Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Ann Hematol ; 103(6): 1887-1896, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38581547

RESUMO

We evaluated the impact of the genotype on clinical and hematochemical features, hepatic and cardiac iron levels, and endocrine, hepatic, and cardiovascular complications in non-transfusion-dependent (NTD) ß-thalassemia intermedia (TI) patients. Sixty patients (39.09 ± 11.11 years, 29 females) consecutively enrolled in the Myocardial Iron Overload in Thalassemia project underwent Magnetic Resonance Imaging to quantify iron overload, biventricular function parameters, and atrial areas and to detect replacement myocardial fibrosis. Three groups of patients were identified: homozygous ß+ (N = 18), heterozygous ß0ß+ (N = 22), and homozygous ß0 (N = 20). The groups were homogeneous for sex, age, splenectomy, hematochemical parameters, chelation therapy, and iron levels. The homozygous ß° genotype was associated with significantly higher biventricular end-diastolic and end-systolic volume indexes and bi-atrial area indexes. No difference was detected in biventricular ejection fractions or myocardial fibrosis. Extramedullary hematopoiesis and leg ulcers were significantly more frequent in the homozygous ß° group compared to the homozygous ß+ group. No association was detected between genotype and liver cirrhosis, hypogonadism, hypothyroidism, osteoporosis, heart failure, arrhythmias, and pulmonary hypertension. Heart remodelling related to a high cardiac output state cardiomyopathy, extramedullary hematopoiesis, and leg ulcers were more pronounced in patients with the homozygous ß° genotype compared to the other genotypes analyzed. The knowledge of the genotype can assist in the clinical management of NTD ß-TI patients.


Assuntos
Genótipo , Sobrecarga de Ferro , Ferro , Talassemia beta , Humanos , Talassemia beta/genética , Talassemia beta/complicações , Feminino , Masculino , Adulto , Pessoa de Meia-Idade , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/etiologia , Ferro/metabolismo , Úlcera da Perna/etiologia , Úlcera da Perna/genética , Hematopoese Extramedular/genética , Imageamento por Ressonância Magnética , Miocárdio/patologia , Miocárdio/metabolismo , Cirrose Hepática/genética , Cirrose Hepática/complicações , Homozigoto
2.
Clin Lab ; 70(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38213217

RESUMO

BACKGROUND: ß-thalassemia is an inherited disorder caused by defects in the synthesis of the beta-globin chain. One of the significant clinical complications in ß-thalassemia intermedia is iron overload toxicity, which may be attributed to reduced levels of hepcidin. This reduction in hepcidin leads to increased absorption of iron in the intestines, ultimately resulting in iron overload. The objective of this study was to assess the impact of curcumin on the expression of growth differentiating factor-15 (GDF-15) and hepcidin genes in patients with beta-thalassemia intermedia. METHODS: This study was designed as a randomized controlled double-blind clinical trial. Prior to and after the intervention period with curcumin, a blood sample of 5 mL was collected from both the placebo and curcumin-treated groups for the assessment of hepcidin and growth differentiating factor-15 gene expression. RESULTS: This study revealed a significant reduction in the expression of growth differentiating factor-15 in the curcumin group compared to the placebo group during the 3-month treatment period. Furthermore, curcumin supplementation led to a remarkable 10.1-fold increase in the levels of hepcidin in the curcumin group compared to the placebo group. CONCLUSIONS: The results of this study show that curcumin administration increases the mRNA levels of hepcidin in whole blood of thalassemia intermedia patients and supports the idea that curcumin could be a potential treatment to reduce suppression of hepcidin in thalassemias and other iron-loading anemias. CONCLUSIONS: The results of this study show that curcumin administration increases the mRNA levels of hepcidin in whole blood of thalassemia intermedia patients and supports the idea that curcumin could be a potential treatment to reduce suppression of hepcidin in thalassemias and other iron-loading anemias.


Assuntos
Curcumina , Sobrecarga de Ferro , Talassemia beta , Humanos , Hepcidinas/genética , Fator 15 de Diferenciação de Crescimento/genética , Talassemia beta/tratamento farmacológico , Talassemia beta/genética , Curcumina/farmacologia , Curcumina/uso terapêutico , Ferro , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/genética , RNA Mensageiro/genética , Expressão Gênica
3.
Am J Hematol ; 99(1): 12-20, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37867341

RESUMO

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.


Assuntos
Anemia , Apoferritinas , Deficiências de Ferro , Sobrecarga de Ferro , Feminino , Humanos , Anemia/genética , Apoferritinas/genética , Apoferritinas/metabolismo , Ferritinas , Ferro/metabolismo , Deficiências de Ferro/genética , Sobrecarga de Ferro/genética
4.
FASEB J ; 37(11): e23245, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37874260

RESUMO

Iron overload is one of the secondary osteoporosis etiologies. Cellular and molecular mechanisms involved in iron-related osteoporosis are not fully understood. AIM: The aim of the study was to investigate the respective roles of iron excess and hepcidin, the systemic iron regulator, in the development of iron-related osteoporosis. MATERIAL AND METHODS: We used mice models with genetic iron overload (GIO) related to hepcidin deficiency (Hfe-/- and Bmp6-/- ) and secondary iron overload (SIO) exhibiting a hepcidin increase secondary to iron excess. Iron concentration and transferrin saturation levels were evaluated in serum and hepatic, spleen, and bone iron concentrations were assessed by ICP-MS and Perl's staining. Gene expression was evaluated by quantitative RT-PCR. Bone micro-architecture was evaluated by micro-CT. The osteoblastic MC3T3 murine cells that are able to mineralize were exposed to iron and/or hepcidin. RESULTS: Despite an increase of bone iron concentration in all overloaded mice models, bone volume/total volume (BV/TV) and trabecular thickness (Tb.Th) only decreased significantly in GIO, at 12 months for Hfe-/- and from 6 months for Bmp6-/- . Alterations in bone microarchitecture in the Bmp6-/- model were positively correlated with hepcidin levels (BV/TV (ρ = +.481, p < .05) and Tb.Th (ρ = +.690, p < .05). Iron deposits were detected in the bone trabeculae of Hfe-/- and Bmp6-/- mice, while iron deposits were mainly visible in bone marrow macrophages in secondary iron overload. In cell cultures, ferric ammonium citrate exposure abolished the mineralization process for concentrations above 5 µM, with a parallel decrease in osteocalcin, collagen 1, and alkaline phosphatase mRNA levels. Hepcidin supplementation of cells had a rescue effect on the collagen 1 and alkaline phosphatase expression level decrease. CONCLUSION: Together, these data suggest that iron in excess alone is not sufficient to induce osteoporosis and that low hepcidin levels also contribute to the development of osteoporosis.


Assuntos
Hemocromatose , Sobrecarga de Ferro , Osteoporose , Animais , Camundongos , Ferro/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Hemocromatose/genética , Fosfatase Alcalina/metabolismo , Proteína da Hemocromatose/genética , Antígenos de Histocompatibilidade Classe I/genética , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Fígado/metabolismo , Osteoporose/genética , Colágeno/metabolismo , Camundongos Knockout
5.
JAMA Netw Open ; 6(10): e2338995, 2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37870835

RESUMO

Importance: HFE gene-associated hereditary hemochromatosis type 1 (HH1) is underdiagnosed, resulting in missed opportunities for preventing morbidity and mortality. Objective: To assess whether screening for p.Cys282Tyr homozygosity is associated with recognition and management of asymptomatic iron overload. Design, Setting, and Participants: This cross-sectional study obtained data from the Geisinger MyCode Community Health Initiative, a biobank of biological samples and linked electronic health record data from a rural, integrated health care system. Participants included those who received a p.Cys282Tyr homozygous result via genomic screening (MyCode identified), had previously diagnosed HH1 (clinically identified), and those negative for p.Cys282Tyr homozygosity between 2017 and 2018. Data were analyzed from April 2020 to August 2023. Exposure: Disclosure of a p.Cys282Tyr homozygous result. Main Outcomes and Measures: Postdisclosure management and HFE-associated phenotypes in MyCode-identified participants were analyzed. Rates of HFE-associated phenotypes in MyCode-identified participants were compared with those of clinically identified participants. Relevant laboratory values and rates of laboratory iron overload among participants negative for p.Cys282Tyr homozygosity were compared with those of MyCode-identified participants. Results: A total of 86 601 participants had available exome sequences at the time of analysis, of whom 52 994 (61.4%) were assigned female at birth, and the median (IQR) age was 62.0 (47.0-73.0) years. HFE p.Cys282Tyr homozygosity was disclosed to 201 participants, of whom 57 (28.4%) had a prior clinical HH1 diagnosis, leaving 144 participants who learned of their status through screening. There were 86 300 individuals negative for p.Cys282Tyr homozygosity. After result disclosure, among MyCode-identified participants, 99 (68.8%) had a recommended laboratory test and 36 (69.2%) with laboratory or liver biopsy evidence of iron overload began phlebotomy or chelation. Fifty-three (36.8%) had iron overload; rates of laboratory iron overload were higher in MyCode-identified participants than participants negative for p.Cys282Tyr homozygosity (females: 34.1% vs 2.1%, P < .001; males: 39.0% vs 2.9%, P < .001). Iron overload (females: 34.1% vs 79.3%, P < .001; males: 40.7% vs 67.9%, P = .02) and some liver-associated phenotypes were observed at lower frequencies in MyCode-identified participants compared with clinically identified individuals. Conclusions and Relevance: Results of this cross-sectional study showed the ability of genomic screening to identify undiagnosed iron overload and encourage relevant management, suggesting the potential benefit of population screening for HFE p.Cys282Tyr homozygosity. Further studies are needed to examine the implications of genomic screening for health outcomes and cost-effectiveness.


Assuntos
Hemocromatose , Sobrecarga de Ferro , Masculino , Recém-Nascido , Humanos , Feminino , Pessoa de Meia-Idade , Idoso , Hemocromatose/diagnóstico , Hemocromatose/genética , Hemocromatose/terapia , Estudos Transversais , Proteína da Hemocromatose/genética , Sobrecarga de Ferro/diagnóstico , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/complicações , Testes Genéticos
6.
Hematol Oncol Stem Cell Ther ; 16(3): 238-244, 2023 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-34883086

RESUMO

BACKGROUND: Blood transfusion (BT) is essential in treating sickle cell disease (SCD); however, it leads to iron overload (IO) and oxidative stress. We studied the relationship between oxidative stress, iron status parameters, hepcidin mRNA gene expression, and IO in SCD patients. METHODS: We classified all SCD patients (n = 90) into two groups: Group I, 45 children (s.ferritin ≥ 938 ng/mL) and Group II, 45 children (s.ferritin < 938 ng/mL). A total of 55 children, age and sex matched, participated as a control group. Malondialdehyde (MDA), nitrite, s.iron, s.total iron-binding capacity (sTIBC), transferrin saturation %, s.ferritin, s.hepcidin, and hepcidin mRNA gene expression were assessed. RESULTS: Among SCD BT-dependent patients (>3 times/year), 63% were from Group I and 37% from Group II, p < .01. The two patient groups had significantly lower s.hepcidin and hepcidin gene expression than controls ( p < .001). TIBC, s.iron, s.ferritin, transferrin saturation %, ferritin/hepcidin ratio, and MDA levels were higher among SCD patients than controls ( p < .001). Group I had higher mean level of ferritin/hepcidin ratio and MDA than Group II ( p < .01). The higher level of MDA and increased frequency of BT were the significant predicting risk factors for IO ( p < .05). A receiver-operating characteristic curve indicates that MDA is the outstanding significant biomarker for high level of s.ferritin with subsequent IO progression. CONCLUSION: MDA may serve as a biomarker of oxidative stress and IO in SCD patients. This result paid attention for urgent initiation of antioxidant and chelation therapy on detecting increased MDA level.


Assuntos
Anemia Falciforme , Sobrecarga de Ferro , Humanos , Criança , Hepcidinas/metabolismo , Sobrecarga de Ferro/genética , Ferro/metabolismo , Anemia Falciforme/complicações , Anemia Falciforme/genética , Ferritinas , Estresse Oxidativo , Biomarcadores/metabolismo , Transferrinas/metabolismo
7.
Am J Hematol ; 97(10): 1324-1336, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36071579

RESUMO

ß-thalassemia is a genetic disorder caused by mutations in the ß-globin gene, and characterized by anemia, ineffective erythropoiesis and iron overload. Patients affected by the most severe transfusion-dependent form of the disease (TDT) require lifelong blood transfusions and iron chelation therapy, a symptomatic treatment associated with several complications. Other therapeutic opportunities are available, but none is fully effective and/or applicable to all patients, calling for the identification of novel strategies. Transferrin receptor 2 (TFR2) balances red blood cells production according to iron availability, being an activator of the iron-regulatory hormone hepcidin in the liver and a modulator of erythropoietin signaling in erythroid cells. Selective Tfr2 deletion in the BM improves anemia and iron-overload in non-TDT mice, both as a monotherapy and, even more strikingly, in combination with iron-restricting approaches. However, whether Tfr2 targeting might represent a therapeutic option for TDT has never been investigated so far. Here, we prove that BM Tfr2 deletion improves anemia, erythrocytes morphology and ineffective erythropoiesis in the Hbbth1/th2 murine model of TDT. This effect is associated with a decrease in the expression of α-globin, which partially corrects the unbalance with ß-globin chains and limits the precipitation of misfolded hemoglobin, and with a decrease in the activation of unfolded protein response. Remarkably, BM Tfr2 deletion is also sufficient to avoid long-term blood transfusions required for survival of Hbbth1/th2 animals, preventing mortality due to chronic anemia and reducing transfusion-associated complications, such as progressive iron-loading. Altogether, TFR2 targeting might represent a promising therapeutic option also for TDT.


Assuntos
Sobrecarga de Ferro , Receptores da Transferrina , Talassemia beta , Animais , Transfusão de Sangue , Modelos Animais de Doenças , Ferro/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Camundongos , Receptores da Transferrina/genética , Globinas beta , Talassemia beta/genética , Talassemia beta/terapia
8.
Free Radic Biol Med ; 161: 187-197, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33080340

RESUMO

Increasing populations are found to bear mild hepatic iron overload (HIO) due to unhealthy lifestyles, metabolic diseases, etc., whether this mild but chronic HIO induces hepatic inflammation is unknown. In the present study, mice receiving a 12-months 0.3% dextran-iron diet show mild HIO with no detectable oxidative damages in the liver but have infiltrated macrophages and increased IL-6, TNFα, AST and ALT since 6-months. The HNF4α/miR-122/CCL2 pathway, identified by our previous studies to induce macrophages infiltration, is initiated by chronic mild HIO. After excluding the role of DNA methylation, a modified transcription factor microarray is applied to find that transcription factor YY1 is responsible for HIO-decreased HNF4α expression. Then the E3 ubiquitin ligase TRIP12 is identified by an immunoprecipitation coupled LC-MS/MS and proved to bind and ubiquitinate YY1, leading to its degradation. The overexpression or silence of YY1 in the liver regulates the HNF4α/miR-122/CCL2 pathway. More importantly, YY1 overexpression alleviates chronic mild HIO induced hepatic inflammatory responses. In conclusion, these results elucidate an oxidative-stress-independent, TRIP12/YY1/HNF4α/miR-122/CCL2 pathway of chronic mild HIO inducing hepatic inflammation, implying that effective measures in addition to antioxidants are needed for individuals at the risk of chronic mild HIO.


Assuntos
Sobrecarga de Ferro , Espectrometria de Massas em Tandem , Ubiquitina-Proteína Ligases , Animais , Cromatografia Líquida , Inflamação/genética , Sobrecarga de Ferro/genética , Fígado , Camundongos , Ubiquitina-Proteína Ligases/genética , Fator de Transcrição YY1/genética
9.
Signal Transduct Target Ther ; 5(1): 138, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32732975

RESUMO

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.


Assuntos
Auranofina/farmacologia , Ferroptose/efeitos dos fármacos , Hemocromatose , Sobrecarga de Ferro , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Feminino , Ferroptose/genética , Células HEK293 , Hemocromatose/tratamento farmacológico , Hemocromatose/genética , Hemocromatose/metabolismo , Hemocromatose/patologia , Humanos , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais/genética
10.
Food Funct ; 10(8): 5203-5214, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31380553

RESUMO

Alcoholic liver disease (ALD) is associated with significant morbidity and mortality globally. In this study, the soybean lecithin-gallic acid complex was synthesized, and its physicochemical properties were evaluated, which confirmed the complex formation. Compared with the free state of the drug, gallic acid exhibited significantly different physicochemical properties after it was complexed with soybean lecithin. To clarify the binding mode between two monomers, computational investigation was performed. From the computational data, we deduced the structure of the compound and predicted that it has a high affinity for human phosphatidylcholine transfer protein and exhibits strong pharmacological activities in vivo. The complex not only effectively ameliorated liver fibrosis, lipid peroxidation, and oxidative stress, but also reduced liver iron overload in a mouse ALD model induced by alcohol (p < 0.05). Additionally, it regulated iron metabolism by inhibiting TfR1 expression (p < 0.05) and promoting hepcidin expression (p < 0.05). These results suggest that the soybean lecithin-gallic acid complex ameliorates hepatic damage and iron overload induced by alcohol and exert hepatoprotective effects.


Assuntos
Ácido Gálico/administração & dosagem , Sobrecarga de Ferro/tratamento farmacológico , Lecitinas/administração & dosagem , Hepatopatias Alcoólicas/tratamento farmacológico , Animais , Biologia Computacional , Ácido Gálico/química , Hepcidinas/genética , Hepcidinas/metabolismo , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Lecitinas/química , Fígado/metabolismo , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Glycine max/química
11.
Int J Mol Sci ; 20(3)2019 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-30708965

RESUMO

Iron toxicity is associated with organ injury and has been reported in various clinical conditions, such as hemochromatosis, thalassemia major, and myelodysplastic syndromes. Therefore, iron chelation therapy represents a pivotal therapy for these patients during their lifetime. The aim of the present study was to assess the iron chelating properties of α-lipoic acid (ALA) and how such an effect impacts on iron overload mediated toxicity. Human mesenchymal stem cells (HS-5) and animals (zebrafish, n = 10 for each group) were treated for 24 h with ferric ammonium citrate (FAC, 120 µg/mL) in the presence or absence of ALA (20 µg/mL). Oxidative stress was evaluated by reduced glutathione content, reactive oxygen species formation, mitochondrial dysfunction, and gene expression of heme oxygenase-1b and mitochondrial superoxide dismutase; organ injury, iron accumulation, and autophagy were measured by microscopical, cytofluorimetric analyses, and inductively coupled plasma‒optical mission Spectrometer (ICP-OES). Our results showed that FAC results in a significant increase of tissue iron accumulation, oxidative stress, and autophagy and such detrimental effects were reversed by ALA treatment. In conclusion, ALA possesses excellent iron chelating properties that may be exploited in a clinical setting for organ preservation, as well as exhibiting a good safety profile and low cost for the national health system.


Assuntos
Compostos Férricos/efeitos adversos , Quelantes de Ferro/administração & dosagem , Sobrecarga de Ferro/tratamento farmacológico , Compostos de Amônio Quaternário/efeitos adversos , Ácido Tióctico/administração & dosagem , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Glutationa/metabolismo , Heme Oxigenase-1/genética , Humanos , Quelantes de Ferro/farmacologia , Sobrecarga de Ferro/induzido quimicamente , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/genética , Ácido Tióctico/farmacologia , Peixe-Zebra
12.
Genomics ; 111(6): 1504-1513, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30391296

RESUMO

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.


Assuntos
Bombyx/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Insetos/genética , Sobrecarga de Ferro/genética , Ferro da Dieta/administração & dosagem , Ferro/administração & dosagem , Transcriptoma/efeitos dos fármacos , Animais , Bombyx/efeitos dos fármacos , Feminino , Proteínas de Insetos/metabolismo , Sobrecarga de Ferro/fisiopatologia , Masculino
13.
Artif Cells Nanomed Biotechnol ; 46(sup3): S18-S27, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30033763

RESUMO

Early evaluation of iron overload (IO) and prompt iron-chelation therapy reduce the haematopoietic damage wrought by IO-induced reactive oxygen species (ROS). We examined whether MagA could simultaneously increase the sensitivity of magnetic resonance imaging (MRI) for iron measurement and attenuate oxidative damage to the haematopoietic microenvironment. After generation of a transgenic (Tg) mouse model, MRI, transmission electron microscopy and cytotoxicity assays were used to assess various parameters in mesenchymal stem cells (MSCs). Transverse relaxation rate (R2*) of MagA-expressing MSCs in the presence of iron supplement was higher compared with that of control cells. Besides, R2* value of liver from IO magA Tg mice was higher than that of wild type mice. Moreover, MagA contributed to reduce the cytotoxicity of iron against MSCs, reduce expression of p-p38 mitogen-activated protein kinase and ferritin, and reduce inhibition of the osteogenic differentiation caused by IO. These data support the use of magA as a reporter gene for cell tracking with MRI and indicate exciting new possibilities for use of MagA in the attenuation of injury due to oxidative stress caused by exogenous iron.


Assuntos
Proteínas de Bactérias , Medula Óssea , Proteínas de Transporte de Cátions , Genes Reporter , Hematopoese , Sobrecarga de Ferro , Ferro/metabolismo , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Medula Óssea/diagnóstico por imagem , Medula Óssea/metabolismo , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Sobrecarga de Ferro/diagnóstico por imagem , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Camundongos , Camundongos Transgênicos
14.
Free Radic Biol Med ; 115: 127-135, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29157668

RESUMO

Iron promotes formation of hydroxyl radicals by the Fenton reaction, subsequently leading to potential oxidatively generated damage of nucleic acids. Oxidatively generated damage to RNA, measured as 8-oxo-7,8-dihydroguanosine (8-oxoGuo) in urine, is increased in patients with genetic iron overload, which have led us to test the hypothesis that high iron status, assessed by iron biomarkers and genetic disposition, increases urinary excretion of 8-oxoGuo. In a general Danish population study we used a Mendelian randomization design with HFE genotypes as a proxy for iron status and supplemented with ex vivo experiments in mice muscle tissue exposed to iron(II) sulfate to attempt to clarify this hypothesis. The biomarkers ferritin, transferrin, and transferrin saturation (TS) were associated with 8-oxoGuo (in linear univariable and multivariable regression analyses: P < 0.001). Mendelian randomization indicated a causal pathway between genetically elevated iron biomarkers (assessed by ferritin and TS) and high levels of 8-oxoGuo. The ex vivo experiments showed a monotonically increase in 8-oxoGuo with increased iron concentration (ANOVA: P = 0.0008) that was prevented with iron chelation (P = 0.01). Our results indicate a causal relationship between iron biomarkers and 8-oxoGuo. Furthermore, the ex vivo experiment shows a mechanistic link between iron and 8-oxoGuo formation. Both iron overload and the biomarker 8-oxoGuo have been linked to e.g. diabetes, which merits future studies to investigate if iron induced 8-oxoGuo is involved in disease development.


Assuntos
Biomarcadores/urina , Diabetes Mellitus/metabolismo , Genótipo , Guanosina/análogos & derivados , Proteína da Hemocromatose/genética , Hemocromatose/metabolismo , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Adulto , Idoso , Animais , Estudos Transversais , Dinamarca , Diabetes Mellitus/genética , Feminino , Guanosina/urina , Hemocromatose/genética , Humanos , Sobrecarga de Ferro/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Oxirredução , Polimorfismo de Nucleotídeo Único , RNA/metabolismo
15.
Ann Hematol ; 96(9): 1541-1546, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28707012

RESUMO

We retrospectively evaluated the relationship between serum transferrin receptor-1 (sTfR1) and some fundamental events in the life and the management (the age at diagnosis, the age at the first red blood cells transfusion, the age at splenectomy, and the overall need of chelation therapy) of 111 patients with non-transfusion-dependent thalassemia (NTDT) subdivided in four genetic entities: patients with homozygous or compound heterozygous state for ß-thalassemia, patients with triplicated α genotype associated with ß heterozygosity, patients with deletional HbH, and patients with the combination of a ß defect plus a ß chain variant. We found that the group with homozygous or compound heterozygous state for ß-thalassemia had the highest sTfR1 levels and that the presence of increased sTfR1 levels (>5 times normal) was associated with a complex and severe history of disease requiring splenectomy, occasional red blood cells transfusions, and early start and continuous iron chelation therapy.The complexity in the management of NTDT patients is an emerging issue due to the wide heterogeneity of clinical behavior. Our data indicate that the measurement of sTfR1 levels, a common laboratory test, could contribute to correctly stratify disease history and the iron chelation strategy in NTDT patients.


Assuntos
Antígenos CD/sangue , Sobrecarga de Ferro/sangue , Sobrecarga de Ferro/diagnóstico , Receptores da Transferrina/sangue , Talassemia beta/sangue , Adolescente , Adulto , Fatores Etários , Antígenos CD/genética , Criança , Pré-Escolar , Transfusão de Eritrócitos/efeitos adversos , Feminino , Humanos , Quelantes de Ferro/administração & dosagem , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/genética , Masculino , Pessoa de Meia-Idade , Receptores da Transferrina/genética , Talassemia beta/genética , Talassemia beta/terapia
16.
Int J Med Sci ; 14(3): 275-283, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28367088

RESUMO

Iron overload has recently been associated with the changes in the bone microstructure that occur in osteoporosis. However, the effect of iron overload on osteoblasts is unclear. The purpose of this study was to explore the function of divalent metal transporter 1 (DMT1) in the pathological processes of osteoporosis. Osteoblast hFOB1.19 cells were cultured in medium supplemented with different concentrations (0, 50, 100, 200, 300, 400, 500 µmol/L) of ferric ammonium citrate (FAC) as a donor of ferric ions. We used western blotting and immunofluorescence to determine the levels of DMT1 after treatment with FAC. Apoptosis was evaluated by detecting the levels of cleaved caspase 3, BCL2, and BAX with western blotting. Autophagy was evaluated by detecting the levels of LC3 with western blotting and immunofluorescence. Beclin-1 expression was also assessed with western blotting. The autophagy inhibitor 3-methyladenine was used to determine whether autophagy affects the apoptosis induced by FAC. Our results show that FAC increased the levels of DMT1, upregulated the expression of BCL2, and downregulated the apoptosis-related proteins cleaved caspase 3 and BAX. Both LC3I/LC3II levels and beclin-1 were also increased, indicating that FAC increases the accumulation of autophagosomes in hFOB1.19 cells. FAC-induced autophagy was increased by the apoptosis inhibitor 3-MA but was reduced in DMT1 shRNA hFOB1.19 cells. These results suggest that the increased expression of DMT1 induces iron overload and iron overload induces osteoblast autophagy and apoptosis, thus affecting the pathological processes of osteoporosis. Clarifying the mechanisms underlying the effects of DMT1 will allow the identification of novel targets for the prevention and treatment of osteoporosis.


Assuntos
Apoptose/genética , Autofagia/genética , Osteoblastos/metabolismo , Osteoporose/genética , Fatores de Transcrição/genética , Caspase 3/biossíntese , Compostos Férricos/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ferro/administração & dosagem , Ferro/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Osteoblastos/patologia , Osteoporose/metabolismo , Osteoporose/patologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Compostos de Amônio Quaternário/administração & dosagem , Proteína X Associada a bcl-2/biossíntese
17.
Int J Hematol ; 105(3): 353-360, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27848180

RESUMO

Transfusion is believed to be the main cause of iron overload in Japan. A nationwide survey on post-transfusional iron overload subsequently led to the establishment of guidelines for iron chelation therapy in this country. To date, however, detailed clinical information on the entire iron overload population in Japan has not been fully investigated. In the present study, we obtained and studied detailed clinical information on the iron overload patient population in Japan. Of 1109 iron overload cases, 93.1% were considered to have occurred post-transfusion. There were, however, 76 cases of iron overload of unknown origin, which suggest that many clinicians in Japan may encounter some difficulty in correctly diagnosing and treating iron overload. Further clinical data were obtained for 32 cases of iron overload of unknown origin; median of serum ferritin was 1860.5 ng/mL. As occurs in post-transfusional iron overload, liver dysfunction was found to be as high as 95.7% when serum ferritin levels exceeded 1000 ng/mL in these patients. Gene mutation analysis of the iron metabolism-related genes in 27 cases of iron overload with unknown etiology revealed mutations in the gene coding hemojuvelin, transferrin receptor 2, and ferroportin; this indicates that although rare, hereditary hemochromatosis does occur in Japan.


Assuntos
Sobrecarga de Ferro/epidemiologia , Sobrecarga de Ferro/etiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Ferritinas/sangue , Hemocromatose/diagnóstico , Hemocromatose/epidemiologia , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/genética , Japão/epidemiologia , Hepatopatias/etiologia , Masculino , Redes e Vias Metabólicas/genética , Pessoa de Meia-Idade , Mutação , Inquéritos e Questionários , Reação Transfusional , Adulto Jovem
18.
Hematology ; 21(1): 46-53, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26288192

RESUMO

OBJECTIVES: Estimating the prevalence of glutathione S-transferase gene polymorphism (GSTM1) null genotype among patients with beta thalassemia major (ß-TM) in relation to myocardial status assessed by tissue Doppler and cardiac siderosis assessed by cardiac magnetic resonance imaging (MRI) T2*. METHODS: Hundred patients with ß-TM and 100 healthy controls were enrolled. Complete blood count (CBC), mean serum ferritin and GSTM1 genotyping, echocardiography, tissue Doppler, and cardiac MRI T2* were done. RESULTS: Serum ferritin ranged from 1200 to 8000 ng/ml, and mean T2* value was 27.10 ± 11.20 ms. Of patients, 68 (68%) had no cardiac siderosis, while 24 (24%) with mild to moderate, and 8 (8%) with sever cardiac siderosis. T2* values were not correlated with serum ferritin (r = -0.09, P = 0.50). GSTM1 null genotype was prevalent in 46% of patients and 40% of controls (P = 0.69). Patients with null genotype had significantly shorter T2* (P = 0.001), higher left ventricular end-diastolic diameter (P = 0.002), and shorter ejection time (P = 0.005) with no significant relation to serum ferritin (P = 0.122). GSTM1 null genotype was the only predictor for cardiac iron overload (P = 0.002). DISCUSSION: Serum ferritin concentrations have been shown to correlate poorly with all stages of cardiac dysfunction. Low cardiac MRI T2* values occur in patients with ß-TM despite good chelation therapy, suggesting a possible role of genetic factors in cardiac siderosis. CONCLUSION: GSTM1 null genotype is significantly associated with cardiac iron overload independent of serum ferritin in Egyptian patients with ß-TM.


Assuntos
Glutationa Transferase/genética , Sobrecarga de Ferro/genética , Ferro/metabolismo , Polimorfismo Genético , Siderose/genética , Talassemia beta/terapia , Adolescente , Estudos de Casos e Controles , Criança , Egito , Feminino , Ferritinas/sangue , Ferritinas/genética , Expressão Gênica , Genótipo , Glutationa Transferase/deficiência , Humanos , Sobrecarga de Ferro/etiologia , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Índice de Gravidade de Doença , Siderose/etiologia , Siderose/metabolismo , Siderose/patologia , Reação Transfusional , Talassemia beta/genética , Talassemia beta/patologia
19.
Expert Rev Gastroenterol Hepatol ; 10(3): 359-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26561304

RESUMO

A number of human disorders are related to chronic iron overload, either of genetic or acquired origin. The multi-organ damage produced by iron excess leads, in adults and in children, to severe clinical consequences, affecting both quality of life and life expectancy. The diagnosis is increasingly based on a non-invasive strategy, resorting to clinical, biological and imaging data. The treatment rests on either venesection or chelation therapy, depending on the etiology. Major advances in the fields of molecular biology, pharmacology, and biotechnology pave the road for key improvements in the diagnostic and therapeutic management of the patients.


Assuntos
Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/diagnóstico , Sobrecarga de Ferro/terapia , Flebotomia , Biomarcadores/sangue , Doença Crônica , Predisposição Genética para Doença , Humanos , Ferro/sangue , Quelantes de Ferro/efeitos adversos , Sobrecarga de Ferro/sangue , Sobrecarga de Ferro/genética , Fenótipo , Flebotomia/efeitos adversos , Valor Preditivo dos Testes , Fatores de Risco , Resultado do Tratamento
20.
Rev Prat ; 65(10): 1305-11, 2015 Dec.
Artigo em Francês | MEDLINE | ID: mdl-26979029

RESUMO

Chronic iron overload, either of genetic (hemochromatoses) or acquired (transfusions) origin, leads to frequent disorders, affecting both the quality of life and life expectancy. Major recent advances in the knowledge of iron metabolism, together with advances in biology, imaging and drug design have already significantly improved the diagnostic and therapeutic approaches. These conceptual and technological ameliorations should, in the near future, continue to benefit the clinical management of iron overloaded patients.


Assuntos
Suplementos Nutricionais , Hepcidinas/administração & dosagem , Quelantes de Ferro/administração & dosagem , Sobrecarga de Ferro/diagnóstico , Sobrecarga de Ferro/tratamento farmacológico , Qualidade de Vida , Reação Transfusional , Predisposição Genética para Doença , Hemocromatose/genética , Humanos , Sobrecarga de Ferro/etiologia , Sobrecarga de Ferro/genética , Expectativa de Vida , Prognóstico , Fatores de Risco , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA