There was no evidence of target-mediated clearance or accumulation in peripheral organs except lung. In conclusion, TXB2 is a high affinity, species cross-reactive, and brain-selective VNAR antibody to TfR1 that rapidly crosses the BBB and exhibits a favorable pharmacokinetic and safety profile and can be readily adapted to carry a wide variety of biotherapeutics from blood to brain.


Asunto(s)
Afinidad de Anticuerpos , Antígenos CD/inmunología , Transporte Biológico/inmunología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Receptores de Transferrina/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Bacteriófagos/inmunología , Transporte Biológico/genética , Reacciones Cruzadas , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Receptores de Antígenos/inmunología , Receptores de Antígenos/metabolismo , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Anticuerpos de Cadena Única/farmacocinética , Transfección
11.
Biosci Rep ; 40(4)2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32186328

RESUMEN

In the light of hepatocyte growth factor (HGF) the inhibiting role on the expression of hepcidin, we hypothesized that HGF might be able to reduce cell and tissue iron by increasing ferroportin 1 (Fpn1) content and Fpn1-mediated iron release from cells and tissues. The hypothesized ability of HGF to reduce iron might be one of the mechanisms associated with its neuroprotective action under the conditions of ischemia/reperfusion (I/R). Here, we investigated the effects of HGF on the expression of hepcidin as well as transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), Fpn1, ferritin and iron regulatory proteins (IRPs) in oxygen-glucose deprivation and reoxygenation (OGD/R)-treated PC12 cells by real-time PCR and Western blot analysis. We demonstrated that HGF could completely reverse the OGD/R-induced reduction in Fpn1 and IRP1 expression and increase in ferritin light chain protein and hepcidin mRNA levels in PC12 cells. It was concluded that HGF protects PC12 cells against OGD/R-induced injury mainly by reducing cell iron contents via the up-regulation of Fpn1 and increased Fpn1-mediated iron export from cells. Our findings suggested that HGF may also be able to ameliorate OGD/R or I/R-induced overloading of brain iron by promoting Fpn1 expression.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Proteínas de Transporte de Catión/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Hierro/metabolismo , Daño por Reperfusión/prevención & control , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Hipoxia de la Célula , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/uso terapéutico , Hepcidinas/metabolismo , Humanos , Hierro/análisis , Proteína 1 Reguladora de Hierro/metabolismo , Células PC12 , Ratas , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Daño por Reperfusión/patología , Regulación hacia Arriba/efectos de los fármacos
12.
J Biol Chem ; 295(7): 2068-2083, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31915245

RESUMEN

Many secretory proteins are activated by cleavage at specific sites. The proprotein convertases (PCs) form a family of nine secretory subtilisin-like serine proteases, seven of which cleave at specific basic residues within the trans-Golgi network, granules, or at the cell surface/endosomes. The seventh member, PC7, is a type-I transmembrane (TM) protein with a 97-residue-long cytosolic tail (CT). PC7 sheds human transferrin receptor 1 (hTfR1) into soluble shTfR1 in endosomes. To better understand the physiological roles of PC7, here we focused on the relationship between the CT-regulated trafficking of PC7 and its ability to shed hTfR1. Deletion of the TMCT resulted in soluble PC7 and loss of its hTfR1 shedding activity. Extensive CT deletions and mutagenesis analyses helped us zoom in on three residues in the CT, namely Glu-719, Glu-721, and Leu-725, that are part of a novel motif, EXEXXXL725, critical for PC7 activity on hTfR1. NMR studies of two 14-mer peptides mimicking this region of the CT and its Ala variants revealed that the three exposed residues are on the same side of the molecule. This led to the identification of adaptor protein 2 (AP-2) as a protein that recognizes the EXEXXXL725 motif, thus representing a potentially new regulator of PC7 trafficking and cleavage activity. Immunocytochemistry of the subcellular localization of PC7 and its Ala variants of Leu-725 and Glu-719 and Glu-721 revealed that Leu-725 enhances PC7 localization to early endosomes and that, together with Glu-719 and Glu-721, it increases the endosomal activity of PC7 on hTfR1.


Asunto(s)
Antígenos CD/genética , Citosol/metabolismo , Transporte de Proteínas/genética , Receptores de Transferrina/genética , Subtilisinas/genética , Factor de Transcripción AP-2/genética , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos/genética , Antígenos CD/química , Membrana Celular/genética , Movimiento Celular/genética , Citosol/química , Endosomas/genética , Células HEK293 , Humanos , Receptores de Transferrina/química , Subtilisinas/química , Red trans-Golgi/genética
13.
Eur J Nucl Med Mol Imaging ; 46(13): 2848-2858, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31342134

RESUMEN

Almost 50 million people worldwide are affected by Alzheimer's disease (AD), the most common neurodegenerative disorder. Development of disease-modifying therapies would benefit from reliable, non-invasive positron emission tomography (PET) biomarkers for early diagnosis, monitoring of disease progression, and assessment of therapeutic effects. Traditionally, PET ligands have been based on small molecules that, with the right properties, can penetrate the blood-brain barrier (BBB) and visualize targets in the brain. Recently a new class of PET ligands based on antibodies have emerged, mainly in applications related to cancer. While antibodies have advantages such as high specificity and affinity, their passage across the BBB is limited. Thus, to be used as brain PET ligands, antibodies need to be modified for active transport into the brain. Here, we review the development of radioligands based on antibodies for visualization of intrabrain targets. We focus on antibodies modified into a bispecific format, with the capacity to undergo transferrin receptor 1 (TfR1)-mediated transcytosis to enter the brain and access pathological proteins, e.g. amyloid-beta. A number of such antibody ligands have been developed, displaying differences in brain uptake, pharmacokinetics, and ability to bind and visualize the target in the brain of transgenic mice. Potential pathological changes related to neurodegeneration, e.g. misfolded proteins and neuroinflammation, are suggested as future targets for this novel type of radioligand. Challenges are also discussed, such as the temporal match of radionuclide half-life with the ligand's pharmacokinetic profile and translation to human use. In conclusion, brain PET imaging using bispecific antibodies, modified for receptor-mediated transcytosis across the BBB, is a promising method for specifically visualizing molecules in the brain that are difficult to target with traditional small molecule ligands.


Asunto(s)
Anticuerpos/genética , Encéfalo/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Ingeniería de Proteínas , Animales , Anticuerpos/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Humanos , Transporte de Proteínas
14.
Cell Mol Biol Lett ; 24: 31, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31131011

RESUMEN

BACKGROUND: While microRNAs (miRNAs) are known to play a critical role in the progression of colorectal cancer, the role of miR-107 remains unknown. We evaluated its role and explored the underlying mechanism. MATERIALS & METHODS: MTT, wound-healing, transwell migration and transwell invasion assays were performed to evaluate the role of miR-107 in SW629 cell proliferation, migration and invasion. Real time-PCR and dual-luciferase reporter gene, TFR1 overexpression and western blotting assays were used to explore the underlying mechanism. RESULTS: MiR-107 is downregulated in colorectal cancer tissues and several human colorectal cancer cell lines. Low miR-107 expression often indicates a poor survival rate for colorectal cancer patients. MiR-107 suppresses the proliferation, migration and invasion of SW620 cells by negatively regulating transferrin receptor 1 (TFR1). CONCLUSION: MiR-107 suppresses the metastasis of colorectal cancer and could be a potential therapy target in colorectal cancer patients.


Asunto(s)
Neoplasias Colorrectales/genética , Genes Supresores de Tumor , MicroARNs/genética , Receptores de Transferrina/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Invasividad Neoplásica , Regulación hacia Arriba/genética
15.
Neurosci Lett ; 662: 1-5, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28987816

RESUMEN

To understand the potential mechanisms involved in the beneficial effects of aspirin (ASA) in mood disorders, Alzheimer's (AD) and Parkinson's disease (PD), we investigated the effects of ASA on the expression of iron transport proteins transferrin receptor 1 (TfR1), ferroportin 1 (Fpn1), and iron storage protein ferritin light chain (Ft-L) in interleukin-6 (IL-6)-treated PC-12 cells. We demonstrated that IL-6 alone could induce a severe decline in Fpn1 expression and cell viability, and an increase in Ft-L protein, while ASA could markedly diminish the effects of IL-6 on these parameters. We also found that IL-6 significantly increased hepcidin expression and janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) phosphorylation, while ASA also observably suppressed these IL-6-induced effects. The data imply that ASA increases Fpn1 expression by inhibiting hepcidin expression via the IL-6/JAK/STAT3 pathway and show that the reduced content of Ft-L is due to the increased Fpn1 and subsequent iron release in the cells. The reduction of iron in neuronal cells by the increased expression of Fpn1 might be partly associated with the beneficial effects of ASA on mood disorders, AD and PD.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Proteínas de Transporte de Catión/metabolismo , Hepcidinas/antagonistas & inhibidores , Interleucina-6/farmacología , Quinasas Janus/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Células PC12 , Fosforilación , Ratas , Transducción de Señal
16.
Neurochem Int ; 91: 72-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26522688

RESUMEN

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.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Hierro/metabolismo , Microglía/metabolismo , Receptores de Transferrina/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Citocinas/biosíntesis , Ferritinas/biosíntesis , Hepcidinas/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
17.
Neuropeptides ; 48(3): 161-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24786977

RESUMEN

The existence of all components of the renin-angiotensin system (RAS) and the iron metabolism system, and the recent findings on the functions of angiotensin II (ANGII) in peripheral iron metabolism imply that ANGII might play a role in iron homeostasis by regulating expression of iron transport proteins in the brain. Here, we investigated effects of ANGII on uptake and release of iron as well as expression of cell iron transport proteins in cultured astrocytes. We demonstrated that ANGII could significantly inhibit transferrin-bound iron (Tf-Fe) uptake and iron release as well as the expression of transferrin receptor 1 (TfR1) and the iron exporter ferroportin 1 (Fpn1) in cultured astrocytes. This indicated that the inhibitory role of ANGII on Tf-Fe uptake and iron release is mediated by its negative effect on the expression of TfR1 and Fpn1. We also provided evidence that ANGII had no effect on divalent metal transporter 1 (DMT1) expression as well as non-transferrin-bound iron (NTBI) uptake in the cells. Our findings showed that ANGII has a role to affect expression of iron transport proteins in astrocytes in vitro and also suggested that ANGII might have a physiological function in brain iron homeostasis.


Asunto(s)
Angiotensina II/farmacología , Astrocitos/metabolismo , Hierro/metabolismo , Transferrina/metabolismo , Animales , Astrocitos/efectos de los fármacos , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA