Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3802, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714719

RESUMEN

The interaction between nuclear receptor coactivator 4 (NCOA4) and the iron storage protein ferritin is a crucial component of cellular iron homeostasis. The binding of NCOA4 to the FTH1 subunits of ferritin initiates ferritinophagy-a ferritin-specific autophagic pathway leading to the release of the iron stored inside ferritin. The dysregulation of NCOA4 is associated with several diseases, including neurodegenerative disorders and cancer, highlighting the NCOA4-ferritin interface as a prime target for drug development. Here, we present the cryo-EM structure of the NCOA4-FTH1 interface, resolving 16 amino acids of NCOA4 that are crucial for the interaction. The characterization of mutants, designed to modulate the NCOA4-FTH1 interaction, is used to validate the significance of the different features of the binding site. Our results explain the role of the large solvent-exposed hydrophobic patch found on the surface of FTH1 and pave the way for the rational development of ferritinophagy modulators.


Asunto(s)
Microscopía por Crioelectrón , Ferritinas , Coactivadores de Receptor Nuclear , Ferritinas/metabolismo , Ferritinas/química , Ferritinas/genética , Humanos , Coactivadores de Receptor Nuclear/metabolismo , Coactivadores de Receptor Nuclear/química , Coactivadores de Receptor Nuclear/genética , Unión Proteica , Sitios de Unión , Hierro/metabolismo , Autofagia , Modelos Moleculares , Células HEK293 , Oxidorreductasas/metabolismo , Oxidorreductasas/química , Oxidorreductasas/genética , Proteolisis , Mutación
2.
JCI Insight ; 9(5)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38301068

RESUMEN

Acute bacterial orchitis (AO) is a prevalent cause of intrascrotal inflammation, often resulting in sub- or infertility. A frequent cause eliciting AO is uropathogenic Escherichia coli (UPEC), a gram negative pathovar, characterized by the expression of various iron acquisition systems to survive in a low-iron environment. On the host side, iron is tightly regulated by iron regulatory proteins 1 and 2 (IRP1 and -2) and these factors are reported to play a role in testicular and immune cell function; however, their precise role remains unclear. Here, we showed in a mouse model of UPEC-induced orchitis that the absence of IRP1 results in less testicular damage and a reduced immune response. Compared with infected wild-type (WT) mice, testes of UPEC-infected Irp1-/- mice showed impaired ERK signaling. Conversely, IRP2 deletion led to a stronger inflammatory response. Notably, differences in immune cell infiltrations were observed among the different genotypes. In contrast with WT and Irp2-/- mice, no increase in monocytes and neutrophils was detected in testes of Irp1-/- mice upon UPEC infection. Interestingly, in Irp1-/- UPEC-infected testes, we observed an increase in a subpopulation of macrophages (F4/80+CD206+) associated with antiinflammatory and wound-healing activities compared with WT. These findings suggest that IRP1 deletion may protect against UPEC-induced inflammation by modulating ERK signaling and dampening the immune response.


Asunto(s)
Proteína 1 Reguladora de Hierro , Orquitis , Masculino , Humanos , Ratones , Animales , Proteína 1 Reguladora de Hierro/genética , Proteína 1 Reguladora de Hierro/metabolismo , Orquitis/microbiología , Inflamación , Proteína 2 Reguladora de Hierro/genética , Proteína 2 Reguladora de Hierro/metabolismo , Hierro/metabolismo
3.
Am J Hematol ; 99(1): 12-20, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37867341

RESUMEN

Ferritin is a hetero-oligomeric nanocage, composed of 24 subunits of two types, FTH1 and FTL. It protects the cell from excess reactive iron, by storing iron in its cavity. FTH1 is essential for the recruitment of iron into the ferritin nanocage and for cellular ferritin trafficking, whereas FTL contributes to nanocage stability and iron nucleation inside the cavity. Here we describe a female patient with a medical history of severe hypoferritinemia without anemia. Following inadequate heavy IV iron supplementation, the patient developed severe iron overload and musculoskeletal manifestations. However, her serum ferritin levels rose only to normal range. Genetic analyses revealed an undescribed homozygous variant of FTL (c.92A > G), which resulted in a Tyr31Cys substitution (FTLY31C ). Analysis of the FTL structure predicted that the Y31C mutation will reduce the variant's stability. Expression of the FTLY31C variant resulted in significantly lower cellular ferritin levels compared with the expression of wild-type FTL (FTLWT ). Proteasomal inhibition significantly increased the initial levels of FTLY31C , but could not protect FTLY31C subunits from successive degradation. Further, variant subunits successfully incorporated into hetero-polymeric nanocages in the presence of sufficient levels of FTH1. However, FTLY31C subunits poorly assembled into nanocages when FTH1 subunit levels were low. These results indicate an increased susceptibility of unassembled monomeric FTLY31C subunits to proteasomal degradation. The decreased cellular assembly of FTLY31C -rich nanocages may explain the low serum ferritin levels in this patient and emphasize the importance of a broader diagnostic approach of hypoferritinemia without anemia, before IV iron supplementation.


Asunto(s)
Anemia , Apoferritinas , Deficiencias de Hierro , Sobrecarga de Hierro , Femenino , Humanos , Anemia/genética , Apoferritinas/genética , Apoferritinas/metabolismo , Ferritinas , Hierro/metabolismo , Deficiencias de Hierro/genética , Sobrecarga de Hierro/genética
4.
Elife ; 122023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37561022

RESUMEN

Postmenopausal atherosclerosis (AS) has been attributed to estrogen deficiency. However, the beneficial effect of hormone replacement therapy (HRT) is lost in late postmenopausal women with atherogenesis. We asked whether aging-related iron accumulation affects estrogen receptor α (ERα) expression, thus explaining HRT inefficacy. A negative correlation has been observed between aging-related systemic iron deposition and ERα expression in postmenopausal AS patients. In an ovariectomized Apoe-/- mouse model, estradiol treatment had contrasting effects on ERα expression in early versus late postmenopausal mice. ERα expression was inhibited by iron treatment in cell culture and iron-overloaded mice. Combined treatment with estradiol and iron further decreased ERα expression, and the latter effect was mediated by iron-regulated E3 ligase Mdm2. In line with these observations, cellular cholesterol efflux was reduced, and endothelial homeostasis was disrupted. Consequently, AS was aggravated. Accordingly, systemic iron chelation attenuated estradiol-triggered progressive AS in late postmenopausal mice. Thus, iron and estradiol together downregulate ERα through Mdm2-mediated proteolysis, providing a potential explanation for failures of HRT in late postmenopausal subjects with aging-related iron accumulation. This study suggests that immediate HRT after menopause, along with appropriate iron chelation, might provide benefits from AS.


Asunto(s)
Aterosclerosis , Receptor alfa de Estrógeno , Humanos , Femenino , Ratones , Animales , Receptor alfa de Estrógeno/genética , Posmenopausia , Terapia de Reemplazo de Estrógeno , Aterosclerosis/metabolismo , Estradiol , Terapia de Reemplazo de Hormonas , Quelantes del Hierro
5.
Int J Mol Sci ; 24(5)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36902088

RESUMEN

Due to its advantageous redox properties, iron plays an important role in the metabolism of nearly all life. However, these properties are not only a boon but also the bane of such life forms. Since labile iron results in the generation of reactive oxygen species by Fenton chemistry, iron is stored in a relatively safe form inside of ferritin. Despite the fact that the iron storage protein ferritin has been extensively researched, many of its physiological functions are hitherto unresolved. However, research regarding ferritin's functions is gaining momentum. For example, recent major discoveries on its secretion and distribution mechanisms have been made as well as the paradigm-changing finding of intracellular compartmentalization of ferritin via interaction with nuclear receptor coactivator 4 (NCOA4). In this review, we discuss established knowledge as well as these new findings and the implications they may have for host-pathogen interaction during bacterial infection.


Asunto(s)
Infecciones Bacterianas , Ferritinas , Humanos , Ferritinas/metabolismo , Hierro/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Coactivadores de Receptor Nuclear/metabolismo
6.
Int J Mol Sci ; 23(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36361875

RESUMEN

The sequestration of iron in case of infection, termed nutritional immunity, is an established strategy of host defense. However, the interaction between pathogens and the mammalian iron storage protein ferritin is hitherto not completely understood. To better characterize the function of ferritin in Gram-negative infections, we incubated iron-starved cultures of Salmonella Typhimurium and knockout mutant strains defective for major iron uptake pathways or Escherichia coli with horse spleen ferritin or ionic iron as the sole iron source. Additionally, we added bovine superoxide dismutase and protease inhibitors to the growth medium to assess the effect of superoxide and bacterial proteases, respectively, on Salmonella proliferation and reductive iron release. Compared to free ionic iron, ferritin-bound iron was less available to Salmonella, but was still sufficient to significantly enhance the growth of the bacteria. In the absence of various iron acquisition genes, the availability of ferritin iron further decreased. Supplementation with superoxide dismutase significantly reduced the growth of the ΔentC knockout strain with holoferritin as the sole iron source in comparison with ionic ferrous iron. In contrast, this difference was not observed in the wildtype strain, suggesting that superoxide dismutase undermines bacterial iron uptake from ferritin by siderophore-independent mechanisms. Ferritin seems to diminish iron availability for bacteria in comparison to ionic iron, and its iron sequestering effect could possibly be enhanced by host superoxide dismutase activity.


Asunto(s)
Ferritinas , Hierro , Bovinos , Animales , Caballos , Ferritinas/metabolismo , Hierro/metabolismo , Enterobacteriaceae , Salmonella typhimurium , Superóxido Dismutasa/metabolismo , Escherichia coli/metabolismo , Mamíferos/metabolismo
7.
J Am Chem Soc ; 142(46): 19551-19557, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33166133

RESUMEN

Biomineralization is mediated by specialized proteins that guide and control mineral sedimentation. In many cases, the active regions of these biomineralization proteins are intrinsically disordered. High-resolution structures of these proteins while they interact with minerals are essential for understanding biomineralization processes and the function of intrinsically disordered proteins (IDPs). Here we used the cavity of ferritin as a nanoreactor where the interaction between M6A, an intrinsically disordered iron-binding domain, and an iron oxide particle was visualized at high resolution by cryo-EM. Taking advantage of the differences in the electron-dose sensitivity of the protein and the iron oxide particles, we developed a method to determine the irregular shape of the particles found in our density maps. We found that the folding of M6A correlates with the detection of mineral particles in its vicinity. M6A interacts with the iron oxide particles through its C-terminal side, resulting in the stabilization of a helix at its N-terminal side. The stabilization of the helix at a region that is not in direct contact with the iron oxide particle demonstrates the ability of IDPs to respond to signals from their surroundings by conformational changes. These findings provide the first glimpse toward the long-suspected mechanism for biomineralization protein control over mineral microstructure, where unstructured regions of these proteins become more ordered in response to their interaction with the nascent mineral particles.


Asunto(s)
Apoferritinas/química , Proteínas Bacterianas/química , Microscopía por Crioelectrón/métodos , Compuestos Férricos/química , Proteínas Intrínsecamente Desordenadas/química , Péptidos/química , Sitios de Unión , Biomineralización , Nanopartículas Magnéticas de Óxido de Hierro/química , Magnetospirillum/química , Modelos Moleculares , Tamaño de la Partícula , Unión Proteica , Conformación Proteica , Pliegue de Proteína
8.
Elife ; 82019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31793879

RESUMEN

Lysosomal acidification is a key feature of healthy cells. Inability to maintain lysosomal acidic pH is associated with aging and neurodegenerative diseases. However, the mechanisms elicited by impaired lysosomal acidification remain poorly understood. We show here that inhibition of lysosomal acidification triggers cellular iron deficiency, which results in impaired mitochondrial function and non-apoptotic cell death. These effects are recovered by supplying iron via a lysosome-independent pathway. Notably, iron deficiency is sufficient to trigger inflammatory signaling in cultured primary neurons. Using a mouse model of impaired lysosomal acidification, we observed a robust iron deficiency response in the brain, verified by in vivo magnetic resonance imaging. Furthermore, the brains of these mice present a pervasive inflammatory signature associated with instability of mitochondrial DNA (mtDNA), both corrected by supplementation of the mice diet with iron. Our results highlight a novel mechanism linking impaired lysosomal acidification, mitochondrial malfunction and inflammation in vivo.


Asunto(s)
Ácidos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Deficiencias de Hierro , Lisosomas/metabolismo , Animales , Apoptosis , Encéfalo/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Proliferación Celular , ADN Mitocondrial/genética , Modelos Animales de Enfermedad , Transporte de Electrón , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , Concentración de Iones de Hidrógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata , Inflamación/genética , Hierro/farmacología , Lisosomas/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Biogénesis de Organelos , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , ATPasas de Translocación de Protón Vacuolares/metabolismo , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo
9.
Food Funct ; 10(3): 1763-1766, 2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30794278

RESUMEN

This commentary re-emphasizes the aim of our recent review (David et al., 2018) and addresses some of the points raised in the adjacent commentary by M. Weiner and J. McKim, Food Funct., 2019, 10, DOI: 10.1039/C8FO01282B. In agreement with the commentary, the discussed review highlights the need to adequately understand the complex physicochemistry of the food additive carrageenan (CGN) and its fate in the alimentary canal. In fact, there is a realm of scientific findings that justify the continuation of an open discussion of CGN safety. This response emphasizes that there is sparse information on [i] the physicochemical properties of commercial CGN, [ii] human levels of exposure to CGN from foods, [iii] the role of CGN in gut microbiome dysbiosis and inflammation, and [iv] the effects of CGN on susceptible populations. As long as the determinants of the increased prevalence of chronic and autoimmune diseases are not identified, we must continue to explore the possible beneficial or deleterious effects that may arise from extrinsic factors, including food additives, and do so in meticulous independent studies.


Asunto(s)
Carragenina/efectos adversos , Carragenina/química , Aditivos Alimentarios/efectos adversos , Aditivos Alimentarios/química , Carragenina/metabolismo , Digestión , Aditivos Alimentarios/metabolismo , Análisis de los Alimentos , Humanos
10.
PLoS One ; 13(10): e0204471, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30321179

RESUMEN

The exact route of iron through the kidney and its regulation during iron overload are not completely elucidated. Under physiologic conditions, non-transferrin and transferrin bound iron passes the glomerular filter and is reabsorbed through kidney epithelial cells, so that hardly any iron is found in the urine. To study the route of iron reabsorption through the kidney, we analyzed the location and regulation of iron metabolism related proteins in kidneys of mice with iron overload, elicited by iron dextran injections. Transferrin Receptor 1 was decreased as expected, following iron overload. In contrast, the multi-ligand hetero-dimeric receptor-complex megalin/cubilin, which also mediates the internalization of transferrin, was highly up-regulated. Moreover, with increasing iron, intracellular ferritin distribution shifted in renal epithelium from an apical location to a punctate distribution throughout the epithelial cells. In addition, in contrast to many other tissues, the iron exporter ferroportin was not reduced by iron overload in the kidney. Iron accumulated mainly in interstitial macrophages, and more prominently in the medulla than in the cortex. This suggests that despite the reduction of Transferrin Receptor 1, alternative pathways may effectively mediate re-absorption of iron that cycles through the kidney during parenterally induced iron-overload. The most iron consuming process of the body, erythropoiesis, is regulated by the renal erythropoietin producing cells in kidney interstitium. We propose, that the efficient re-absorption of iron by the kidney, also during iron overload enables these cells to sense systemic iron and regulate its usage based on the systemic iron state.


Asunto(s)
Transporte Biológico/fisiología , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Riñón/metabolismo , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Ferritinas/metabolismo , Espacio Intracelular/metabolismo , Sobrecarga de Hierro/patología , Complejo Hierro-Dextran , Riñón/patología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Transferrina/metabolismo , Bazo/metabolismo , Bazo/patología
11.
Sci Rep ; 8(1): 5118, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29572489

RESUMEN

Iron is essential for growth and proliferation of mammalian cells. The maintenance of cellular iron homeostasis is regulated by iron regulatory proteins (IRPs) through binding to the cognate iron-responsive elements in target mRNAs and thereby regulating the expression of target genes. Irp1 or Irp2-null mutation is known to reduce the cellular iron level by decreasing transferrin receptor 1 and increasing ferritin. Here, we report that Irp1 or Irp2-null mutation also causes downregulation of frataxin and IscU, two of the core components in the iron-sulfur cluster biogenesis machinery. Interestingly, while the activities of some of iron-sulfur cluster-containing enzymes including mitochondrial aconitase and cytosolic xanthine oxidase were not affected by the mutations, the activities of respiratory chain complexes were drastically diminished resulting in mitochondrial dysfunction. Overexpression of human ISCU and frataxin in Irp1 or Irp2-null cells was able to rescue the defects in iron-sulfur cluster biogenesis and mitochondrial quality. Our results strongly suggest that iron regulatory proteins regulate the part of iron sulfur cluster biogenesis tailored specifically for mitochondrial electron transport chain complexes.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteína 1 Reguladora de Hierro/deficiencia , Proteína 2 Reguladora de Hierro/deficiencia , Proteínas de Unión a Hierro/biosíntesis , Animales , Embrión de Mamíferos/patología , Ferritinas/metabolismo , Fibroblastos/patología , Humanos , Ratones , Ratones Noqueados , Mitocondrias/patología , Mutación , Frataxina
12.
Food Funct ; 9(3): 1344-1352, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29469913

RESUMEN

Carrageenan (CGN), a family of marine polysaccharides isolated from seaweeds, has been at the heart of considerable debate in recent years. To date, CGN is generally recognized as safe based on a history of safe use, various acute toxicology studies and some recent chronic toxicology tests. This review offers readers an overview of evidence on CGN characteristics and digestive fate that highlight various gaps in our understanding. Specifically, three unresolved gaps are identified. Firstly, little information can be found on the current levels of public exposure to CGN. Secondly, the link between CGN physicochemical properties, its impact on digestive proteolysis, the colon microbiome and inflammation are yet to be fully resolved. Thirdly, scant scientific evidence exists on the differential digestive fate of CGN in the gut of liable and predisposed populations, such as elderly people or IBD patients. Altogether, revisiting the scientific evidence indicates that more research is needed to elucidate the possibility that continued exposure to increasing levels of CGN in the human diet may compromise human health and well-being.


Asunto(s)
Carragenina/efectos adversos , Carragenina/metabolismo , Extractos Vegetales/efectos adversos , Extractos Vegetales/metabolismo , Algas Marinas/metabolismo , Animales , Carragenina/química , Digestión , Aditivos Alimentarios/efectos adversos , Aditivos Alimentarios/química , Aditivos Alimentarios/metabolismo , Humanos , Extractos Vegetales/química , Algas Marinas/química
13.
Blood ; 131(3): 342-352, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29074498

RESUMEN

Ferritin turnover plays a major role in tissue iron homeostasis, and ferritin malfunction is associated with impaired iron homeostasis and neurodegenerative diseases. In most eukaryotes, ferritin is considered an intracellular protein that stores iron in a nontoxic and bioavailable form. In insects, ferritin is a classically secreted protein and plays a major role in systemic iron distribution. Mammalian ferritin lacks the signal peptide for classical endoplasmic reticulum-Golgi secretion but is found in serum and is secreted via a nonclassical lysosomal secretion pathway. This study applied bioinformatics and biochemical tools, alongside a protein trafficking mouse models, to characterize the mechanisms of ferritin secretion. Ferritin trafficking via the classical secretion pathway was ruled out, and a 2:1 distribution of intracellular ferritin between membrane-bound compartments and the cytosol was observed, suggesting a role for ferritin in the vesicular compartments of the cell. Focusing on nonclassical secretion, we analyzed mouse models of impaired endolysosomal trafficking and found that ferritin secretion was decreased by a BLOC-1 mutation but increased by BLOC-2, BLOC-3, and Rab27A mutations of the cellular trafficking machinery, suggesting multiple export routes. A 13-amino-acid motif unique to ferritins that lack the secretion signal peptide was identified on the BC-loop of both subunits and plays a role in the regulation of ferritin secretion. Finally, we provide evidence that secretion of iron-rich ferritin was mediated via the multivesicular body-exosome pathway. These results enhance our understanding of the mechanism of ferritin secretion, which is an important piece in the puzzle of tissue iron homeostasis.


Asunto(s)
Ferritinas/metabolismo , Vesículas Secretoras/metabolismo , Secuencias de Aminoácidos , Animales , Biomarcadores/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Exosomas/metabolismo , Exosomas/ultraestructura , Ferritinas/sangre , Ferritinas/química , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7
14.
Reprod Fertil Dev ; 29(10): 2005-2010, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28063464

RESUMEN

Mitochondrial ferritin (FtMt) is a functional ferritin targeted to mitochondria that is highly expressed in the testis. To investigate the role of FtMt in the testis we set up a series of controlled matings between FtMt gene-deletion mice (FtMt-/-) with FtMt+/+ mice. We found that the number of newborns per litter and the fertility rate were strongly reduced for the FtMt-/- males, but not for the females, indicating that FtMt has an important role for male fertility. The morphology of the testis and of the spermatozoa of FtMt-/- mice was normal and we did not detect alterations in sperm parameters or in oxidative stress indices. In contrast, we observed that the cauda epididymides of FtMt-/- mice were significantly lighter and contained a lower number of spermatozoa compared with the controls. Also, the ATP content of FtMt-/- spermatozoa was found to be lower than that of FtMt+/+ spermatozoa. These data show that FtMt contributes to sperm epididymis maturation and to male fertility.


Asunto(s)
Ferritinas/genética , Fertilidad/genética , Infertilidad Masculina/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Animales , Ferritinas/metabolismo , Infertilidad Masculina/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo/genética , Motilidad Espermática/genética , Espermatozoides/metabolismo , Testículo/metabolismo
15.
Mol Nutr Food Res ; 61(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27718308

RESUMEN

SCOPE: The objective of this study was to interrogate two mechanisms by which commercial Carrageenans (E407) (CGN) may adversely affect human health: (i) Through modification of gastric proteolysis and (ii) Through affecting gut epithelial structure and function. METHODS AND RESULTS: Three commercial CGN samples with distinct zeta-potentials (stable at the pH range of 3-7 and varied with physiological levels of CaCl2 ) were mixed with milk, soy or egg protein isolates, then subjected to a semi-dynamic in vitro digestion model and analyzed by SDS-PAGE. This revealed varying levels of interference with gastric digestive proteolysis and a significant decrease in pepsin activity. Further, a Caco-2 cell model was used to explore various effects of physiologically digested CGN (pdCGN) on various epithelial cell functions and characteristics. Samples of pdCGN (0.005-0.5 mg/mL) affected the epithelial barrier function, including redistribution of the tight-junction protein Zonula Occludens (Zo)-1, changes in cellular F-actin architecture and increased monolayer permeability to the transfer of macromolecules. Moreover, pdCGN induced elevation in the levels of the pro-inflammatory IL-8 receptor CXCR1. CONCLUSION: This work raises the possibility that CGN may reduce protein and peptide bioaccessibility, disrupt normal epithelial function, promote intestinal inflammation, and consequently compromise consumer health.


Asunto(s)
Carragenina/efectos adversos , Carragenina/farmacocinética , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/citología , Células CACO-2 , Carragenina/química , Digestión , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Aditivos Alimentarios/efectos adversos , Aditivos Alimentarios/farmacocinética , Humanos , Mucosa Intestinal/efectos de los fármacos , Proteolisis , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
16.
PLoS One ; 11(10): e0164183, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27711215

RESUMEN

Hepcidin is the key regulator of systemic iron availability that acts by controlling the degradation of the iron exporter ferroportin. It is expressed mainly in the liver and regulated by iron, inflammation, erythropoiesis and hypoxia. The various agents that control its expression act mainly via the BMP6/SMAD signaling pathway. Among them are exogenous heparins, which are strong hepcidin repressors with a mechanism of action not fully understood but that may involve the competition with the structurally similar endogenous Heparan Sulfates (HS). To verify this hypothesis, we analyzed how the overexpression of heparanase, the HS degrading enzyme, modified hepcidin expression and iron homeostasis in hepatic cell lines and in transgenic mice. The results showed that transient and stable overexpression of heparanase in HepG2 cells caused a reduction of hepcidin expression and of SMAD5 phosphorylation. Interestingly, the clones showed also altered level of TfR1 and ferritin, indices of a modified iron homeostasis. The heparanase transgenic mice showed a low level of liver hepcidin, an increase of serum and liver iron with a decrease in spleen iron content. The hepcidin expression remained surprisingly low even after treatment with the inflammatory LPS. The finding that modification of HS structure mediated by heparanase overexpression affects hepcidin expression and iron homeostasis supports the hypothesis that HS participate in the mechanisms controlling hepcidin expression.


Asunto(s)
Regulación de la Expresión Génica , Glucuronidasa/genética , Hepcidinas/genética , Homeostasis , Hierro/metabolismo , Animales , Proteína Morfogenética Ósea 6/metabolismo , Proteína Morfogenética Ósea 6/farmacología , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Heparina/farmacología , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Interleucina-6/farmacología , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo
18.
Front Pharmacol ; 5: 194, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25202274

RESUMEN

Epithelial barriers are found in many tissues such as the intestine, kidney and brain where they separate the external environment from the body or a specific compartment from its periphery. Due to the tight junctions that connect epithelial barrier-cells (EBCs), the transport of compounds takes place nearly exclusively across the apical or basolateral membrane, the cell-body and the opposite membrane of the polarized EBC, and is regulated on numerous levels including barrier-specific adapted trafficking-machineries. Iron is an essential element but toxic at excess. Therefore, all iron-requiring organisms tightly regulate iron concentrations on systemic and cellular levels. In contrast to most cell types that control just their own iron homeostasis, EBCs also regulate homeostasis of the compartment they enclose or the body as a whole. Iron is transported across EBCs by specialized transporters such as the transferrin receptor and ferroportin. Recently, the iron storage protein ferritin was also attributed a role in the regulation of systemic iron homeostasis and we gathered evidence from the literature and original data that ferritin is polarized in EBC, suggesting also a role for ferritin in iron trafficking across EBCs.

19.
J Biol Chem ; 289(4): 2318-30, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24302717

RESUMEN

There is compelling evidence that aminoglycoside (AG) antibiotics can induce the mammalian ribosome to suppress disease-causing nonsense mutations and partially restore the expression of functional proteins. However, prolonged AG treatment can cause detrimental side effects in patients, including most prominently, ototoxicity. Recent mechanistic discussions have considered the relative contributions of mitochondrial and cytoplasmic protein synthesis inhibition to AG-induced ototoxicity. We show that AGs inhibit mitochondrial protein synthesis in mammalian cells and perturb cell respiration, leading to a time- and dose-dependent increase in superoxide overproduction and accumulation of free ferrous iron in mitochondria caused by oxidative damage of mitochondrial aconitase, ultimately leading to cell apoptosis via the Fenton reaction. These deleterious effects increase with the increased potency of AG to inhibit the mitochondrial rather than cytoplasmic protein synthesis, which in turn correlates with their ototoxic potential in both murine cochlear explants and the guinea pig in vivo. The deleterious effects of AGs were alleviated in synthetic derivatives specially designed for the treatment of genetic diseases caused by nonsense mutations and possessing low affinity toward mitochondrial ribosomes. This work highlights the benefit of a mechanism-based drug redesign strategy that can maximize the translational value of "readthrough therapy" while mitigating drug-induced side effects. This approach holds promise for patients suffering from genetic diseases caused by nonsense mutations.


Asunto(s)
Aminoglicósidos/farmacología , Citoplasma/metabolismo , Mitocondrias/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Ribosomas/metabolismo , Aminoglicósidos/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Cóclea/metabolismo , Relación Dosis-Respuesta a Droga , Enfermedades Genéticas Congénitas/tratamiento farmacológico , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Cobayas , Células HeLa , Humanos , Ratones , Proteínas Mitocondriales/biosíntesis , Consumo de Oxígeno/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/efectos adversos , Especies Reactivas de Oxígeno/metabolismo
20.
BMC Med ; 11: 185, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23968282

RESUMEN

BACKGROUND: Over the last few years, accumulating data have implicated a role for ferritin as a signaling molecule and direct mediator of the immune system. Hyperferritinemia is associated with a multitude of clinical conditions and with worse prognosis in critically ill patients. DISCUSSION: There are four uncommon medical conditions characterized by high levels of ferritin, namely the macrophage activation syndrome (MAS), adult onset Still's disease (AOSD), catastrophic antiphospholipid syndrome (cAPS) and septic shock, that share a similar clinical and laboratory features, and also respond to similar treatments, suggesting a common pathogenic mechanism. Ferritin is known to be a pro-inflammatory mediator inducing expression of pro-inflammatory molecules, yet it has opposing actions as a pro-inflammatory and as an immunosuppressant. We propose that the exceptionally high ferritin levels observed in these uncommon clinical conditions are not just the product of the inflammation but rather may contribute to the development of a cytokine storm. SUMMARY: Here we review and compare four clinical conditions and the role of ferritin as an immunomodulator. We would like to propose including these four conditions under a common syndrome entity termed "Hyperferritinemic Syndrome".


Asunto(s)
Síndrome Antifosfolípido/sangre , Ferritinas/sangre , Síndrome de Activación Macrofágica/sangre , Choque Séptico/sangre , Enfermedad de Still del Adulto/sangre , Animales , Síndrome Antifosfolípido/diagnóstico , Síndrome Antifosfolípido/epidemiología , Catarata/sangre , Catarata/congénito , Catarata/diagnóstico , Catarata/epidemiología , Enfermedad Catastrófica/epidemiología , Humanos , Mediadores de Inflamación/sangre , Trastornos del Metabolismo del Hierro/sangre , Trastornos del Metabolismo del Hierro/congénito , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/epidemiología , Síndrome de Activación Macrofágica/diagnóstico , Síndrome de Activación Macrofágica/epidemiología , Choque Séptico/diagnóstico , Choque Séptico/epidemiología , Enfermedad de Still del Adulto/diagnóstico , Enfermedad de Still del Adulto/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA