Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 39(3): 413-431, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30700131

RESUMEN

Objective- Calcific aortic valve disease is a prominent finding in elderly and in patients with chronic kidney disease. We investigated the potential role of iron metabolism in the pathogenesis of calcific aortic valve disease. Approach and Results- Cultured valvular interstitial cells of stenotic aortic valve with calcification from patients undergoing valve replacement exhibited significant susceptibility to mineralization/osteoblastic transdifferentiation in response to phosphate. This process was abrogated by iron via induction of H-ferritin as reflected by lowering ALP and osteocalcin secretion and preventing extracellular calcium deposition. Cellular phosphate uptake and accumulation of lysosomal phosphate were decreased. Accordingly, expression of phosphate transporters Pit1 and Pit2 were repressed. Translocation of ferritin into lysosomes occurred with high phosphate-binding capacity. Importantly, ferritin reduced nuclear accumulation of RUNX2 (Runt-related transcription factor 2), and as a reciprocal effect, it enhanced nuclear localization of transcription factor Sox9 (SRY [sex-determining region Y]-box 9). Pyrophosphate generation was also increased via upregulation of ENPP2 (ectonucleotide pyrophosphatase/phosphodiesterase-2). 3H-1, 2-dithiole-3-thione mimicked these beneficial effects in valvular interstitial cell via induction of H-ferritin. Ferroxidase activity of H-ferritin was essential for this function, as ceruloplasmin exhibited similar inhibitory functions. Histological analysis of stenotic aortic valve revealed high expression of H-ferritin without iron accumulation and its relative dominance over ALP in noncalcified regions. Increased expression of H-ferritin accompanied by elevation of TNF-α (tumor necrosis factor-α) and IL-1ß (interleukin-1ß) levels, inducers of H-ferritin, corroborates the essential role of ferritin/ferroxidase via attenuating inflammation in calcific aortic valve disease. Conclusions- Our results indicate that H-ferritin is a stratagem in mitigating valvular mineralization/osteoblastic differentiation. Utilization of 3H-1, 2-dithiole-3-thione to induce ferritin expression may prove a novel therapeutic potential in valvular mineralization.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Apoferritinas/fisiología , Calcificación Vascular/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/patología , Apoferritinas/antagonistas & inhibidores , Apoferritinas/farmacología , Transporte Biológico , Núcleo Celular/metabolismo , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Canales Iónicos/biosíntesis , Hierro/farmacología , Lisosomas/metabolismo , Fosfatos/metabolismo , Hidrolasas Diéster Fosfóricas/biosíntesis , Hidrolasas Diéster Fosfóricas/genética , Factor de Transcripción SOX9/metabolismo , Tionas/farmacología , Tiofenos/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Calcificación Vascular/patología
2.
J Neurochem ; 147(6): 831-848, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30152072

RESUMEN

For more than 150 years, it is known that occupational overexposure of manganese (Mn) causes movement disorders resembling Parkinson's disease (PD) and PD-like syndromes. However, the mechanisms of Mn toxicity are still poorly understood. Here, we demonstrate that Mn dose- and time-dependently blocks the protein translation of amyloid precursor protein (APP) and heavy-chain Ferritin (H-Ferritin), both iron homeostatic proteins with neuroprotective features. APP and H-Ferritin are post-transcriptionally regulated by iron responsive proteins, which bind to homologous iron responsive elements (IREs) located in the 5'-untranslated regions (5'-UTRs) within their mRNA transcripts. Using reporter assays, we demonstrate that Mn exposure repressed the 5'-UTR-activity of APP and H-Ferritin, presumably via increased iron responsive proteins-iron responsive elements binding, ultimately blocking their protein translation. Using two specific Fe2+ -specific probes (RhoNox-1 and IP-1) and ion chromatography inductively coupled plasma mass spectrometry (IC-ICP-MS), we show that loss of the protective axis of APP and H-Ferritin resulted in unchecked accumulation of redox-active ferrous iron (Fe2+ ) fueling neurotoxic oxidative stress. Enforced APP expression partially attenuated Mn-induced generation of cellular and lipid reactive oxygen species and neurotoxicity. Lastly, we could validate the Mn-mediated suppression of APP and H-Ferritin in two rodent in vivo models (C57BL6/N mice and RjHan:SD rats) mimicking acute and chronic Mn exposure. Together, these results suggest that Mn-induced neurotoxicity is partly attributable to the translational inhibition of APP and H-Ferritin resulting in impaired iron metabolism and exacerbated neurotoxic oxidative stress. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Asunto(s)
Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Apoferritinas/antagonistas & inhibidores , Hierro/metabolismo , Intoxicación por Manganeso/metabolismo , Regiones no Traducidas 5' , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apoferritinas/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Modificación Traduccional de las Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
3.
Phys Chem Chem Phys ; 17(29): 19001-11, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26126644

RESUMEN

Cationic dendrimers, such as PAMAM, are known to be positively charged at neutral pH allowing their unspecific interaction with proteins and other cellular components. Especially, ferritin, which has an important role in iron homeostasis, presents a negative electrostatic potential at the 3-fold channel. This channel is important in the functionality of ferritin because it allows the iron entry into its inner cavity. In this way, the interaction between the protonated terminal amines of the dendrimer and the negatively charged 3-fold channels of ferritin is expected. Experimental measurements demonstrated that PAMAM G4 inhibits the iron storage properties of L-chain human ferritin (L-Ftn). Molecular dynamics simulations have been used to analyze the specific interaction between PAMAM G4 and L-Ftn. Results show that PAMAM G4 effectively interacts with the 3-fold channels of L-Ftn, suggesting that this interaction is responsible for the inhibition of the iron storage properties of L-Ftn.


Asunto(s)
Apoferritinas/antagonistas & inhibidores , Dendrímeros/farmacología , Hierro/metabolismo , Nylons/farmacología , Apoferritinas/genética , Apoferritinas/metabolismo , Dendrímeros/química , Humanos , Simulación de Dinámica Molecular , Nylons/química
4.
FEBS J ; 281(10): 2410-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24673886

RESUMEN

Siderophores are best known as small iron-binding molecules that facilitate iron uptake in bacteria and fungi. In our previous study, we demonstrated that eukaryotes also produce siderophore-like molecules via a remarkably conserved biosynthetic pathway. A member of the short-chain dehydrogenase family of reductases, 3-hydroxybutyrate dehydrogenase-2, catalyzes a rate-limiting step in the biogenesis of the mammalian siderophore. Physiologically, depletion of the mammalian siderophore by inhibiting expression of the 3-hydroxybutyrate dehydrogenase-2 gene (Bdh2) results in abnormal accumulation of intracellular iron, increased oxidative stress, and mitochondrial iron deficiency. Thus, the mammalian siderophore is an important regulator of cellular iron homeostasis. The cellular iron storage protein ferritin also regulates iron metabolism and protects cells from oxidative stress. Depletion of ferritin results in intracellular iron accumulation, predisposes to oxidative stress, and confers a growth advantage to cells. We therefore hypothesize that the siderophore and ferritin coregulate cellular iron metabolism/homeostasis in eukaryotes. We tested this prediction by depleting both the siderophore and ferritin. This resulted in a marked accumulation of cellular iron, and caused increased sensitivity to oxidants. Interestingly, cells lacking both the siderophore and ferritin proliferated at a higher rate than cells lacking either of these components alone. Taken together, our findings suggest that the siderophore and ferritin synergistically regulate cellular iron levels.


Asunto(s)
Apoferritinas/metabolismo , Hidroxibutirato Deshidrogenasa/metabolismo , Hierro/metabolismo , Animales , Apoferritinas/antagonistas & inhibidores , Apoferritinas/genética , Línea Celular , Técnicas de Silenciamiento del Gen , Homeostasis , Hidroxibutirato Deshidrogenasa/antagonistas & inhibidores , Hidroxibutirato Deshidrogenasa/genética , Ratones , Estrés Oxidativo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Sideróforos/metabolismo
5.
Chem Commun (Camb) ; 50(6): 701-3, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24284378

RESUMEN

A selective and specific inhibition of the catalase mimetic activity but not SOD-like activity of Pt-apoferritin can be achieved through the choice of the inhibitor. The recovery of activity using a reducing agent was explored and proven to successfully restore the surface-chemistry of NPs.


Asunto(s)
Apoferritinas/química , Apoferritinas/metabolismo , Biomimética , Platino (Metal)/química , Apoferritinas/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Peróxido de Hidrógeno/química , Nanopartículas/química , Sustancias Reductoras/química , Propiedades de Superficie
6.
Free Radic Biol Med ; 67: 19-29, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24231192

RESUMEN

The efficacy of doxorubicin (DOX) as an antitumor agent is greatly limited by the induction of cardiomyopathy, which results from mitochondrial dysfunction and iron-catalyzed oxidative stress in the cardiomyocyte. Metformin (MET) has been seen to have a protective effect against the oxidative stress induced by DOX in cardiomyocytes through its modulation of ferritin heavy chain (FHC), the main iron-storage protein. This study aimed to assess the involvement of FHC as a pivotal molecule in the mitochondrial protection offered by MET against DOX cardiotoxicity. The addition of DOX to adult mouse cardiomyocytes (HL-1 cell line) increased the cytosolic and mitochondrial free iron pools in a time-dependent manner. Simultaneously, DOX inhibited complex I activity and ATP generation and induced the loss of mitochondrial membrane potential. The mitochondrial dysfunction induced by DOX was associated with the release of cytochrome c to the cytosol, the activation of caspase 3, and DNA fragmentation. The loss of iron homeostasis, mitochondrial dysfunction, and apoptosis induced by DOX were prevented by treatment with MET 24h before the addition of DOX. The involvement of FHC and NF-κB was determined through siRNA-mediated knockdown. Interestingly, the presilencing of FHC or NF-κB with specific siRNAs blocked the protective effect induced by MET against DOX cardiotoxicity. These findings were confirmed in isolated primary neonatal rat cardiomyocytes. In conclusion, these results deepen our knowledge of the protective action of MET against DOX-induced cardiotoxicity and suggest that therapeutic strategies based on FHC modulation could protect cardiomyocytes from the mitochondrial damage induced by DOX by restoring iron homeostasis.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Apoferritinas/genética , Cardiotónicos/farmacología , Doxorrubicina/farmacología , Metformina/farmacología , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoferritinas/antagonistas & inhibidores , Apoferritinas/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Fragmentación del ADN/efectos de los fármacos , Regulación de la Expresión Génica , Hierro/metabolismo , Ratones , Mitocondrias/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal
7.
Int J Oncol ; 43(5): 1481-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23969999

RESUMEN

The development of resistance of cancer cells to therapeutic agents is the major obstacle in the successful treatment of breast cancer and the main cause of breast cancer recurrence. The results of several studies have demonstrated an important role of altered cellular iron metabolism in the progression of breast cancer and suggested that iron metabolism may be involved in the acquisition of a cancer cell drug-resistant phenotype. In the present study, we show that human MCF-7 breast cancer cells with an acquired resistance to the chemotherapeutic drugs doxorubicin (MCF-7/DOX) and cisplatin (MCF-7/CDDP) exhibited substantial alterations in the intracellular iron content and levels of iron-regulatory proteins involved in the cellular uptake, storage and export of iron, especially in profoundly increased levels of ferritin light chain (FTL) protein. The increased levels of FTL in breast cancer indicate that FTL may be used as a diagnostic and prognostic marker for breast cancer. Additionally, we demonstrate that targeted downregulation of FTL protein by the microRNA miR-133a increases sensitivity of MCF-7/DOX and MCF-7/CDDP cells to doxorubicin and cisplatin. These results suggest that correction of iron metabolism abnormalities may substantially improve the efficiency of breast cancer treatment.


Asunto(s)
Apoferritinas/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Cisplatino/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Hierro/metabolismo , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Apoferritinas/antagonistas & inhibidores , Apoferritinas/genética , Western Blotting , Neoplasias de la Mama/patología , Espectroscopía de Resonancia por Spin del Electrón , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , MicroARNs/genética , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
8.
J Biol Chem ; 281(49): 37616-27, 2006 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-17056593

RESUMEN

Chemokine receptor-initiated signaling plays critical roles in cell differentiation, proliferation, and migration. However, the regulation of chemokine receptor signaling under physiological and pathological conditions is not fully understood. In the present study, we demonstrate that the CXC chemokine receptor 4 (CXCR4) formed a complex with ferritin heavy chain (FHC) in a ligand-dependent manner. Our in vitro binding assays revealed that purified FHC associated with both the glutathione S-transferase-conjugated N-terminal and C-terminal domains of CXCR4, thereby suggesting the presence of more than one FHC binding site in the protein sequence of CXCR4. Using confocal microscopy, we observed that stimulation with CXCL12, the receptor ligand, induced colocalization of the internalized CXCR4 with FHC into internal vesicles. Furthermore, after CXCL12 treatment, FHC underwent time-dependent nuclear translocation and phosphorylation at serine residues. By contrast, a mutant form of FHC in which serine 178 was replaced by alanine (S178A) failed to undergo phosphorylation, suggesting that serine 178 is the major phosphorylation site. Compared with the wild type FHC, the FHC-S178A mutant exhibited reduced association with CXCR4 and constitutive nuclear translocation. We also found that CXCR4-mediated extracellular signal-regulated kinase 1/2 (ERK1/2) activation and chemotaxis were inhibited by overexpression of wild type FHC but not FHC-S178A mutant, and were prolonged by FHC knockdown. In addition to CXCR4, other chemokine receptor-initiated signaling appeared to be similarly regulated by FHC, because CXCR2-mediated ERK1/2 activation was also inhibited by FHC overexpression and prolonged by FHC knockdown. Altogether, our data provide strong evidence for an important role of FHC in chemokine receptor signaling and receptor-mediated cell migration.


Asunto(s)
Apoferritinas/metabolismo , Quimiocinas CXC/metabolismo , Receptores CXCR4/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Apoferritinas/antagonistas & inhibidores , Apoferritinas/genética , Línea Celular , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimiocinas CXC/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Células Jurkat , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores CXCR4/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA