Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biometals ; 37(4): 955-969, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38483766

RESUMEN

Iron is an essential element for the normal functioning of living organisms, but excessive iron deposition can lead to organ damage. This study aims to investigate the interaction between the endoplasmic reticulum stress signaling pathway and the PI3K/AKT/mTOR signaling pathway in liver injury induced by iron overload in chicks. Rspectively, 150 one-day-old broilers were divided into three groups and supplemented with 50 (C), 500 (E1), and 1000 (E2) mg ferrous sulfate monohydrate/kg in the basal diet. Samples were taken after continuous feeding for 14 days. The results showed that iron overload could upregulate the levels of ALT and AST. Histopathological examination revealed bleeding in the central vein of the liver accompanied by inflammatory cell infiltration. Hoechst staining showed that the iron overload group showed significant bright blue fluorescence, and ultrastructural observations showed chromatin condensation as well as mitochondrial swelling and cristae disorganization in the iron overload group. RT-qPCR and Western blot results showed that iron overload upregulated the expression of Bax, Caspase-3, Caspase-9, GRP78, GRP94, P-PERK, ATF4, eIF2α, IRE1, and ATF6, while downregulating the expression of Bcl-2 and the PI3K/AKT/mTOR pathway. XBP-1 splicing experiment showed significant splicing of XBP-1 gene after iron overload. PCA and correlation analysis suggested a potential association between endoplasmic reticulum stress, the PI3K/AKT/mTOR signaling pathway, and liver injury in chicks. In summary, iron overload can induce cell apoptosis and liver injury by affecting endoplasmic reticulum stress and the PI3K/AKT/mTOR signaling pathway.


Asunto(s)
Pollos , Estrés del Retículo Endoplásmico , Sobrecarga de Hierro , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/metabolismo , Hígado/patología
2.
Ecotoxicol Environ Saf ; 275: 116241, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38522287

RESUMEN

Iron overload occurs due to excessive iron intake compared to the body's demand, leading to iron deposition and impairment of multiple organ functions. Our previous study demonstrated that chronic oral administration of ferric citrate (FC) caused colonic inflammatory injury. However, the precise mechanism underlying this inflammatory response remains unclear. The current study aims to investigate the mechanism by which iron overload induced by FC exposure leads to colonic inflammation. To accomplish this, mice were orally exposed to three different concentrations of FC (71 mg/kg/bw (L), 143 mg/kg/bw (M) and 286 mg/kg/bw (H)) for continuous 16 weeks, with the control group receiving ultrapure water (C). Exposure to FC caused disturbances in the excretory system, altered colonic flora alpha diversity, and enriched pathogenic bacteria, such as Mucispirillum, Helicobacter, Desulfovibrio, and Shigella. These changes led to structural disorders of the colonic flora and an inflammatory response phenotype characterized by inflammatory cells infiltration, atrophy of intestinal glands, and irregular thickening of the intestinal wall. Mechanistic studies revealed that FC-exposure activated the NF-κB signaling pathway by up-regulating TLR4, MyD88, and NF-κB mRNA levels and protein expression. This activation resulted in increased production of pro-inflammatory cytokines, further contributing to the colonic inflammation. Additionally, in vitro experiments in SW480 cells confirmed the activation of NF-κB signaling pathway by FC exposure, consistent with the in vivo findings. The significance of this study lies in its elucidation of the mechanism by which iron overload caused by FC exposure leads to colonic inflammation. By identifying the role of pathogenic bacteria and the NF-κB signaling pathway, this study could potentially offer a crucial theoretical foundation for the research on iron overload, as well as provide valuable insights for clinical iron supplementation.


Asunto(s)
Compuestos Férricos , Sobrecarga de Hierro , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Inflamación/inducido químicamente , Inflamación/patología , Sobrecarga de Hierro/patología , Hierro/metabolismo
3.
Redox Biol ; 60: 102616, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36746004

RESUMEN

OBJECTIVE: Rheumatoid arthritis is an inflammatory joint disease in which synovial iron deposition has been described. Transferrin receptor 2 (Tfr2) represents a critical regulator of systemic iron levels. Loss of Tfr2 function in humans and mice results in iron overload. As iron contributes to inflammatory processes, we investigated whether Tfr2-deletion affects the pathogenesis of inflammatory arthritis in an iron-dependent manner. METHODS: Using a global and conditional genetic disruption of Tfr2, we assessed the relevance of Tfr2 in K/BxN serum-transfer arthritis (STA) and macrophage polarization. RESULTS: Male Tfr2-/- mice subjected to STA developed pronounced joint swelling, and bone erosion as compared to Tfr2+/+ littermate-controls (P < 0.01). Furthermore, an increase of neutrophils and macrophages/monocytes was observed in the inflammatory infiltrate within the paws of Tfr2-/- mice. To elucidate whether Tfr2 in myeloid cells has a direct role in the pathogenesis of arthritis or whether the effects were mediated via the systemic iron overload, we induced STA in Tfr2fl/fl-LysMCre + mice, which showed normal iron-loading. Cre + female mice displayed increased disease development compared to Cre-controls. As macrophages regulate iron availability and innate immunity, we hypothesized that Tfr2-deficiency would polarize macrophages toward a pro-inflammatory state (M1) that contributes to arthritis progression. In response to IFN-γ stimulation, Tfr2-/- macrophages showed increased expression of M1-like cytokines, IFN-γ-target genes, nitric-oxide production, and prolonged STAT1 activation compared to Tfr2+/+ macrophages (P < 0.01), while pre-treatment with ruxolitinib abolished Tfr2-driven M1-like polarization. CONCLUSION: Taken together, these findings suggest a protective role of Tfr2 in macrophages on the progression of arthritis via suppression of M1-like polarization.


Asunto(s)
Artritis , Sobrecarga de Hierro , Humanos , Ratones , Masculino , Femenino , Animales , Ratones Noqueados , Hierro/metabolismo , Sobrecarga de Hierro/patología , Macrófagos/metabolismo , Artritis/metabolismo , Receptores de Transferrina/genética
4.
Pharm Biol ; 61(1): 37-49, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36573499

RESUMEN

CONTEXT: Gallic acid (GA) and lecithin showed important roles in antioxidant and drug delivery, respectively. A complex synthesized from GA and soybean lecithin (SL-GAC), significantly improved bioavailability of GA and pharmacological activities. However, the antioxidant activity of SL-GAC and its effect on iron-overload-induced liver injury remains unexplored. OBJECTIVE: This study investigates the antioxidant properties of SL-GAC in vitro and in mice, and its remediating effects against liver injury by iron-overloaded. MATERIALS AND METHODS: In vitro, free radical scavenging activity, lipid peroxidation inhibition, and ferric reducing power of SL-GAC were measured by absorbance photometry. In vivo, C57BL/6J mice were randomized into 4 groups: control, iron-overloaded, iron-overloaded + deferoxamine, and iron-overloaded + SL-GAC. Treatments with deferoxamine (150 mg/kg/intraperitioneally) and SL-GAC (200 mg/kg/orally) were given to the desired groups for 12 weeks, daily. Iron levels, oxidative stress, and biochemical parameters were determined by histopathological examination and molecular biological techniques. RESULTS: In vitro, SL-GAC showed DPPH and ABTS free radicals scavenging activity with IC50 values equal to 24.92 and 128.36 µg/mL, respectively. In C57BL/6J mice, SL-GAC significantly reduced the levels of serum iron (22.82%), liver iron (50.29%), aspartate transaminase (25.97%), alanine transaminase (38.07%), gamma glutamyl transferase (42.11%), malondialdehyde (19.82%), total cholesterol (45.96%), triglyceride (34.90%), ferritin light chain (18.51%) and transferrin receptor (27.39%), while up-regulated the levels of superoxide dismutase (24.69%), and glutathione (11.91%). CONCLUSIONS: These findings encourage the use of SL-GAC to treat liver injury induced by iron-overloaded. Further in vivo and in vitro studies are needed to validate its potential in clinical medicine.


Asunto(s)
Sobrecarga de Hierro , Hepatopatías , Ratones , Animales , Lecitinas/metabolismo , Lecitinas/farmacología , Lecitinas/uso terapéutico , Antioxidantes/uso terapéutico , Glycine max , Ácido Gálico/farmacología , Deferoxamina/farmacología , Deferoxamina/metabolismo , Deferoxamina/uso terapéutico , Ratones Endogámicos C57BL , Hepatopatías/tratamiento farmacológico , Estrés Oxidativo , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado , Hierro/metabolismo , Peroxidación de Lípido
5.
Free Radic Biol Med ; 190: 234-246, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35981695

RESUMEN

Iron overload is a common phenomenon in the elderly population. Many clinical studies have indicated an association between iron overload and the incidence and pathological progression of intervertebral disc degeneration (IVDD). However, the role and underlying mechanism by which iron participates in the progression of IVDD has not yet been reported. In the present study, we aimed to elucidate the connection between iron overload and IVDD, and explore the underlying mechanisms of disease. Firstly, a clinical epidemiology study was conducted and revealed that iron overload is an independent risk factor for human IVDD. To elucidate the role of iron overload in IVDD, an iron overload mouse model was established, and we observed that iron overload promoted IVDD and cartilage endplate degeneration in a dose dependent manner. Endplate chondrocytes were further isolated and treated with FAC to mimic iron overload in vitro. Excess iron significantly promoted mineralization of endplate chondrocytes in addition to their degeneration via oxidative stress. Moreover, a high dose of excess iron promoted chondrocytes ferroptosis. An iron chelator (DFO), an antioxidant (NAC) and a ferroptosis inhibitor (Fer-1) demonstrated effective inhibition of endplate chondrocyte degeneration induced by iron overload, and our in vivo studies further demonstrated that DFO, NAC and Fer-1 could rescue high dose iron-induced IVDD and cartilage endplate calcification. In conclusion, our results indicate that iron overload is strongly associated with the onset and development of IVDD via oxidative stress and ferroptosis. Inhibiting oxidative stress or ferroptosis could therefore be promising therapeutic strategies for IVDD induced by iron overload.


Asunto(s)
Ferroptosis , Degeneración del Disco Intervertebral , Sobrecarga de Hierro , Anciano , Animales , Condrocitos , Humanos , Degeneración del Disco Intervertebral/patología , Hierro/farmacología , Sobrecarga de Hierro/patología , Ratones , Estrés Oxidativo
6.
Semin Ultrasound CT MR ; 43(4): 364-370, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35738822

RESUMEN

Liver Iron content is best correlated to total body iron stores and is thus the organ of choice for evaluation in iron overload diseases. Liver biopsy was the historic standard for iron evaluation, but the evaluation is localized, comes with increased risks due to its invasiveness, and is costly. MRI is now widely used for liver iron evaluation. The superparamagnetic properties of iron cause a disturbance in magnetic resonance imaging, which can be evaluated with various techniques. These include signal intensity ratio (SIR), T2 relaxometry, T2* relaxometry, and Dixon-based solutions. Each of the methods has its own advantages and disadvantages, and factors such as availability, ease of use, accuracy, reproducibility, and cost can all play a role in the ultimate technique used for liver iron quantification. Quantitative susceptibility mapping, and ultrashort TE sequences are promising supplemental methods, but are primarily used as research sequences. These may become more clinically accepted in the near future. Dual energy CT is also being explored as an alternative but is still in the nascent stages. Overall, accurate liver iron concentration is feasible with the current tools available at most MR imaging centers and is highly valuable for evaluation of iron overload diseases.


Asunto(s)
Sobrecarga de Hierro , Hierro , Humanos , Sobrecarga de Hierro/diagnóstico por imagen , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/patología , Hígado/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Reproducibilidad de los Resultados
7.
J Cachexia Sarcopenia Muscle ; 13(2): 1250-1261, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35118832

RESUMEN

BACKGROUND: Iron excess has been proposed as an essential factor in skeletal muscle wasting. Studies have reported correlations between muscle iron accumulation and atrophy, either through ageing or by using experimental models of secondary iron overload. However, iron treatments performed in most of these studies induced an extra-pathophysiological iron overload, more representative of intoxication or poisoning. The main objective of this study was to determine the impact of iron excess closer to pathophysiological conditions on structural and metabolic adaptations (i) in differentiated myotubes and (ii) in skeletal muscle exhibiting oxidative (i.e. the soleus) or glycolytic (i.e. the gastrocnemius) metabolic phenotypes. METHODS: The impact of iron excess was assessed in both in vitro and in vivo models. Murine differentiated myotubes were exposed to ferric ammonium citrate (FAC) (i.e. 10 and 50 µM) for the in vitro component. The in vivo model was achieved by a single iron dextran subcutaneous injection (1 g/kg) in mice. Four months after the injection, soleus and gastrocnemius muscles were harvested for analysis. RESULTS: In vitro, iron exposure caused dose-dependent increases of iron storage protein ferritin (P < 0.01) and dose-dependent decreases of mRNA TfR1 levels (P < 0.001), which support cellular adaptations to iron excess. Extra-physiological iron treatment (50 µM FAC) promoted myotube atrophy (P = 0.018), whereas myotube size remained unchanged under pathophysiological treatment (10 µM FAC). FAC treatments, whatever the doses tested, did not affect the expression of proteolytic markers (i.e. NF-κB, MurF1, and ubiquitinated proteins). In vivo, basal iron content and mRNA TfR1 levels were significantly higher in the soleus compared with the gastrocnemius (+130% and +127%; P < 0.001, respectively), supporting higher iron needs in oxidative skeletal muscle. Iron supplementation induced muscle iron accumulation in the soleus and gastrocnemius muscles (+79%, P < 0.001 and +34%, P = 0.002, respectively), but ferritin protein expression only increased in the gastrocnemius (+36%, P = 0.06). Despite iron accumulation, muscle weight, fibre diameter, and myosin heavy chain distribution remained unchanged in either skeletal muscle. CONCLUSIONS: Together, these data support that under pathophysiological conditions, skeletal muscle can protect itself from the related deleterious effects of excess iron.


Asunto(s)
Sobrecarga de Hierro , Atrofia Muscular , Animales , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Ratones , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Estrés Oxidativo
8.
Radiology ; 302(2): 345-354, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34783592

RESUMEN

Background Standardized manual region of interest (ROI) sampling strategies for hepatic MRI steatosis and iron quantification are time consuming, with variable results. Purpose To evaluate the performance of automatic MRI whole-liver segmentation (WLS) for proton density fat fraction (PDFF) and iron estimation (transverse relaxometry [R2*]) versus manual ROI, with liver biopsy as the reference standard. Materials and Methods This prospective, cross-sectional, multicenter study recruited participants with chronic liver disease who underwent liver biopsy and chemical shift-encoded 3.0-T MRI between January 2017 and January 2021. Biopsy evaluation included histologic grading and digital pathology. MRI liver sampling strategies included manual ROI (two observers) and automatic whole-liver (deep learning algorithm) segmentation for PDFF- and R2*-derived measurements. Agreements between segmentation methods were measured using intraclass correlation coefficients (ICCs), and biases were evaluated using Bland-Altman analyses. Linear regression analyses were performed to determine the correlation between measurements and digital pathology. Results A total of 165 participants were included (mean age ± standard deviation, 55 years ± 12; 96 women; 101 of 165 participants [61%] with nonalcoholic fatty liver disease). Agreements between mean measurements were excellent, with ICCs of 0.98 for both PDFF and R2*. The median bias was 0.5% (interquartile range, -0.4% to 1.2%) for PDFF and 2.7 sec-1 (interquartile range, 0.2-5.3 sec-1) for R2* (P < .001 for both). Margins of error were lower for WLS than ROI-derived parameters (-0.03% for PDFF and -0.3 sec-1 for R2*). ROI and WLS showed similar performance for steatosis (ROI AUC, 0.96; WLS AUC, 0.97; P = .53) and iron overload (ROI AUC, 0.85; WLS AUC, 0.83; P = .09). Correlations with digital pathology were high (P < .001) between the fat ratio and PDFF (ROI r = 0.89; WLS r = 0.90) and moderate (P < .001) between the iron ratio and R2* (ROI r = 0.65; WLS r = 0.64). Conclusion Proton density fat fraction and transverse relaxometry measurements derived from MRI automatic whole-liver segmentation (WLS) were accurate for steatosis and iron grading in chronic liver disease and correlated with digital pathology. Automated WLS estimations were higher, with a lower margin of error than manual region of interest estimations. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Moura Cunha and Fowler in this issue.


Asunto(s)
Aprendizaje Profundo , Sobrecarga de Hierro/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Enfermedad del Hígado Graso no Alcohólico/diagnóstico por imagen , Biopsia , Enfermedad Crónica , Estudios Transversales , Femenino , Humanos , Sobrecarga de Hierro/patología , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/patología , Estudios Prospectivos
9.
Sci Rep ; 11(1): 12581, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131221

RESUMEN

Deferasirox (DFX) is the newest among three different chelators available to treat iron overload in iron-loading anaemias, firstly released as Dispersible Tablets (DT) and more recently replaced by Film-Coated Tablets (FCT). In this retrospective observational study, pharmacokinetics, pharmacodynamics, and safety features of DFX treatment were analyzed in 74 patients that took both formulations subsequently under clinical practice conditions. Bioavailability of DFX FCT compared to DT resulted higher than expected [Cmax: 99.5 (FCT) and 69.7 (DT) µMol/L; AUC: 1278 (FCT) and 846 (DT), P < 0.0001]. DFX FCT was also superior in scalability among doses. After one year of treatment for each formulation, no differences were observed between the treatments in the overall iron overload levels; however, DFX FCT but not DT showed a significant dose-response correlation [Spearman r (dose-serum ferritin variation): - 0.54, P < 0.0001]. Despite being administered at different dosages, the long-term safety profile was not different between formulations: a significant increase in renal impairment risk was observed for both treatments and it was reversible under strict monitoring (P < 0.002). Altogether, these data constitute a comprehensive comparison of DFX formulations in thalassaemia and other iron-loading anaemias, confirming the effectiveness and safety characteristics of DFX and its applicability for treatment tailoring.


Asunto(s)
Anemia/tratamiento farmacológico , Deferasirox/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Talasemia/tratamiento farmacológico , Adulto , Anemia/sangre , Anemia/epidemiología , Anemia/patología , Terapia por Quelación/tendencias , Deferasirox/farmacocinética , Femenino , Ferritinas/sangre , Humanos , Hierro/sangre , Hierro/metabolismo , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/farmacocinética , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/epidemiología , Sobrecarga de Hierro/patología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Talasemia/sangre , Talasemia/epidemiología , Talasemia/patología
10.
Exp Physiol ; 106(8): 1772-1784, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34148259

RESUMEN

NEW FINDINGS: What is the central question of this study? The current literature indicates that oxidative stress plays a major role in iron overload. Although exercise is a well-established approach to treat/prevent cardiovascular diseases, its effects on iron overload are not known. What is the main finding and its importance? Moderate-intensity aerobic training had benefits in a rodent model of iron-overload cardiomyopathy by improving the antioxidant capacity of the heart. After further confirmation by translational and clinical studies, we should consider using this non-pharmacological, highly accessible and easily executable adjuvant approach allied to other therapies to improve the quality of life of iron-overloaded patients. ABSTRACT: Iron is an essential micronutrient for several life processes, but its excess can damage organs owing to oxidative stress, with cardiomyopathy being the leading cause of death in iron-overloaded patients. Although exercise has long been considered as a cardioprotective tool, its effects on iron overload are not known. This study was designed to investigate the effects of moderate-intensity aerobic training in rats previously submitted to chronic iron overload. Wistar rats received i.p. injections of iron dextran (100 mg/kg, 5 days/week for 4 weeks); thereafter, the rats were kept sedentary or exercised (60 min/day, progressive aerobic training, 60-70% of maximal speed, 5 days/week on a treadmill) for 8 weeks. At the end of the experimental period, haemodynamics were recorded and blood samples, livers and hearts harvested. Myocardial mechanics of papillary muscles were assessed in vitro, and cardiac remodelling was evaluated by histology and immunoblotting. Iron overload led to liver iron deposition, liver fibrosis and increased serum alanine aminotransferase and aspartate aminotransferase. Moreover, cardiac iron accumulation was accompanied by impaired myocardial mechanics, increased cardiac collagen type I and lipid peroxidation (TBARS), and release of creatine phosphokinase-MB to the serum. Although exercise did not influence iron levels, tissue injury markers were significantly reduced. Likewise, myocardial contractility and inotropic responsiveness were improved in exercised rats, in association with an increase in the endogenous antioxidant enzyme catalase. In conclusion, moderate-intensity aerobic exercise was associated with attenuated oxidative stress and cardiac damage in a rodent model of iron overload, thereby suggesting its potential role as a non-pharmacological adjuvant therapy for iron-overload cardiomyopathy.


Asunto(s)
Sobrecarga de Hierro , Calidad de Vida , Animales , Corazón , Humanos , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Miocardio/metabolismo , Estrés Oxidativo , Ratas , Ratas Wistar
11.
Biomarkers ; 26(5): 425-433, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33843382

RESUMEN

Background: Iron-overload is a well-known cause for the development of chronic liver diseases and known to induce DNA damage.Material and methods: The protective effect of argan oil (AO) from the Argania spinosa fruit and olive oil (OO) (6% AO or OO for 28 days) was evaluated on a mouse model of iron overload (3.5mg Fe2+/liter) and in human fibroblasts where DNA damage was induced via culture under hyperoxia (40% oxygen).Results: Iron treatment induced DNA damage in liver tissue while both oils were able to decrease it. We confirmed this effect in vitro in MRC-5 fibroblasts under hyperoxia. A cell-free ABTS assay suggested that improvement of liver toxicity by both oils might depend on a high content in tocopherol, phytosterol and polyphenol compounds known for their antioxidant potential. The antioxidant effect of AO was confirmed in fibroblasts by reduced intracellular peroxide levels after hyperoxia. However, we could not find a significant decrease of genes encoding pro-inflammatory cytokines (TNFα, IL-6, IL-1ß, COX-2) or senescence markers (p16 and p21) for the oils in mouse liver.Conclusion: We found a striking effect of AO by ameliorating DNA damage after iron overload in a mouse liver model and in human fibroblasts by hyperoxia adding compelling evidence to the protective mechanisms of AO and OO.


Asunto(s)
Antioxidantes/farmacología , Daño del ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Sobrecarga de Hierro/tratamiento farmacológico , Hígado/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Aceites de Plantas/farmacología , Animales , Hipoxia de la Célula , Línea Celular , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Mediadores de Inflamación/metabolismo , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Aceite de Oliva/farmacología
12.
Hematology ; 26(1): 123-133, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33491605

RESUMEN

OBJECTIVES: We aim to explore and analyze the related influencing factors of liver and cardiac iron overload in MDS patients detected by magnetic resonance imaging (MRI). METHODS: We have detected cardiac T2* and liver T2* by MRI in 105 MDS patients. Among them, 20 patients accepted MRI examination before and after iron chelation therapy (ICT). Results: We found that adjusted ferritin (ASF) was significantly correlated with liver T2* and cardiac T2*. RBC transfusion volume, brain natriuretic peptide (BNP) and age were the related factors of cardiac T2*, while RBC transfusion volume and erythropoietin (EPO) were related factors of liver T2*. After ICT, the changes of ASF and liver T2* were earlier than cardiac T2*. Chronic hepatitis but virus copy normal's has no significant effect on liver iron deposition. CONCLUSION: These results showed special attention should be paid to these related influencing factors of liver and cardiac T2* expression when we evaluated iron overload and detected the efficacy of ICT in MDS patients.


Asunto(s)
Corazón/diagnóstico por imagen , Sobrecarga de Hierro/diagnóstico por imagen , Sobrecarga de Hierro/etiología , Hígado/diagnóstico por imagen , Adulto , Anciano , Anciano de 80 o más Años , Transfusión de Eritrocitos , Femenino , Humanos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/patología , Sobrecarga de Hierro/terapia , Hígado/patología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Miocardio/patología , Factores de Riesgo , Adulto Joven
13.
Mol Nutr Food Res ; 65(4): e2000772, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33325123

RESUMEN

SCOPE: Accumulating evidence indicates that micronutrients are related to metabolic diseases. However, comparatively less attention has been devoted to their influence on each other during the development of metabolic diseases. To investigate the underlying mechanisms, the effects of iron and vitamin D on pancreatic ß cell functions are examined. METHODS AND RESULTS: Iron overload is induced in INS-1 rat insulinoma pancreatic ß cells and it is found that iron overload dramatically reduce expression of the vitamin D receptor (VDR). Iron overload-induced ß cell dysfunction is rescued by 1,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ) cotreatment via restoration of VDR level and the consequent maintenance of Ca2+ homeostasis. Iron accumulation is also observed in the islets of 22-month-old C57BL/6 mice fed with a chow diet (1000 IU vitamin D3 per kg). In contrast, islet iron accumulation and hyperinsulinemia are ameliorated in mice fed with a vitamin D3 -supplemented diet (20 000 IU kg-1 ). CONCLUSION: The authors show that functional failure of ß cells due to iron accumulation is rescued by 1,25(OH)2 D3 , and iron overload significantly reduces VDR levels in ß cells. These results suggest that iron and vitamin D inversely influence pancreatic ß cell function.


Asunto(s)
Calcio/metabolismo , Colecalciferol/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Sobrecarga de Hierro/patología , Receptores de Calcitriol/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Homeostasis/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Hierro/metabolismo , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratas , Vitamina D/análogos & derivados , Vitamina D/farmacología
14.
Signal Transduct Target Ther ; 5(1): 138, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32732975

RESUMEN

Iron homeostasis is essential for health; moreover, hepcidin-deficiency results in iron overload in both hereditary hemochromatosis and iron-loading anemia. Here, we identified iron modulators by functionally screening hepcidin agonists using a library of 640 FDA-approved drugs in human hepatic Huh7 cells. We validated the results in C57BL/6J mice and a mouse model of hemochromatosis (Hfe-/- mice). Our screen revealed that the anti-rheumatoid arthritis drug auranofin (AUR) potently upregulates hepcidin expression. Interestingly, we found that canonical signaling pathways that regulate iron, including the Bmp/Smad and IL-6/Jak2/Stat3 pathways, play indispensable roles in mediating AUR's effects. In addition, AUR induces IL-6 via the NF-κB pathway. In C57BL/6J mice, acute treatment with 5 mg/kg AUR activated hepatic IL-6/hepcidin signaling and decreased serum iron and transferrin saturation. Whereas chronically treating male Hfe-/- mice with 5 mg/kg AUR activated hepatic IL-6/hepcidin signaling, decreasing systemic iron overload, but less effective in females. Further analyses revealed that estrogen reduced the ability of AUR to induce IL-6/hepcidin signaling in Huh7 cells, providing a mechanistic explanation for ineffectiveness of AUR in female Hfe-/- mice. Notably, high-dose AUR (25 mg/kg) induces ferroptosis and causes lipid peroxidation through inhibition of thioredoxin reductase (TXNRD) activity. We demonstrate the ferroptosis inhibitor ferrostatin significantly protects liver toxicity induced by high-dose AUR without comprising its beneficial effect on iron metabolism. In conclusion, our findings provide compelling evidence that TXNRD is a key regulator of ferroptosis, and AUR is a novel activator of hepcidin and ferroptosis via distinct mechanisms, suggesting a promising approach for treating hemochromatosis and hepcidin-deficiency related disorders.


Asunto(s)
Auranofina/farmacología , Ferroptosis/efectos de los fármacos , Hemocromatosis , Sobrecarga de Hierro , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Femenino , Ferroptosis/genética , Células HEK293 , Hemocromatosis/tratamiento farmacológico , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hemocromatosis/patología , Humanos , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética
15.
Rev Med Interne ; 41(11): 769-775, 2020 Nov.
Artículo en Francés | MEDLINE | ID: mdl-32682623

RESUMEN

Aceruloplasminemia is a rare iron-overload disease that should be better known by physicians. It is an autosomal recessive disorder due to mutations in ceruloplasmin gene causing systemic iron overload, including cerebral and liver parenchyma. The impairment of ferroxidase ceruloplasmin activity leads to intracellular iron retention leading aceruloplasminemia symptoms. Neurologic manifestations include cognitive impairment, ataxia, extrapyramidal syndrome, abnormal movements, and psychiatric-like syndromes. Physicians should search for aceruloplasminemia in several situations with high ferritin levels: microcytic anaemia, diabetes mellitus, neurological and psychiatric disorders. Diagnosis approach is based on the study of transferrin saturation and hepatic iron content evaluated by magnetic resonance imaging of the liver. Ceruloplasmin dosage is required in case of low transferrin saturation and high hepatic iron content and genetic testing is mandatory in case of serum ceruloplasmin defect. Neurological manifestations occur in the sixties decade and leads to disability. Iron chelators are widely used. Despite their efficacy on systemic and cerebral iron overload, iron chelators tolerance is poor. Early initiation of iron chelation therapy might prevent or slowdown neurodegeneration, highlighting the need for an early diagnosis but their clinical efficacy remains uncertain.


Asunto(s)
Ceruloplasmina/deficiencia , Trastornos del Metabolismo del Hierro/diagnóstico , Enfermedades Neurodegenerativas/diagnóstico , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Diagnóstico Diferencial , Humanos , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/complicaciones , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/terapia , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/patología , Enfermedades Neurodegenerativas/complicaciones , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/terapia , Trastornos Parkinsonianos/diagnóstico , Trastornos Parkinsonianos/etiología , Trastornos Parkinsonianos/metabolismo , Enfermedades Raras
16.
Nephrol Dial Transplant ; 35(6): 946-954, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32259248

RESUMEN

BACKGROUND: The iron-based phosphate binders, sucroferric oxyhydroxide (SFOH) and ferric citrate (FC), effectively lower serum phosphorus in clinical studies, but gastrointestinal iron absorption from these agents appears to differ. We compared iron uptake and tissue accumulation during treatment with SFOH or FC using experimental rat models. METHODS: Iron uptake was evaluated during an 8-h period following oral administration of SFOH, FC, ferrous sulphate (oral iron supplement) or control (methylcellulose vehicle) in rat models of anaemia, iron overload and inflammation. A 13-week study evaluated the effects of SFOH and FC on iron accumulation in different organs. RESULTS: In the pharmacokinetic experiments, there was a minimal increase in serum iron with SFOH versus control during the 8-h post-treatment period in the iron overload and inflammation rat models, whereas a moderate increase was observed in the anaemia model. Significantly greater increases (P < 0.05) in serum iron were observed with FC versus SFOH in the rat models of anaemia and inflammation. In the 13-week iron accumulation study, total liver iron content was significantly higher in rats receiving FC versus SFOH (P < 0.01), whereas liver iron content did not differ between rats in the SFOH and control groups. CONCLUSIONS: Iron uptake was higher from FC versus SFOH following a single dose in anaemia, iron overload and inflammation rat models and 13 weeks of treatment in normal rats. These observations likely relate to different physicochemical properties of SFOH and FC and suggest distinct mechanisms of iron absorption from these two phosphate binders.


Asunto(s)
Anemia/tratamiento farmacológico , Compuestos Férricos/administración & dosificación , Inflamación/tratamiento farmacológico , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/farmacocinética , Sacarosa/administración & dosificación , Administración Oral , Anemia/patología , Animales , Combinación de Medicamentos , Femenino , Inflamación/patología , Sobrecarga de Hierro/patología , Cinética , Masculino , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Distribución Tisular
17.
Aging (Albany NY) ; 11(21): 9846-9861, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31699955

RESUMEN

Iron homeostasis is critical for maintaining normal brain physiological functions, and its mis-regulation can cause neurotoxicity and play a part in the development of many neurodegenerative disorders. The high incidence of iron deficiency makes iron supplementation a trend, and ferric citrate is a commonly used iron supplement. In this study, we found that the chronic oral administration of ferric citrate (2.5 mg/day and 10 mg/day) for 16 weeks selectively induced iron accumulation in the corpus striatum (CPu), substantia nigra (SN) and hippocampus, which typically caused parkinsonism phenotypes in middle-aged mice. Histopathological analysis showed that apoptosis- and oxidative stress-mediated neurodegeneration and dopaminergic neuronal loss occurred in the brain, and behavioral tests showed that defects in the locomotor and cognitive functions of these mice developed. Our research provides a new perspective for ferric citrate as a food additive or in clinical applications and suggests a new potential approach to develop animal models for Parkinson's disease (PD).


Asunto(s)
Encéfalo/metabolismo , Compuestos Férricos/efectos adversos , Sobrecarga de Hierro/inducido químicamente , Trastornos Parkinsonianos/inducido químicamente , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Compuestos Férricos/administración & dosificación , Compuestos Férricos/metabolismo , Sobrecarga de Hierro/patología , Masculino , Ratones , Estrés Oxidativo , Trastornos Parkinsonianos/patología
18.
Oxid Med Cell Longev ; 2019: 2340392, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31781327

RESUMEN

It has been recognized that iron overload may harm the body's health. Vascular endothelial cells (VECs) are one of the main targets of iron overload injury, and the mechanism involved was thought to be related to the excessive generation of reactive oxygen species (ROS). However, the subcellular and temporal characteristics of ROS generation, potential downstream mechanisms, and target organelles in VECs injured by iron overload have not been expounded yet. In this study, we elucidated the abovementioned issues through both in vivo and in vitro experiments. Mice were fed pellet diets that were supplemented with iron for 4 consecutive months. Results showed that the thoracic aortic strips' endothelium-dependent dilation was significantly impaired and associated with inflammatory changes, noticeable under brown TUNEL-positive staining in microscopy analysis. In addition, the serum content of asymmetric dimethylarginine (ADMA) increased, whereas nitric oxide (NO) levels decreased. Furthermore, the dimethylarginine dimethylaminohydrolase II (DDAHII) expression and activity, as well as the phosphorylation of endothelial nitric oxide synthase (eNOS) in aortic tissue, were inhibited. Human umbilical vein endothelial cells were treated with 50 µM iron dextran for 48 hours, after which the cell viability, NO content, DDAHII expression and activity, and phosphorylation of eNOS decreased and lactate dehydrogenase and caspase-3 activity, ADMA content, and apoptotic cells significantly increased. After the addition of L-arginine (L-Arg) or pAD/DDAHII, the abovementioned changes were reversed. By dynamically detecting the changes of ROS generation in the cytoplasm and mitochondria and interfering with different aspects of signaling pathways, we have confirmed for the first time that excessive ROS originates from the cytoplasm and activates the ROS-induced ROS release (RIRR) mechanism, leading to mitochondrial dysfunction. Together, our data suggested that excessive free iron ions produced excess ROS in the cytoplasm. Thus, excess ROS create one vicious circle by activating the ADMA/eNOS/DDAHII/NO pathway and another vicious circle by activation of the RIRR mechanism, which, when combined, induce a ROS burst, resulting in mitochondrial dysfunction and damaged VECs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Sobrecarga de Hierro/metabolismo , Mitocondrias/metabolismo , Transducción de Señal , Amidohidrolasas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Sobrecarga de Hierro/patología , Ratones , Mitocondrias/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Especies Reactivas de Oxígeno/metabolismo
19.
Am J Pathol ; 189(9): 1814-1830, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31287995

RESUMEN

The liver secretes hepcidin (Hepc) into the bloodstream to reduce blood iron levels. Hepc accomplishes this by triggering degradation of the only known cellular iron exporter ferroportin in the gut, macrophages, and liver. We previously demonstrated that systemic Hepc knockout (HepcKO) mice, which have high serum iron, develop retinal iron overload and degeneration. However, it was unclear whether this is caused by high blood iron levels or, alternatively, retinal iron influx that would normally be regulated by retina-produced Hepc. To address this question, retinas of liver-specific and retina-specific HepcKO mice were studied. Liver-specific HepcKO mice had elevated blood and retinal pigment epithelium (RPE) iron levels and increased free (labile) iron levels in the retina, despite an intact blood-retinal barrier. This led to RPE hypertrophy associated with lipofuscin-laden lysosome accumulation. Photoreceptors also degenerated focally. In contrast, there was no change in retinal or RPE iron levels or degeneration in the retina-specific HepcKO mice. These data indicate that high blood iron levels can lead to retinal iron accumulation and degeneration. High blood iron levels can occur in patients with hereditary hemochromatosis or result from use of iron supplements or multiple blood transfusions. Our results suggest that high blood iron levels may cause or exacerbate retinal disease.


Asunto(s)
Hepcidinas/fisiología , Sobrecarga de Hierro/etiología , Hierro/metabolismo , Hígado/metabolismo , Retina/metabolismo , Degeneración Retiniana/etiología , Animales , Barrera Hematorretinal , Femenino , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
20.
Magn Reson Imaging ; 61: 267-272, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31128226

RESUMEN

Brain iron overload is chronic and slow progressing and plays an important role in the pathogenesis of neurodegenerative disorders. Magnetic resonance imaging (MRI) is a useful noninvasive tool for determining liver iron content, but it has not been proven to be adequate for evaluating brain iron overload. We evaluated the usefulness of MRI-derived parameters to determine brain iron concentration in ß-thalassemic mice and the effects of the membrane permeable iron chelator, deferiprone. Sixteen ß-thalassemic mice underwent 1.5T MRI of the brain that included a multiecho T2*-weighted sequence. Brain T2* values ranged from 28 to 31ms for thalassemic mice. For the iron overloaded thalassemic mice, brain T2* values decreased, ranging from 8 to 12ms, which correlated with the iron overload status of the animals. In addition, brain T2* values increased in the group with the treatment of deferiprone, ranging from 18 to 24ms. Our results may be useful to understand brain pathology in iron overload. Moreover, data could lead to an earlier diagnosis, assist in following disease progression, and demonstrate the benefits of iron chelation therapy.


Asunto(s)
Encéfalo/diagnóstico por imagen , Quelantes del Hierro/farmacología , Sobrecarga de Hierro/diagnóstico por imagen , Imagen por Resonancia Magnética , Talasemia beta/diagnóstico por imagen , Animales , Encéfalo/patología , Quelantes/farmacología , Gráficos por Computador , Deferiprona , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Hierro , Sobrecarga de Hierro/patología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Interfaz Usuario-Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA