Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38658189

RESUMEN

Iron is a fundamental element for biological life, starting from bacteria till humans. Iron is essential for cell function and survival, energy production and metabolism, whereas increased levels cause oxidative stress. It is also a constituent of haemoglobin and thus it is necessary for oxygen transportation through the body. Given these multiple functions, the regulation of iron metabolism is complex and tight coupled with oxygen homeostasis at tissue and cellular levels, thanks to the interaction with the hypoxia inducible factor (HIF) system. In patients with chronic kidney disease (CKD), iron deficiency significantly contributes to anaemia development. This frequently overlaps with chronic inflammation, causing iron- restricted erythropoiesis. To add further complexity, metabolic hyperferritinemia may, on one side, increase the risk for CKD and, on the other, overlaps with functional iron deficiency. Excessive intracellular iron in certain cell types during CKD can also mediate cellular death (called ferroptosis), and contribute to the pathogenesis of kidney damage, atherosclerosis and vascular calcifications. This review is aimed at broadening the perspective of iron metabolism in the setting of CKD not just as a contributor to anaemia in CKD patients, but also as an important player with an impact on cell metabolism, renal fibrosis, and the cardiovascular system.

2.
Mol Cell Neurosci ; 111: 103589, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33422672

RESUMEN

Iron has a key role in the activation of the autophagic pathway in rats with intracerebral hemorrhage (ICH), and hepcidin has the ability to reduce brain iron in ICH-rats. We therefore hypothesized that hepcidin might be able to inhibit autophagy by reducing iron in an ICH brain. Here, we investigated the effects of Ad-hepcidin and/or hepcidin peptide on autophagic activities in ICH models in vitro and in vivo. We demonstrated that ad-hepcidin and hepcidin peptide both inhibited hemin-induced increase in LC3-II/LC3-I conversion ratio and reversed the reduction in p62 content in cortical neurons in vitro. We also showed that ad-hepcidin inhibited ICH-induced increase in LC3-II/LC3-I conversion ratio and reversed ICH-induced reduction in p62 content in the brain cortex of rats in vivo. Based on these findings plus previous data on the effects of ad-hepcidin and/or hepcidin peptide on iron contents in ICH models, we suggested that hepcidin-induced inhibition of autophagy might be mediated via reducing iron in hemin-treated neurons in vitro and ICH-rat brain in vivo.


Asunto(s)
Autofagia , Hemorragia Cerebral/metabolismo , Hepcidinas/metabolismo , Adenoviridae/genética , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Vectores Genéticos/genética , Hepcidinas/genética , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Sequestosoma-1/metabolismo
3.
J Cell Physiol ; 236(6): 4515-4527, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33442879

RESUMEN

Ischemic preconditioned (IP) neurons protect astrocytes against ischemia/reperfusion (I/R)-induced injury by inhibiting oxidative stress. However, the relevant mechanisms are unknown. Based on the role of nuclear factor-κB (NF-κB) in cell survival and adaption to oxidative stress, we hypothesized that NF-κB might be associated with astroprotection induced by IP neurons via upregulation of antioxidant enzymes. Here, we investigated the effects of IP neurons on NF-κB activation, cell viability, reactive oxygen species (ROS), expression of antioxidant enzymes, erythropoietin (EPO), and tumor necrosis factor α (TNF-α), in the presence or absence of BAY11-7082 (an NF-κB inhibitor), anti-EPO, and anti-TNF-α antibodies, in astrocytes treated with or without I/R. We found that IP neurons could keep NF-κB activation at a relatively higher but beneficial level, and in turn, upregulated the activity of antioxidant enzymes and hence enhanced cell viability and reduced ROS in I/R treated astrocytes. The results collectively indicated that IP neurons are able to significantly inhibit the I/R-induced NF-κB overactivation, probably via EPO and TNF-α, being essential for IP neuron-induced astroprotection under the conditions of I/R. We concluded that NF-κB-mediated antioxidative stress is one of the mechanisms by which IP neurons protect astrocytes against I/R injury.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Comunicación Paracrina , Daño por Reperfusión/prevención & control , Animales , Antioxidantes/metabolismo , Astrocitos/enzimología , Astrocitos/patología , Hipoxia de la Célula , Células Cultivadas , Corteza Cerebral/patología , Medios de Cultivo Condicionados/metabolismo , Eritropoyetina/metabolismo , Glucosa/deficiencia , Neuronas/patología , Estrés Oxidativo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
4.
Med Res Rev ; 40(2): 633-653, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31471929

RESUMEN

Abnormally high brain iron, resulting from the disrupted expression or function of proteins involved in iron metabolism in the brain, is an initial cause of neuronal death in neuroferritinopathy and aceruloplasminemia, and also plays a causative role in at least some of the other neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Friedreich's ataxia. As such, iron is believed to be a novel target for pharmacological intervention in these disorders. Reducing iron toward normal levels or hampering the increases in iron associated with age in the brain is a promising therapeutic strategy for all iron-related neurodegenerative disorders. Hepcidin is a crucial regulator of iron homeostasis in the brain. Recent studies have suggested that upregulating brain hepcidin levels can significantly reduce brain iron content through the regulation of iron transport protein expression in the blood-brain barrier and in neurons and astrocytes. In this review, we focus on the discussion of the therapeutic potential of hepcidin in iron-associated neurodegenerative diseases and also provide a systematic overview of recent research progress on how misregulated brain iron metabolism is involved in the development of multiple neurodegenerative disorders.


Asunto(s)
Hepcidinas/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ferroptosis/efectos de los fármacos , Hepcidinas/farmacología , Humanos , Hierro/metabolismo , Modelos Biológicos
5.
Biochem Biophys Res Commun ; 522(2): 322-327, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31761321

RESUMEN

BACKGROUND: Hepcidin is a key regulator of iron homeostasis. Some studies showed that exogenous hepcidin decreased the expression of divalent metal transporter (DMT1) rather than ferroportin(FPN1) to regulate renal iron metabolism. This study explored the effects of hepcidin synthesized by the kidney and its mechanism of iron regulation. METHODS: In the in vivo experiments, mice were divided into a unilateral ureter obstruction (UUO) model group and a sham operation group, and mice in the UUO model group were sacrificed on days 1, 3, 5 and 7. The expression of renal hepcidin, FPN1, DMT1 and the retention of renal iron were studied. In the in vitro experiments, we overexpressed hepcidin in HK-2 cells. Then we tested the expression of renal hepcidin, FPN1, DMT1 and observed the production of intracellular ferrous ions. RESULTS: Renal hepcidin expression was consistently higher in the UUO group than in the sham group from the first day. The expression of FPN1 gradually decreased, and the expression of DMT1 gradually increased in the UUO model. Intracellular ferrous ions significantly increased on the first day of the UUO model. In hepcidin overexpressed HK-2 cells, the expression of FPN1 was decreased, while the expression of DMT1 has no significant change. In addition, production of intracellular ferrous ions increased. CONCLUSION: local hepcidin can regulate iron metabolism in the kidney by adjusting the expression of FPN1.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hepcidinas/metabolismo , Espacio Intracelular/metabolismo , Sobrecarga de Hierro/metabolismo , Riñón/metabolismo , Animales , Ferritinas/metabolismo , Hierro/metabolismo , Masculino , Ratones Endogámicos C57BL
6.
J Comput Assist Tomogr ; 44(5): 708-713, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32936578

RESUMEN

OBJECTIVE: Assessing collateral status is important in acute ischemic stroke (AIS). The purpose of this study was to establish an easy and rapid method for evaluating collateral flow. METHODS: A total of 60 patients with AIS were enrolled. The patients were aged 18 to 85 years with endovascular therapy treatment within 10 hours after the appearance of stroke symptoms, prestroke modified Rankin Scale ≤1, Alberta Stroke Program Early CT Score ≥6, and the occlusion of large vessels in anterior circulation. We reformed imaging strategies by conducting a small-dose group-injection test before normal computed tomography angiography (CTA) scanning and selected the visual collateral score and the regional leptomeningeal score scales as the single-phase CTA collateral flow assessment scales with the replacement of the parasagittal anterior cerebral artery territory by anterior cerebral artery regions adjacent to the longitudinal fissure and then verified, respectively, the consistencies between the 2 single-phase CTA-based collateral scales and the digital subtraction angiography (DSA)-based American Society of Interventional and Therapeutic Neuroradiology/Society of Interventional Radiology scale and compared the prognosis of endovascular therapy between the AIS patients in the poor-collateral-flow group and the other patients' group assessed by 2 single-phase CTA-based collateral scales. RESULTS: There was a high consistency between the 2 single-phase CTA-based collateral flow scales with DSA-based American Society of Interventional and Therapeutic Neuroradiology/Society of Interventional Radiology scale. The assessment by using CTA-based collateral flow assessment methods generated consistent results. CONCLUSION: The single-phase CTA-based visual collateral score scale and regional leptomeningeal score scale can be used as the imaging evidence for the evaluation of collateral flow in AIS patients in the majority of grassroots hospitals where DSA is difficult to carry out.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Angiografía Cerebral/métodos , Circulación Colateral/fisiología , Angiografía por Tomografía Computarizada/métodos , Accidente Cerebrovascular/diagnóstico por imagen , Anciano , Angiografía de Substracción Digital/métodos , Encéfalo/irrigación sanguínea , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología
7.
J Cell Physiol ; 234(4): 3158-3169, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30370692

RESUMEN

Hydrogen sulfide (H2 S) has a significant effect on the regulation of interleukin-6 (IL-6) and signal transducer and activator of transcription 3 (STAT3) activities, while IL-6 directly regulates hepcidin expression via STAT3. We therefore hypothesized that H 2 S has a role in body iron homeostasis by regulating the expression of iron transport proteins via the IL-6/STAT3/Hepcidin pathway. Here, we investigated the effects of two H 2 S donors sodium hydrosulfide and GYY4137 on the expression of ferroportin-1 (Fpn1), transferrin receptor-1 (TfR1), hepcidin, IL-6 and pSTAT3 in the spleen of mice in vivo and peritoneal macrophage in vitro. We also examined the effects of H 2 S on serum iron, transferrin saturation, and ferritin light chain contents in the spleen, and on nitrite content, nuclear factor erythroid 2-related factor-2 (Nrf2) and iron regulatory protein 1 (IRP1) in the macrophages. We demonstrated that H 2 S regulates the expression of TfR1 and Fpn1 in the spleen in vivo and in peritoneal macrophages in vitro predominantly via the IL-6/pSTAT3/hepcidin pathway, under the conditions of inflammation induced by lipopolysaccharides. We also provide evidence that under uninflamed conditions, the regulation of Fpn1 and TfR1 expression by H 2 S, both in vivo and in vitro, are mediated by the nitric oxide (NO)/Nrf2 and iron regulatory protein/iron responsive element pathways, respectively, which are independent of IL-6/pSTAT3/hepcidin signals. These findings show that H 2 S is a key player in iron homeostasis under not only the inflamed conditions but also uninflamed conditions.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Sulfuro de Hidrógeno/farmacología , Hierro/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Morfolinas/farmacología , Compuestos Organotiofosforados/farmacología , Receptores de Transferrina/metabolismo , Bazo/efectos de los fármacos , Sulfuros/farmacología , Animales , Células Cultivadas , Hepcidinas/genética , Hepcidinas/metabolismo , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones Endogámicos C57BL , Morfolinas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/metabolismo , Compuestos Organotiofosforados/metabolismo , Fosforilación , Factor de Transcripción STAT3/metabolismo , Bazo/metabolismo , Sulfuros/metabolismo
8.
Hepatology ; 67(1): 21-35, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28859237

RESUMEN

Cystathionine ß-synthase (CBS) catalyzes the transsulfuration pathway and contributes, among other functions, to the generation of hydrogen sulfide. In view of the exceptionally high expression of CBS in the liver and the common interleukin-6 pathway used in the regulatory systems of hydrogen sulfide and hepcidin, we speculate that CBS is involved in body iron homeostasis. We found that CBS knockout (CBS-/- ) mice exhibited anemia and a significant increase in iron content in the serum, liver, spleen, and heart, along with severe damage to the liver, displaying a hemochromatosis-like phenotype. A high level of hepatic and serum hepcidin was also found. A major cause of the systemic iron overload is the reduced iron usage due to suppressed erythropoiesis, which is consistent with an increase in interleukin-6 and reduced expression of erythropoietin. Importantly, in the liver, absence of CBS caused both a reduction in the transcriptional factor nuclear factor erythroid 2-related factor-2 and an up-regulation of hepcidin that led to a decrease in the iron export protein ferroportin 1. The resulting suppression of iron export exacerbates iron retention, causing damage to hepatocytes. Finally, administration of CBS-overexpressing adenovirus into CBS mutant mice could partially reverse the iron-related phenotype. CONCLUSION: Our findings point to a critical role of CBS in iron homeostasis of the body, and the liver in particular; it is likely that a hemochromatosis-like phenotype in patients can be induced by aberration not only in the expression of key molecules in the hepcidin pathway but also of those related to CBS. (Hepatology 2018;67:21-35).


Asunto(s)
Anemia Ferropénica/enzimología , Anemia Ferropénica/patología , Cistationina betasintasa/metabolismo , Hepatocitos/enzimología , Hierro/metabolismo , Hígado/enzimología , Anemia Ferropénica/metabolismo , Animales , Biopsia con Aguja , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Hepatocitos/metabolismo , Hepcidinas/metabolismo , Homeostasis , Humanos , Inmunohistoquímica , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis Multivariante , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Valores de Referencia
9.
J Cell Physiol ; 233(1): 30-37, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28338217

RESUMEN

The significant positive correlation between ghrelin and iron and hepcidin levels in the plasma of children with iron deficiency anemia prompted us to hypothesize that ghrelin may affect iron metabolism. Here, we investigated the effects of fasting or ghrelin on the expression of hepcidin, ferroportin 1 (Fpn1), transferrin receptor 1 (TfR1), ferritin light chain (Ft-L) proteins, and ghrelin, and also hormone secretagogue receptor 1 alpha (GHSR1α) and ghrelin O-acyltransferase (GOAT) mRNAs in the spleen and/or macrophage. We demonstrated that fasting induces a significant increase in the expression of ghrelin, GHSR1α, GOAT, and hepcidin mRNAs, as well as Ft-L and Fpn1 but not TfR1 proteins in the spleens of mice in vivo. Similar to the effects of fasting on the spleen, ghrelin induced a significant increase in the expression of Ft-L and Fpn1 but not TfR1 proteins in macrophages in vitro. In addition, ghrelin was found to induce a significant enhancement in phosphorylation of ERK as well as translocation of pERK from the cytosol to nuclei. Furthermore, the increased pERK and Fpn1 induced by ghrelin was demonstrated to be preventable by pre-treatment with either GHSR1α antagonist or pERK inhibitor. Our findings support the hypothesis that fasting upregulates Fpn1 expression, probably via a ghrelin/GHSR/MAPK signaling pathway.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ayuno/metabolismo , Ghrelina/metabolismo , Macrófagos Peritoneales/enzimología , Receptores de Ghrelina/metabolismo , Transducción de Señal , Bazo/enzimología , Aciltransferasas/genética , Aciltransferasas/metabolismo , Animales , Apoferritinas/genética , Apoferritinas/metabolismo , Proteínas de Transporte de Catión/genética , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Ghrelina/genética , Antagonistas de Hormonas/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Proteínas de la Membrana , Ratones Endogámicos C57BL , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Ghrelina/antagonistas & inhibidores , Receptores de Ghrelina/genética , Bazo/efectos de los fármacos , Regulación hacia Arriba
10.
Neurochem Res ; 43(8): 1624-1630, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29923037

RESUMEN

A region-specific regulation of inflammation on the expression hepcidin in the brain has been demonstrated, however, it remains unknown whether there is also a cell-specific regulation of inflammation on hepcidin in the brain. Here, we investigated the effects of lipopolysaccharides (LPS) on the expression of hepcidin mRNA and also the expression of IL-6 mRNA, the phosphorylation of STAT3 and the expression of ferroportin 1 (Fpn1) and ferritin light chain (Ft-L) proteins in neurons and astrocytes obtained from wild type (IL-6+/+) and IL-6 knockout (IL-6-/-) mice. We demonstrated that the responses of the expression of hepcidin and IL-6 mRNAs, the phosphorylation of STAT3, and the expression of Fpn1 protein to LPS in IL-6+/+ astrocytes and also the responses of the expression of hepcidin mRNA, the phosphorylation of STAT3 and the expression of Fpn1 protein to IL-6 in IL-6-/- astrocytes were much stronger than those in IL-6+/+ and IL-6-/- neurons. A significant increase in Ft-L was found in LPS-treated IL-6+/+ and IL-6-treated IL-6-/- astrocytes, but not in LPS-treated IL-6+/+ and IL-6-treated IL-6-/- neurons. Our findings provide in vitro evidence for the existence of a cell-specific regulation of LPS on the expression of hepcidin and also Ft-L in the brain.


Asunto(s)
Astrocitos/efectos de los fármacos , Hepcidinas/metabolismo , Lipopolisacáridos/farmacología , Neuronas/efectos de los fármacos , Animales , Apoferritinas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Técnicas de Inactivación de Genes , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo
11.
Ann Nutr Metab ; 72(1): 37-42, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29241202

RESUMEN

BACKGROUND/AIMS: The studies in the patients with iron deficiency anemia (IDA) implied the existence of the association of ghrelin with iron or hepcidin levels in the plasma under the pathophysiological conditions. We hypothesized that fasting may be able to affect iron metabolism via ghrelin under the physiological conditions. METHODS: We investigated the effects of fasting on serum ghrelin and iron contents in healthy volunteers (23-31 years) and C57BL/6 male mice (8-week-olds) under the physiological conditions. RESULTS: Fasting induced a significant elevation in both total ghrelin and acylated ghrelin and a reduction in iron levels in the serum of both human and mice. Correlation analysis demonstrated that total ghrelin or acylated ghrelin is negatively correlated with iron in the serum in human and mice. CONCLUSION: Ghrelin has a role to reduce serum iron under the conditions of fasting.


Asunto(s)
Ayuno/sangre , Ghrelina/sangre , Hierro/sangre , Acilación , Adulto , Anemia Ferropénica/sangre , Animales , Femenino , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Adulto Joven
12.
Biochim Biophys Acta ; 1862(4): 518-525, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26769358

RESUMEN

Alpha-synuclein aggregation is the central hallmark of both sporadic and familial Parkinson's disease (PD). Patients with different PD-causing genetic defects of alpha-synuclein usually show distinctive clinical features that are atypical to sporadic PD. Iron accumulation is invariably found in PD. Recent studies showed that mutant and wild-type alpha-synuclein may have differential interaction with iron and mutant alpha-synuclein toxicity could be preferentially exacerbated by iron. We hence hypothesized that iron overload could selectively influence mutant alpha-synuclein toxicity and disease phenotypes. To test the hypothesis, we investigated if Drosophila melanogaster over-expressing A53T, A30P, and wild-type (WT) alpha-synuclein have different responses to iron treatment. We showed that iron treatment induced similar reduction of survival rate in all flies but induced a more severe motor decline in A53T and A30P mutant alpha-synuclein expressing flies, suggesting interaction between mutant alpha-synuclein and iron. Although no significant difference in total head iron content was found among these flies, we demonstrated that iron treatment induced selective DA neuron loss in motor-related PPM3 cluster only in the flies that express A53T and A30P mutant alpha-synuclein. We provided the first in vivo evidence that iron overload could induce distinctive neuropathology and disease phenotypes in mutant but not WT alpha-synuclein expressing flies, providing insights to the cause of clinical features selectively exhibited by mutant alpha-synuclein carriers.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Hierro/metabolismo , Neuronas Motoras/metabolismo , Mutación Missense , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/biosíntesis , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Neuronas Motoras/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Fenotipo , alfa-Sinucleína/genética
13.
Neurochem Res ; 42(10): 2798-2813, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28508173

RESUMEN

Increasing evidence demonstrates inflammation contributes to neuronal death following cerebral ischemia. Lycium barbarum polysaccharide (LBP) has been reported to prevent scopolamine-induced cognitive and memory deficits. We recently indicated that LBP exerts neuroprotective effect against focal cerebral ischemic injury in mice via attenuating the mitochondrial apoptosis pathway. The aim of this study was to investigate the neuroprotective effects of LBP against the behavioral dysfunction induced by focal cerebral ischemia injury in mice. Following 7 successive days of pretreatment with LBP (10, 20 and 40 mg/kg) and nimodipine (4 mg/kg) by intragastric gavage, mice were subjected to middle cerebral artery occlusion (MCAO). Following reperfusion, cerebral blood flows, the total power of the spontaneous EEG, and morphological changes were estimated. Learning and memory ability, and motor coordination were determined by Morris water maze task, rotarod and grip test. Western blot analysis, Real-Time fluorogenic PCR assays, and immunofluorescence staining were used to examine the expression of proinflammatory mediators and activation of microglia. The present study showed that LBP pretreatment significantly enhanced regional cortical blood flow and the total power of the spontaneous EEG, improved memory and motor coordination impairments, and inhibited over-activation of microglia and astrocytes after MCAO. Further study demonstrated LBP suppressed MCAO-induced activations of P65 NF-κB and P38 MAPK, and prevented up-regulations of proinflammatory mediators in hippocampus. Our data suggest that LBP can exert functional recovery of memory and motor coordination deficits and neuroprotective effect against cerebral ischemic injury in mice.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Muerte Celular/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/metabolismo
14.
Int J Mol Sci ; 17(12)2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27999284

RESUMEN

Aspirin down regulates transferrin receptor 1 (TfR1) and up regulates ferroportin 1 (Fpn1) and ferritin expression in BV-2 microglial cells treated without lipopolysaccharides (LPS), as well as down regulates hepcidin and interleukin 6 (IL-6) in cells treated with LPS. However, the relevant mechanisms are unknown. Here, we investigate the effects of aspirin on expression of hepcidin and iron regulatory protein 1 (IRP1), phosphorylation of Janus kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3) and P65 (nuclear factor-κB), and the production of nitric oxide (NO) in BV-2 microglial cells treated with and without LPS. We demonstrated that aspirin inhibited hepcidin mRNA as well as NO production in cells treated with LPS, but not in cells without LPS, suppresses IL-6, JAK2, STAT3, and P65 (nuclear factor-κB) phosphorylation and has no effect on IRP1 in cells treated with or without LPS. These findings provide evidence that aspirin down regulates hepcidin by inhibiting IL6/JAK2/STAT3 and P65 (nuclear factor-κB) pathways in the cells under inflammatory conditions, and imply that an aspirin-induced reduction in TfR1 and an increase in ferritin are not associated with IRP1 and NO.


Asunto(s)
Aspirina/farmacología , Hepcidinas/biosíntesis , Interleucina-6/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Microglía/efectos de los fármacos , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción ReIA/antagonistas & inhibidores , Animales , Línea Celular , Hepcidinas/genética , Inflamación/patología , Proteína 1 Reguladora de Hierro/biosíntesis , Janus Quinasa 2/metabolismo , Ratones , Óxido Nítrico/biosíntesis , Fosforilación/efectos de los fármacos , ARN Mensajero/biosíntesis , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
15.
Neurobiol Dis ; 66: 74-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24607885

RESUMEN

Based on the studies on the role of complements C3, C1q and factor B, we hypothesized that complement C5a is detrimental to locomotor recovery at the early stage of secondary injury after spinal cord injury (SCI). To test this hypothesis, we investigated the effect of inhibition of complement C5a receptor (C5aR) by using C5aR antagonist PMX53 (C5aRA) and deficiency of complement C5a receptor (C5aR-/- mice) on histological and locomotor recovery after SCI in mice. We demonstrated that the Basso Mouse Scale scores in the mice injected with C5aRA (C5aRA-mice) at 45min before and 24h after SCI and the C5aR-/- mice were markedly higher than those in the mice treated with saline (Saline-mice) and the C5aR+/+ mice respectively between 7 and 28days after SCI. Also, expression of TNF-α and IL-1ß in C5aRA-mice was significantly lower than that in Saline-mice from 1 to 24h after SCI. In addition, the percentage of microglia/macrophage in C5aRA mice and C5aR-/- mice was significantly lower than those in their corresponding control groups from 1 to 14days after SCI. Furthermore, C5aRA mice and C5aR-/- mice had less GFAP expression in the injured spinal cord epicenter as compared to Saline mice and C5aR+/+ mice at day 28 after SCI. These findings provided evidence that inhibition or deficiency of C5aR could significantly improve histological and functional locomotor recovery after SCI in mice.


Asunto(s)
Actividad Motora/fisiología , Receptor de Anafilatoxina C5a/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Animales , Fármacos del Sistema Nervioso Central/farmacología , Femenino , Proteína Ácida Fibrilar de la Glía , Interleucina-1beta/metabolismo , Macrófagos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/fisiología , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Péptidos Cíclicos/farmacología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Índice de Severidad de la Enfermedad , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
16.
Neurochem Res ; 39(5): 893-900, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24682751

RESUMEN

Based on the well-confirmed roles of angiotensin II (ANGII) in iron transport of peripheral organs and cells, the causative link of excess brain iron with and the involvement of ANGII in neurodegenerative disorders, we speculated that ANGII might also have an effect on expression of iron transport proteins in the brain. In the present study, we investigated effects of ANGII on iron uptake and release using the radio-isotope methods as well as expression of cell iron transport proteins by Western blot analysis in cultured neurons. Our findings demonstrated for the first time that ANGII significantly reduced transferrin-bound iron and non-transferrin bound iron uptake and iron release as well as expression of two major iron uptake proteins transferrin receptor 1 and divalent metal transporter 1 and the key iron exporter ferroportin 1 in cultured neurons. The findings suggested that endogenous ANGII might have a physiological significance in brain iron metabolism.


Asunto(s)
Angiotensina II/fisiología , Hierro/metabolismo , Transferrina/metabolismo , Angiotensina II/farmacología , Animales , Antígenos CD/biosíntesis , Proteínas de Transporte de Catión/biosíntesis , Células Cultivadas , Radioisótopos de Hierro/metabolismo , Masculino , Neuronas/metabolismo , Ratas Sprague-Dawley , Receptores de Transferrina/biosíntesis
17.
Sheng Li Xue Bao ; 66(1): 47-54, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24553869

RESUMEN

In recent years, iron has been regarded as a common pathological feature of many neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and Friedreich's ataxia (FRDA). A number of genes involved in iron transport, storage and regulation have been found associated with initiation and progression of neurodegeneration. However, whether iron abnormalities represent a primary or secondary event still remains unknown. Due to the limitation in transgenic rodent model construction and transfection systems, the progress in unraveling the pathogenic role of different iron-related proteins in neurodegenerative diseases has been slow. Drosophila melanogaster, a simple organism which has a shorter lifespan and smaller genome with many conserved genes, and captures many features of human nervous system and neurodegeneration, may help speed up the progress. The characteristics that spatial- and temporal-specific transgenic Drosophila can be easily constructed and raised in large quantity with phenotype easily determined turn Drosophila into an excellent in vivo genetic system for screening iron-related modifiers in different neurodegenerative conditions and hence provide a better picture about the pathogenic contribution of different iron-related protein abnormalities. It is believed that identification of important iron-related genes that can largely stop or even reverse degenerative process in Drosophila models may lead to development of novel therapeutic strategies against neurodegenerative diseases.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila melanogaster , Hierro , Enfermedades Neurodegenerativas/fisiopatología , Enfermedad de Alzheimer/fisiopatología , Animales , Ataxia de Friedreich/fisiopatología , Humanos , Enfermedad de Parkinson/fisiopatología
18.
Bioact Mater ; 38: 55-72, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38699242

RESUMEN

As a natural immune cell and antigen presenting cell, macrophages have been studied and engineered to treat human diseases. Macrophages are well-suited for use as drug carriers because of their biological characteristics, such as excellent biocompatibility, long circulation, intrinsic inflammatory homing and phagocytosis. Meanwhile, macrophages' uniquely high plasticity and easy re-education polarization facilitates their use as part of efficacious therapeutics for the treatment of inflammatory diseases or tumors. Although recent studies have demonstrated promising advances in macrophage-based drug delivery, several challenges currently hinder further improvement of therapeutic effect and clinical application. This article focuses on the main challenges of utilizing macrophage-based drug delivery, from the selection of macrophage sources, drug loading, and maintenance of macrophage phenotypes, to drug migration and release at target sites. In addition, corresponding strategies and insights related to these challenges are described. Finally, we also provide perspective on shortcomings on the road to clinical translation and production.

19.
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
20.
Mol Neurobiol ; 61(4): 2006-2020, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37833459

RESUMEN

Both neuroinflammation and iron accumulation play roles in the pathogenesis of Parkinson's disease (PD). However, whether inflammation induces iron dyshomeostasis in dopaminergic neurons at an early stage of PD, at which no quantifiable dopaminergic neuron loss can be observed, is still unknown. As for the inflammation mediators, although several cytokines have been reported to increase in PD, the functions of these cytokines in the SN are double-edged and controversial. In this study, whether inflammation could induce iron dyshomeostasis in dopaminergic neurons through high mobility group protein B1 (HMGB1) in the early stage of PD is explored. Lipopolysaccharide (LPS), a toxin that primarily activates glia cells, and 6-hydroxydopamine (6-OHDA), the neurotoxin that firstly impacts dopaminergic neurons, were utilized to mimic PD in rats. We found a common and exceedingly early over-production of HMGB1, followed by an increase of divalent metal transporter 1 with iron responsive element (DMT1+) in the dopaminergic neurons before quantifiable neuronal loss. HMGB1 neutralizing antibody suppressed inflammation in the SN, DMT1+ elevation in dopaminergic neurons, and dopaminergic neuronal loss in both LPS and 6-OHDA administration- induced PD models. On the contrary, interleukin-1ß inhibitor diacerein failed to suppress these outcomes induced by 6-OHDA. Our findings not only demonstrate that inflammation could be one of the causes of DMT1+ increase in dopaminergic neurons, but also highlight HMGB1 as a pivotal early mediator of inflammation-induced iron increase and subsequent neurodegeneration, thereby HMGB1 could serve as a potential target for early-stage PD treatment.


Asunto(s)
Proteína HMGB1 , Enfermedad de Parkinson , Trastornos Parkinsonianos , Animales , Ratas , Citocinas/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteína HMGB1/metabolismo , Inflamación/patología , Hierro/metabolismo , Lipopolisacáridos , Oxidopamina , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA