Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807459

RESUMEN

BACKGROUND: Oral iron supplementation causes gastrointestinal side effects. Short-term alterations in dietary iron exacerbate inflammation and alter the gut microbiota, in murine models of colitis. Patients typically take supplements for months. We investigated the impact of long-term changes in dietary iron on colitis and the microbiome in mice. METHODS: We fed mice chow containing differing levels of iron, reflecting deficient (100 ppm), normal (200 ppm), and supplemented (400 ppm) intake for up to 9 weeks, both in absence and presence of dextran sodium sulphate (DSS)-induced chronic colitis. We also induced acute colitis in mice taking these diets for 8 weeks. Impact was assessed (i) clinically and histologically, and (ii) by sequencing the V4 region of 16S rRNA. RESULTS: In mice with long-term changes, the iron-deficient diet was associated with greater weight loss and histological inflammation in the acute colitis model. Chronic colitis was not influenced by altering dietary iron however there was a change in the microbiome in DSS-treated mice consuming 100 ppm and 400 ppm iron diets, and control mice consuming the 400 ppm iron diet. Proteobacteria levels increased significantly, and Bacteroidetes levels decreased, in the 400 ppm iron DSS group at day-63 compared to baseline. CONCLUSIONS: Long-term dietary iron alterations affect gut microbiota signatures but do not exacerbate chronic colitis, however acute colitis is exacerbated by such dietary changes. More work is needed to understand the impact of iron supplementation on IBD. The change in the microbiome, in patients with colitis, may arise from the increased luminal iron and not simply from colitis.


Asunto(s)
Colitis/metabolismo , Sobrecarga de Hierro/fisiopatología , Hierro/metabolismo , Anemia Ferropénica , Animales , Bacterias/genética , Colitis/fisiopatología , Colon/patología , Sulfato de Dextran/farmacología , Dieta , Suplementos Dietéticos/efectos adversos , Modelos Animales de Enfermedad , Disbiosis/etiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Inflamación , Enfermedades Inflamatorias del Intestino/patología , Hierro de la Dieta/efectos adversos , Ratones , Ratones Endogámicos C57BL , Microbiota , ARN Ribosómico 16S/genética
3.
Int J Mol Sci ; 21(22)2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33233561

RESUMEN

Thalassemia syndromes are characterized by the inability to produce normal hemoglobin. Ineffective erythropoiesis and red cell transfusions are sources of excess iron that the human organism is unable to remove. Iron that is not saturated by transferrin is a toxic agent that, in transfusion-dependent patients, leads to death from iron-induced cardiomyopathy in the second decade of life. The availability of effective iron chelators, advances in the understanding of the mechanism of iron toxicity and overloading, and the availability of noninvasive methods to monitor iron loading and unloading in the liver, heart, and pancreas have all significantly increased the survival of patients with thalassemia. Prolonged exposure to iron toxicity is involved in the development of endocrinopathy, osteoporosis, cirrhosis, renal failure, and malignant transformation. Now that survival has been dramatically improved, the challenge of iron chelation therapy is to prevent complications. The time has come to consider that the primary goal of chelation therapy is to avoid 24-h exposure to toxic iron and maintain body iron levels within the normal range, avoiding possible chelation-related damage. It is very important to minimize irreversible organ damage to prevent malignant transformation before complications set in and make patients ineligible for current and future curative therapies. In this clinical case-based review, we highlight particular aspects of the management of iron overload in patients with beta-thalassemia syndromes, focusing on our own experience in treating such patients. We review the pathophysiology of iron overload and the different ways to assess, quantify, and monitor it. We also discuss chelation strategies that can be used with currently available chelators, balancing the need to keep non-transferrin-bound iron levels to a minimum (zero) 24 h a day, 7 days a week and the risk of over-chelation.


Asunto(s)
Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/metabolismo , Reacción a la Transfusión/complicaciones , Talasemia beta/terapia , Adulto , Transfusión Sanguínea , Cardiomiopatías/sangre , Cardiomiopatías/etiología , Cardiomiopatías/fisiopatología , Cardiomiopatías/prevención & control , Terapia por Quelación/efectos adversos , Terapia por Quelación/métodos , Deferoxamina/efectos adversos , Monitoreo de Drogas/instrumentación , Monitoreo de Drogas/métodos , Femenino , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Hierro/toxicidad , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/fisiopatología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología , Transferrina/metabolismo , Reacción a la Transfusión/sangre , Reacción a la Transfusión/fisiopatología , Talasemia beta/metabolismo , Talasemia beta/patología
4.
BMJ Case Rep ; 12(2)2019 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-30782626

RESUMEN

Intrauterine transfusion is one of the mainstays of treatment in isoimmunised pregnancies guided by the changes in middle cerebral artery Doppler of the fetus. The common postnatal complications associated with Rh isoimmunisation are high unconjugated bilirubin requiring blood exchange transfusions, cholestasis due to bile inspissation, thrombocytopenia and anaemia. Hyperferritinaemia is an uncommon adverse effect observed in Rh isoimmunised pregnancies. In this case report, we describe the clinical course of a Rh isoimmunised neonate with hyperferritinaemia and transfusion acquired cytomegalovirus disease which resolved. Iron chelation therapy was not necessary.


Asunto(s)
Transfusión de Sangre Intrauterina/efectos adversos , Insuficiencia de Crecimiento/terapia , Sobrecarga de Hierro/diagnóstico , Fototerapia/métodos , Complicaciones Hematológicas del Embarazo/terapia , Isoinmunización Rh/terapia , Adulto , Antivirales/uso terapéutico , Bilirrubina/sangre , Velocidad del Flujo Sanguíneo , Transfusión de Sangre Intrauterina/métodos , Insuficiencia de Crecimiento/fisiopatología , Femenino , Ferritinas/sangre , Humanos , Recién Nacido , Sobrecarga de Hierro/fisiopatología , Sobrecarga de Hierro/terapia , Arteria Cerebral Media , Embarazo , Complicaciones Hematológicas del Embarazo/fisiopatología , Isoinmunización Rh/complicaciones , Isoinmunización Rh/fisiopatología , Resultado del Tratamiento , Valganciclovir/uso terapéutico
5.
Genomics ; 111(6): 1504-1513, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30391296

RESUMEN

To investigate the biological processes affected by long-term iron supplementation, newly hatched silkworms were exposed to high iron mulberry diet (10 and 100 ppm) and its effect on silkworm transcriptom was determined. The results showed that the silkworm was responsive to iron by increasing iron concentration and ferritin levels in the hemolymph and by regulating the expression of many other genes. A total of 523 and 326 differentially expressed genes were identified in 10 and 100 ppm Fe group compared to the control, respectively. Of these genes, 249 were shared between in both the 10 ppm and 100 ppm Fe group, including 152 up-regulated and 97 down-regulated genes. These shared genes included 19 known Fe regulated, 24 immune-related, 12 serine proteases and serine proteases homologs, 41 cuticular and cuticle genes. Ten genes (carboxypeptidases A, serine protease homologs 85, fibrohexamerin/P25, transferrin, sex-specific storage-protein 2, fungal protease inhibitor F, insect intestinal mucin, peptidoglycan recognition protein B, cuticle protein CPH45, unknown gene) were involved in the regulation of iron overload responses.


Asunto(s)
Bombyx/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Insectos/genética , Sobrecarga de Hierro/genética , Hierro de la Dieta/administración & dosificación , Hierro/administración & dosificación , Transcriptoma/efectos de los fármacos , Animales , Bombyx/efectos de los fármacos , Femenino , Proteínas de Insectos/metabolismo , Sobrecarga de Hierro/fisiopatología , Masculino
6.
Am J Physiol Cell Physiol ; 315(3): C319-C329, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29768045

RESUMEN

Resident adipose tissue macrophages (ATMs) play multiple roles to maintain tissue homeostasis, such as removing excess free fatty acids and regulation of the extracellular matrix. The phagocytic nature and oxidative resiliency of macrophages not only allows them to function as innate immune cells but also to respond to specific tissue needs, such as iron homeostasis. MFehi ATMs are a subtype of resident ATMs that we recently identified to have twice the intracellular iron content as other ATMs and elevated expression of iron-handling genes. Although studies have demonstrated that iron homeostasis is important for adipocyte health, little is known about how MFehi ATMs may respond to and influence adipose tissue iron availability. Two methodologies were used to address this question: dietary iron supplementation and intraperitoneal iron injection. Upon exposure to high dietary iron, MFehi ATMs accumulated excess iron, whereas the iron content of MFelo ATMs and adipocytes remained unchanged. In this model of chronic iron excess, MFehi ATMs exhibited increased expression of genes involved in iron storage. In the injection model, MFehi ATMs incorporated high levels of iron, and adipocytes were spared iron overload. This acute model of iron overload was associated with increased numbers of MFehi ATMs; 17% could be attributed to monocyte recruitment and 83% to MFelo ATM incorporation into the MFehi pool. The MFehi ATM population maintained its low inflammatory profile and iron-cycling expression profile. These studies expand the field's understanding of ATMs and confirm that they can respond as a tissue iron sink in models of iron overload.


Asunto(s)
Tejido Adiposo/metabolismo , Tejido Adiposo/fisiología , Hierro de la Dieta/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiología , Adipocitos/metabolismo , Adipocitos/fisiología , Animales , Línea Celular , Suplementos Dietéticos , Inflamación/metabolismo , Inflamación/fisiopatología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Monocitos/fisiología
7.
PLoS One ; 13(2): e0192890, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29466447

RESUMEN

BACKGROUND: Iron overload-related heart failure is the principal cause of death in transfusion dependent patients, including those with Thalassemia Major. Linking cardiac siderosis measured by T2* to therapy improves outcomes. T1 mapping can also measure iron; preliminary data suggests it may have higher sensitivity for iron, particularly for early overload (the conventional cut-point for no iron by T2* is 20ms, but this is believed insensitive). We compared T1 mapping to T2* in cardiac iron overload. METHODS: In a prospectively large single centre study of 138 Thalassemia Major patients and 32 healthy controls, we compared T1 mapping to dark blood and bright blood T2* acquired at 1.5T. Linear regression analysis was used to assess the association of T2* and T1. A "moving window" approach was taken to understand the strength of the association at different levels of iron overload. RESULTS: The relationship between T2* (here dark blood) and T1 is described by a log-log linear regression, which can be split in three different slopes: 1) T2* low, <20ms, r2 = 0.92; 2) T2* = 20-30ms, r2 = 0.48; 3) T2*>30ms, weak relationship. All subjects with T2*<20ms had low T1; among those with T2*>20ms, 38% had low T1 with most of the subjects in the T2* range 20-30ms having a low T1. CONCLUSIONS: In established cardiac iron overload, T1 and T2* are concordant. However, in the 20-30ms T2* range, T1 mapping appears to detect iron. These data support previous suggestions that T1 detects missed iron in 1 out of 3 subjects with normal T2*, and that T1 mapping is complementary to T2*. The clinical significance of a low T1 with normal T2* should be further investigated.


Asunto(s)
Sangre/diagnóstico por imagen , Técnicas de Imagen Cardíaca/métodos , Sobrecarga de Hierro/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Talasemia beta/diagnóstico por imagen , Adulto , Femenino , Humanos , Sobrecarga de Hierro/fisiopatología , Modelos Lineales , Masculino , Estudios Prospectivos , Talasemia beta/fisiopatología
8.
Nutrition ; 45: 49-58, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29129237

RESUMEN

OBJECTIVES: Iron is involved in processes involving oxygen transfer and utilization. Excess iron is linked to cardiovascular diseases and some types of cancer. Iron overload is associated with oxidative stress development, and may have important interactions with lipid metabolism in the liver favoring the development and progression of non-alcoholic fatty liver disease. The aim of the study described here was to assess the effect of high intake of iron on oxidative stress-related parameters, lipid metabolism, and levels of long-chain polyunsaturated fatty acids (LCPUFAs) in liver and other tissues of the rat. METHODS: Male Wistar rats (21 d old) were fed an iron-rich diet (200 mg iron/kg diet, IRD) versus a control diet (50 mg iron/kg diet; CD) for 21 d. Samples of erythrocytes, liver, adipose tissue, brain, heart, and testicles were evaluated for fatty acid composition and hepatic biochemical and oxidative stress parameters, Δ-6 and Δ-5 desaturase activities, SREBP-1c and PPAR-α mRNA expression and DNA-binding capacity, and lipolytic, lipogenic, and antioxidant enzymatic activities. RESULTS: The IRD caused liver steatosis and increased activity of plasma transaminases, with higher oxidative stress status in plasma and liver. Liver Δ-6 and Δ-5 desaturase exhibited decreased activity, but enhanced expression in response to the IRD compared with the CD, with lower levels of ω-3 and ω-6 LCPUFAs and higher expression and DNA binding of SREBP-1c, whereas expression and DNA-binding activity of PPAR-α were diminished. CONCLUSIONS: IRD induced oxidative stress and a reduction in the desaturation capacity of the liver, with LCPUFA depletion in the different tissues studied, thus promoting a pro-steatotic condition in the liver.


Asunto(s)
Ácidos Grasos Omega-3/sangre , Ácidos Grasos Omega-6/sangre , Sobrecarga de Hierro/fisiopatología , Hierro/efectos adversos , Lipogénesis , Estrés Oxidativo , Animales , Catalasa/genética , Catalasa/metabolismo , delta-5 Desaturasa de Ácido Graso , Ácido Graso Desaturasas/genética , Ácido Graso Desaturasas/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/genética , Glutatión Reductasa/metabolismo , Hierro/administración & dosificación , Linoleoil-CoA Desaturasa/genética , Linoleoil-CoA Desaturasa/metabolismo , Hígado/metabolismo , Masculino , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/etiología , PPAR alfa/genética , PPAR alfa/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
9.
Hum Fertil (Camb) ; 21(4): 294-298, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28643569

RESUMEN

Iron is a 'one-way' element and the primary point of regulation of body iron stores is at the level of intestinal iron absorption. Repeated blood transfusions for congenital anaemias bypass this regulatory checkpoint and inevitably lead to iron overload in the long-term. Iron overload causes multi-organ dysfunction of the heart, liver, pancreas and joints. It also causes reproductive toxicity primarily through its damaging effect on the anterior pituitary leading to hypogonadotrophic hypogonadism. Another less understood mechanism of reproductive toxicity is direct gonadal damage of excess free iron. In this article, we present the case of a 24-year-old woman with Diamond-Blackfan anaemia who presented to our unit seeking fertility assistance. The evaluation revealed a combination of hypogonadotrophic hypogonadism and reduced ovarian reserve along with evidence of severe iron overload. A literature search along with input from clinical experts has allowed us to counsel the patient to help her make an informed choice. A multi-disciplinary approach which would include initial optimization of pre-conceptional health with aggressive iron chelation therapy and subsequent ovulation induction with gonadotrophins has been planned, failing which, egg donation may be the only viable alternative.


Asunto(s)
Anemia de Diamond-Blackfan/complicaciones , Hipogonadismo/complicaciones , Sobrecarga de Hierro/complicaciones , Reserva Ovárica/fisiología , Ovario/fisiopatología , Anemia de Diamond-Blackfan/fisiopatología , Femenino , Humanos , Hipogonadismo/fisiopatología , Sobrecarga de Hierro/fisiopatología , Adulto Joven
10.
Transfus Clin Biol ; 24(3): 223-226, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28673501

RESUMEN

Hemoglobinopathies, thalassemia and sickle cell disease are among the most frequent monogenic diseases in the world. Transfusion has improved dramatically their prognosis, but provokes iron overload, which induces multiple organ damages. Iron overload is related to accumulation of iron released from hemolysis and transfused red cell, but also, in thalassemic patients, secondary to ineffective erythropoiesis, which increases intestinal iron absorption via decreased hepcidin production. Transfusion-related cardiac iron overload remains a main cause of death in thalassemia in well-resourced countries, and is responsible for severe hepatic damages in sickle cell disease. Regular monitoring by Magnetic Resonance Imaging (MRI) using myocardial T2* (ms) and Liver Iron Content (LIC) (mg of iron/g dry weight) are now standards of care in chronically transfused patients. Serum ferritin level measurements and record of the total number of transfused erythrocyte concentrates are also helpful tools. Three iron chelators are currently available, deferoxamine, which must be injected subcutaneously or intravenously, and two oral chelators, deferiprone and deferasirox. We will review the main characteristics of these drugs and their indications.


Asunto(s)
Hemoglobinopatías/complicaciones , Sobrecarga de Hierro/tratamiento farmacológico , Transfusión Sanguínea , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/etiología , Cardiomiopatías/patología , Terapia por Quelación , Ferritinas/análisis , Hemoglobinopatías/terapia , Hemólisis , Hepcidinas/biosíntesis , Humanos , Absorción Intestinal , Hierro/análisis , Hierro/farmacocinética , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/efectos adversos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/fisiopatología , Hierro de la Dieta/farmacocinética , Hepatopatías/diagnóstico por imagen , Hepatopatías/etiología , Hepatopatías/patología , Imagen por Resonancia Magnética
11.
Crit Rev Oncol Hematol ; 113: 156-170, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28427505

RESUMEN

Iron is an essential element for key cellular metabolic processes. However, transfusional iron overload (IOL) may result in significant cellular toxicity. IOL occurs in transfusion dependent hematologic malignancies (HM), may lead to pathological clinical outcomes, and IOL reduction may improve outcomes. In hematopoietic stem cell transplantation (SCT) for HM, IOL may have clinical importance; endpoints examined regarding an impact of IOL and IOL reduction include transplant-related mortality, organ function, infection, relapse risk, and survival. Here we review the clinical consequences of IOL and effects of IOL reduction before, during and following SCT for HM. IOL pathophysiology is discussed as well as available tests for IOL quantification including transfusion history, serum ferritin level, transferrin saturation, hepcidin, labile plasma iron and other parameters of iron-catalyzed oxygen free radicals, and organ IOL by imaging. Data-based recommendations for IOL measurement, monitoring and reduction before, during and following SCT for HM are made.


Asunto(s)
Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/fisiopatología , Transfusión Sanguínea , Neoplasias Hematológicas/complicaciones , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/tratamiento farmacológico
12.
Transfusion ; 57(3): 700-704, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28019032

RESUMEN

BACKGROUND: Use of chronic blood transfusions as a treatment modality in patients with blood disorders places them at risk for iron overload. Since patients with ß-thalassemia major (TM) are transfusion-dependent, most studies on iron overload and chelation have been conducted in this population. While available data suggest that compared to TM, patients with sickle cell disease (SCD) have a lower risk of extrahepatic iron overload, significant iron overload can develop. Further, previous studies have demonstrated a direct relationship between iron overload and morbidity and mortality rates in SCD. However, reports describing the outcome for patients with SCD and cardiac iron overload are rare. STUDY DESIGN AND METHODS: We performed a retrospective analysis and identified two SCD patients with cardiac iron overload. We provide detailed descriptions of both cases and their outcomes. RESULTS: Serum ferritin levels ranged between 17,000 and 19,000 µg/L. Both had liver iron concentrations in excess of 35 mg of iron per gram of dried tissue as well as evidence of cardiac iron deposition on magnetic resonance imaging. One patient died of an arrhythmia and had evidence of severe multiorgan iron overload via autopsy. On the other hand, after appropriate therapy, a second patient had improvement in cardiac function. CONCLUSION: Improper treatment of iron overload in SCD can lead to a fatal outcome. Alternatively, iron overload may potentially be prevented or reversed with judicious use of blood transfusions and early use of chelation therapy, respectively.


Asunto(s)
Anemia de Células Falciformes , Arritmias Cardíacas , Ferritinas/sangre , Sobrecarga de Hierro , Hierro/sangre , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/fisiopatología , Anemia de Células Falciformes/terapia , Arritmias Cardíacas/sangre , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/terapia , Femenino , Humanos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/fisiopatología , Sobrecarga de Hierro/terapia , Masculino
13.
Sci Rep ; 6: 29110, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-27357559

RESUMEN

Superparamagnetic iron oxide nanoparticles (SPIONs) as a contrast agent have been widely used in magnetic resonance imaging for tumor diagnosis and theranostics. However, there has been safety concern of SPIONs with cirrhosis related to excess iron-induced oxidative stress. In this study, the impact of iron overload by SPIONs was assessed on a mouse cirrhosis model. A single dose of SPION injection at 0.5 or 5 mg Fe/kg in the cirrhosis group induced a septic shock response at 24 h with elevated serum levels of liver and kidney function markers and extended impacts over 14 days including high levels of serum cholesterols and persistent low serum iron level. In contrast, full restoration of liver functions was found in the normal group with the same dosages over time. Analysis with PCR array of the toxicity pathways revealed the high dose of SPIONs induced significant expression changes of a distinct subset of genes in the cirrhosis liver. All these results suggested that excess iron of the high dose of SPIONs might be a risk factor for cirrhosis because of the marked impacts of elevated lipid metabolism, disruption of iron homeostasis and possibly, aggravated loss of liver functions.


Asunto(s)
Cirrosis Hepática/fisiopatología , Hígado/efectos de los fármacos , Nanopartículas de Magnetita/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Animales , Medios de Contraste/efectos adversos , Medios de Contraste/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Sobrecarga de Hierro/inducido químicamente , Sobrecarga de Hierro/fisiopatología , Hígado/fisiopatología , Cirrosis Hepática/inducido químicamente , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/uso terapéutico , Ratones , Nanomedicina Teranóstica , Distribución Tisular/efectos de los fármacos
14.
Toxicol Lett ; 258: 159-167, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27320964

RESUMEN

Evidence indicates that hepatic fibrosis is the initial lesion of cirrhosis or hepatocellular carcinoma in diseases such as nonalcoholic steatohepatitis (NASH). To induce NASH, we fed rats a choline-deficient and iron-supplemented L-amino acid-defined (CDAA) diet. Histopathological examination revealed that fibrosis appeared from week 4 and progressed to bridging fibrosis from week 12. Using qRT-PCR assays, we detected increased expression of miR-21, Mmp-9, and Timp-1 in liver that peaked during week 4, when fibrosis was first detected. The expression pattern of miR-21 in plasma paralleled that in liver. Fibrosis tended to be resolved within 12 weeks of a recovery period after 12 weeks of feeding, and the expression of miR-21, Timp-1, and Mmp-9 decreased in liver. Comprehensive analyses of miRNA and mRNA expression in the liver using samples acquired at week 4 detected 16 miRNAs and 11 mRNAs that are mutually-interacting fibrosis-related factors. We therefore conclude that miR-21 was closely associated with fibrosis in a rat model of NASH and has potential as a plasma biomarker for hepatic fibrosis.


Asunto(s)
Modelos Animales de Enfermedad , Cirrosis Hepática/etiología , Hígado/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Biomarcadores/sangre , Colina/uso terapéutico , Deficiencia de Colina/complicaciones , Deficiencia de Colina/dietoterapia , Deficiencia de Colina/etiología , Deficiencia de Colina/fisiopatología , Dieta/efectos adversos , Suplementos Dietéticos/envenenamiento , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/fisiopatología , Hierro de la Dieta/envenenamiento , Hígado/inmunología , Hígado/patología , Hígado/fisiopatología , Cirrosis Hepática/prevención & control , Metaloproteinasa 9 de la Matriz/genética , MicroARNs/sangre , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , ARN Mensajero/metabolismo , Ratas Wistar , Organismos Libres de Patógenos Específicos , Inhibidor Tisular de Metaloproteinasa-1/genética
15.
Annu Rev Nutr ; 36: 241-73, 2016 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-27146016

RESUMEN

Dietary advice is the cornerstone in first-line treatment of metabolic diseases. Nutritional interventions directed at these clinical conditions mainly aim to (a) improve insulin resistance by reducing energy-dense macronutrient intake to obtain weight loss and (b) reduce fluctuations in insulin secretion through avoidance of rapidly absorbable carbohydrates. However, even in the majority of motivated patients selected for clinical trials, massive efforts using this approach have failed to achieve lasting efficacy. Less attention has been given to the role of micronutrients in metabolic diseases. Here, we review the evidence that highlights (a) the importance of iron in pancreatic beta-cell function and dysfunction in diabetes and (b) the integrative pathophysiological effects of tissue iron levels in the interactions among the beta cell, gut microbiome, hypothalamus, innate and adaptive immune systems, and insulin-sensitive tissues. We propose that clinical trials are warranted to clarify the impact of dietary or pharmacological iron reduction on the development of metabolic disorders.


Asunto(s)
Diabetes Mellitus/prevención & control , Medicina Basada en la Evidencia , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Hierro de la Dieta/uso terapéutico , Estrés Oxidativo , Inmunidad Adaptativa , Animales , Apoptosis , Diabetes Mellitus/etiología , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Suplementos Dietéticos/efectos adversos , Microbioma Gastrointestinal/inmunología , Homeostasis , Humanos , Hipotálamo/inmunología , Hipotálamo/metabolismo , Inmunidad Innata , Secreción de Insulina , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Sobrecarga de Hierro/inmunología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Sobrecarga de Hierro/fisiopatología , Hierro de la Dieta/efectos adversos , Hierro de la Dieta/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo
16.
Am J Clin Nutr ; 103(2): 375-81, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26762369

RESUMEN

BACKGROUND: An iron overload may induce pancreatic islet damage and increase risk of diabetes. Women with a history of gestational diabetes mellitus (GDM) are at high risk of developing type 2 diabetes mellitus (T2DM) after pregnancy. OBJECTIVE: We aimed to examine the association of habitual iron intake with long-term risk of T2DM in this high-risk population. DESIGN: We included 3976 women with a history of GDM from the Nurses' Health Study II cohort as part of the ongoing Diabetes & Women's Health Study. The women were followed up through 2009. Iron intake was assessed with the use of a validated food-frequency questionnaire in 1991 and every 4 y thereafter. We used Cox proportional hazards models to estimate HRs and 95% CIs. RESULTS: We documented 641 incident T2DM cases during 57,683 person-years of observation. Adjusted HRs for T2DM for the highest quartile compared with the lowest quartile were 1.64 (95% CI: 1.20, 2.25; P-trend = 0.02) for total iron intake and 1.80 (95% CI: 1.18, 2.74; P-trend = 0.005) for dietary heme iron intake. In addition, women who consumed ≥30.0 mg supplemental Fe/d, compared with nonusers, had an adjusted HR of 1.83 (95% CI: 1.25, 2.70; P-trend = 0.002). CONCLUSION: In women with a history of GDM, greater intakes of total iron, dietary heme iron, and supplemental iron were associated with higher risk of T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Diabetes Gestacional/fisiopatología , Suplementos Dietéticos/efectos adversos , Sobrecarga de Hierro/fisiopatología , Hierro de la Dieta/efectos adversos , Carne/efectos adversos , Adulto , Estudios de Cohortes , Diabetes Mellitus Tipo 2/epidemiología , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Hemo/efectos adversos , Humanos , Incidencia , Sobrecarga de Hierro/etiología , Hierro de la Dieta/envenenamiento , Enfermeras y Enfermeros , Encuestas Nutricionales , Embarazo , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Riesgo , Estados Unidos/epidemiología , Adulto Joven
17.
J Pediatr Hematol Oncol ; 37(8): 605-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26422288

RESUMEN

BACKGROUND: Thalassemia major (TM) is a chronic disease requiring regular transfusions that may result in generalized iron loading, such as in the heart, the liver, endocrine organs, and the lungs. We aimed to determine pulmonary function abnormalities in children with TM in our center. PATIENTS AND METHODS: In this study, pulmonary function tests (PFTs) of 49 patients with TM who received regular blood transfusion and had no history of chronic respiratory disease were evaluated. The relationship between PFTs and the age, the body surface area, pretransfusional hemoglobin, and serum ferritin was evaluated. RESULTS: Among the ß-TM patients included in this study, 61% were male and 39% were female, with a mean age of 10.8±3 years (range, 5 to 17 y). The patients' mean level of ferritin was 3873±2011 ng/dL (range, 676 to 9476 ng/dL). A reduced forced vital capacity (FVC) was found in 33 patients (67%). A reduced forced expiratory volume in 1 second (FEV1) was found in 15 patients (30%). A forced expiratory volume in 1 second to forced vital capacity (FEV1/FVC) ratio of >80% was found in all patients. The peak expiratory flow (PEF) was decreased in 23 patients (46.9%). The forced mid-expiratory flow between 25% and 75% of the exhaled vital capacity (MEF25%-75%) was decreased in 5 patients (10%). FVC and FEV1 values in patients with a high ferritin level (>2500 ng/dL) were decreased compared with patients with a low ferritin level (<2500 ng/dL) (P=0.04, 0.03). FVC, FEV1, and PEF parameters were negatively correlated with the age and the body surface area. Age was a predictor of FVC (ß=-0.450, P<0.001), FEV1 (ß=-0.419, P<0.001), and PEF (ß=-0.505, P<0.001), and hemoglobin was a predictor of FEV1/FVC (ß=0.366, P=0.01) and MEF25%-75% (ß=0.323, P=0.003). CONCLUSIONS: Our results concluded that the respiratory system should be evaluated by PFTs even in asymptomatic patients with high serum ferritin levels during the adolescent period annually to prevent the squeal of pulmonary disease in TM. Patients who have abnormal PFTs should be reevaluated for compliance with chelation therapy and the transfusion program.


Asunto(s)
Pulmón/fisiopatología , Talasemia beta/fisiopatología , Adolescente , Factores de Edad , Enfermedades Asintomáticas , Superficie Corporal , Terapia por Quelación , Niño , Preescolar , Terapia Combinada , Estudios Transversales , Femenino , Ferritinas/sangre , Hemoglobinas/análisis , Humanos , Hierro , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/fisiopatología , Mediciones del Volumen Pulmonar , Masculino , Espirometría , Esplenectomía , Reacción a la Transfusión , Talasemia beta/sangre , Talasemia beta/terapia
18.
J Nutr Biochem ; 26(11): 1174-82, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26239832

RESUMEN

Estrogen deficiency has been considered to be a major cause of osteoporosis, but recent epidemiological evidence and mechanistic studies have indicated that aging and the associated increase in reactive oxygen species (ROS) are the proximal pathogenic factors. Through ROS-mediated reactions, iron can induce disequilibrium of oxidation and antioxidation and can cause bone loss in mice. Therefore, we investigated the effects of resveratrol (RES) on bone mineral density, bone microstructure and the osteoblast functions under iron-overload conditions. Excess iron disrupted the antioxidant/prooxidant equilibrium of the mice and induced the defect and the lesion of the bone trabecula as well as disequilibrium between bone formation and bone resorption in iron-overload mice. Oral administration of RES significantly prevented bone loss in the osteoporotic mice. RES reversed the reduction of Runx2, OCN and type I collagen from excess iron; up-regulated the level of FOXO1; and maintained the antioxidant/prooxidant equilibrium in the mice. RES also reduced the ratio of OPG/RANKL in MC3T3-E1 cells and in mice and significantly inhibited subsequent osteoclastogenesis. These results provide new insights into the antiosteoporosis mechanisms of RES through antioxidative effects, suggesting that RES can be considered a potential natural resource for developing medicines or dietary supplements to prevent and treat osteoporosis.


Asunto(s)
Antioxidantes/metabolismo , Densidad Ósea/efectos de los fármacos , Sobrecarga de Hierro/complicaciones , Osteoporosis/dietoterapia , Estilbenos/farmacología , Animales , Línea Celular , Suplementos Dietéticos , Fémur/efectos de los fármacos , Fémur/metabolismo , Fémur/ultraestructura , Interleucina-6/sangre , Sobrecarga de Hierro/fisiopatología , Masculino , Ratones Endogámicos C57BL , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Resveratrol , Estilbenos/sangre , Factor de Necrosis Tumoral alfa/sangre
19.
Blood Cells Mol Dis ; 55(3): 194-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26227843

RESUMEN

The disorders of iron overload due to primary or secondary cause are one of the important human diseases leading to high mortality if untreated. To understand this, an animal model has been extensively studied. The source of iron administered to the mode of iron administration that can mimic the iron overload in humans has been studied. A safe and orally active iron chelator is still needed as many of the existing compounds have different types of complications and toxicity associated. Hence having a simple animal model which can be availed quickly and can be used to study various compounds for its iron chelating activity would likely to have immense utility for pharmacological studies. In this review we have shown how, using a simple procedure, a large number of small iron overloaded animals can be produced easily for various studies.


Asunto(s)
Modelos Animales de Enfermedad , Sobrecarga de Hierro , Animales , Terapia por Quelación , Humanos , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/fisiopatología , Sobrecarga de Hierro/terapia
20.
Free Radic Biol Med ; 88(Pt A): 3-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26216855

RESUMEN

With repeated blood transfusions, patients with thalassemia major rapidly become loaded with iron, often surpassing hepatic metal accumulation capacity within ferritin shells and infiltrating heart and endocrine organs. That pathological scenario contrasts with the physiological one, which is characterized by an efficient maintenance of all plasma iron bound to circulating transferrin, due to a tight control of iron ingress into plasma by the hormone hepcidin. Within cells, most of the acquired iron becomes protein-associated, as once released from endocytosed transferrin, it is used within mitochondria for the synthesis of protein prosthetic groups or it is incorporated into enzyme active centers or alternatively sequestered within ferritin shells. A few cell types also express the iron extrusion transporter ferroportin, which is under the negative control of circulating hepcidin. However, that system only backs up the major cell regulated iron uptake/storage machinery that is poised to maintain a basal level of labile cellular iron for metabolic purposes without incurring potentially toxic scenarios. In thalassemia and other transfusion iron-loading conditions, once transferrin saturation exceeds about 70%, labile forms of iron enter the circulation and can gain access to various types of cells via resident transporters or channels. Within cells, they can attain levels that exceed their ability to chemically cope with labile iron, which has a propensity for generating reactive oxygen species (ROS), thereby inducing oxidative damage. This scenario occurs in the heart of hypertransfused thalassemia major patients who do not receive adequate iron-chelation therapy. Iron that accumulates in cardiomyocytes forms agglomerates that are detected by T2* MRI. The labile forms of iron infiltrate the mitochondria and damage cells by inducing noxious ROS formation, resulting in heart failure. The very rapid relief of cardiac dysfunction seen after intensive iron-chelation therapy in some patients with thalassemia major is thought to be due to the relief of the cardiac mitochondrial dysfunction caused by oxidative stress or to the removal of labile iron interference with calcium fluxes through cardiac calcium channels. In fact, improvement occurs well before there is any significant improvement in the total level of cardiac iron loading. The oral iron chelator deferiprone, because of its small size and neutral charge, demonstrably enters cells and chelates labile iron, thereby rapidly reducing ROS formation, allowing better mitochondrial activity and improved cardiac function. Deferiprone may also rapidly improve arrhythmias in patients who do not have excessive cardiac iron. It maintains the flux of iron in the direction hemosiderin to ferritin to free iron, and it allows clearance of cardiac iron in the presence of other iron chelators or when used alone. To date, the most commonly used chelator combination therapy is deferoxamine plus deferiprone, whereas other combinations are in the process of assessment. In summary, it is imperative that patients with thalassemia major have iron chelators continuously present in their circulation to prevent exposure of the heart to labile iron, reduce cardiac toxicity, and improve cardiac function.


Asunto(s)
Cardiomiopatías/etiología , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/etiología , Estrés Oxidativo/efectos de los fármacos , Talasemia/complicaciones , Cardiomiopatías/fisiopatología , Cardiomiopatías/prevención & control , Humanos , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/fisiopatología , Talasemia/fisiopatología , Transferrina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA