Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
PLoS One ; 17(7): e0271973, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35895739

RESUMO

BACKGROUND: Screening program participants with iron overload (IO) phenotypes without HFE p.C282Y/p.C282Y are incompletely characterized. METHODS: We studied white participants who had IO phenotypes without p.C282Y/p.C282Y in post-screening clinical examinations (CE). We defined IO phenotypes as a) elevated serum ferritin (SF) and transferrin saturation (TS) at screening and CE, and b) absence of IO treatment, anemia, transfusion >10 units, alcohol intake >30 g/d, hepatitis B or C, and pregnancy. We defined IO-related disease as elevated alanine or aspartate aminotransferase (ALT/AST) or swelling/tenderness of 2nd/3rd metacarpophalangeal (MCP) joints. All participants had HFE p.C282Y and p.H63D genotyping. RESULTS: There were 32 men and 26 women (mean age 54±16 y). Median food/supplemental iron intakes were 14.3/0.0 mg/d. Relative risks of HFE genotypes were 12.9 (p.C282Y/p.H63D), 3.0 (p.H63D/p.H63D), 1.9 (p.C282Y/wt), 0.9 (p.H63D/wt), and 0.5 (wt/wt) compared to 42,640 white screening participants without IO phenotypes or p.C282Y/p.C282Y. Regression on SF revealed positive associations: MCV (p = 0.0006; ß coefficient = 0.4531); swelling/tenderness of MCP joints (p = 0.0033; ß = 0.3455); and p.H63D/wt (p = 0.0015; ß = 0.4146). IO-related disease (18 elevated ALT/AST, one swelling/tenderness of MCP joints) occurred in 19 participants (7 men, 12 women). Median MCV was higher in participants with IO-related disease (97 fL vs. 94 fL; p = 0.0007). Logistic regression on IO-related disease revealed a significant association with diabetes (p = 0.0416; odds ratio 18.9 (95% confidence interval 1.0, 341.1)). CONCLUSIONS: In the present 58 screening program participants who had IO phenotypes without HFE p.C282Y/p.C282Y, relative risks of HFE genotypes p.C282Y/p.H63D, p.H63D/p.H63D, and p.C282Y/wt were significantly higher than in 42,640 white screening participants with neither IO phenotypes nor p.C282Y/p.C282Y. SF was significantly associated with MCV, swelling/tenderness of 2nd/3rd MCP joints, and p.H63D/wt. IO-related disease was significantly associated with MCV and diabetes.


Assuntos
Hemocromatose , Sobrecarga de Ferro , Adulto , Idoso , Feminino , Ferritinas , Genótipo , Hemocromatose/complicações , Proteína da Hemocromatose/genética , Humanos , Sobrecarga de Ferro/diagnóstico , Masculino , Pessoa de Meia-Idade , Fenótipo , Transferrina/genética
2.
Diabetes Metab Res Rev ; 37(4): e3448, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33738915

RESUMO

AIMS: Hereditary haemochromatosis (HH) is a genetic disorder characterised by systemic iron overload and can lead to end-organ failure. However, very few data on this disorder, especially those on endocrine gland involvement in Chinese populations, are currently available. This study aimed to analyse the clinical features of endocrinopathies in patients with HH to generate concern among endocrinologists and improve the management of this disorder. MATERIALS AND METHODS: Chinese patients with HH-related endocrine dysfunction were enrolled at Peking Union Medical College Hospital from January 2010 to December 2018. All clinical data were analysed and summarised. RESULTS: A total of six patients were enrolled in this study, comprising five men and one woman; the average age was 36.5 ± 13.3 years. Mean serum ferritin concentration was 4508.8 ± 1074.3 ng/ml, and median transferrin saturation was 97.9% (96.6%-110.0%). Endocrine gland involvement associated with HH included the pancreas (5/6 patients), the adenohypophysis (5/6 patients) and the bones (1/6 patients); secondary endocrinopathies consisted of diabetes mellitus, hypogonadism, adrenal insufficiency and osteoporosis. Based on phlebotomy and iron chelation therapy, five patients were treated with exogenous insulin preparations, and three patients were treated with exogenous sex hormone replacement therapy. The clinical symptoms of five patients improved, although one patient died of hepatic encephalopathy and multiple organ failure. CONCLUSIONS: HH can cause multiple endocrinopathies. The possibility of HH should be carefully considered in patients with endocrine gland dysfunctions and concomitant elevated serum ferritin levels. Endocrine gland function should also be assessed and followed up in patients with a clear diagnosis of HH.


Assuntos
Doenças do Sistema Endócrino , Hemocromatose , Adulto , China/epidemiologia , Doenças do Sistema Endócrino/sangue , Doenças do Sistema Endócrino/complicações , Feminino , Ferritinas/sangue , Hemocromatose/sangue , Hemocromatose/complicações , Humanos , Masculino , Pessoa de Meia-Idade
3.
Eur J Clin Nutr ; 75(12): 1771-1780, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33712721

RESUMO

There has been no established food and nutrition guidance for diseases characterized by the presence of iron overload (IOL) yet. Hepcidin is a hormone that diminishes iron bioavailability. Its levels increase in response to increased iron stores. Hence, IOL conditions could hypothetically trigger a self-regulatory mechanism for the reduction of the intestinal absorption of iron. In addition, some food substances may modulate intestinal iron absorption and may be useful in the dietary management of patients with IOL. This scoping review aimed to systematize studies that support dietary prescriptions for IOL patients. It was carried out according to the method proposed by the Joanna Briggs Institute and the preferred reporting items for systematic reviews and meta-analyses (PRISMA). Although the need to restrict iron in the diet of individuals with hemochromatosis is quite clear, there is a consensus that IOL diminishes the rate of iron absorption. Reduced iron absorption is also present and has been reported in some diseases with transfusion IOL, in which serum hepcidin is usually high. The consumption of polyphenols and 6-shogaol seems to reduce iron absorption or serum ferritin concentration, while procyanidins do not cause any changes. Vitamin C deficiency is often found in IOL patients. However, vitamin C supplementation and alcohol consumption should be avoided not only because they increase iron absorption, but also because they provoke toxic oxidative reactions when the iron is excessive. Dietary approaches must consider the differences in the pathophysiology and treatment of IOL diseases.


Assuntos
Hemocromatose , Sobrecarga de Ferro , Ferritinas , Hemocromatose/complicações , Hepcidinas/metabolismo , Humanos , Absorção Intestinal , Ferro/metabolismo , Sobrecarga de Ferro/etiologia , Ferro da Dieta/efeitos adversos , Nutrientes
4.
Rural Remote Health ; 19(2): 4844, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31054269

RESUMO

Iron can accumulate in the body due to several causes, resulting in iron overload syndrome. The most common cause is hereditary haemochromatosis (HH), a genetic disorder triggered by inactivation of the iron hormone hepcidin, which results in hyperferraemia and excessive tissue iron deposition. Other causes include repeated blood transfusion, iron-loading anaemias and some chronic liver diseases. Left undiagnosed, HH can cause significant damage to the liver, heart, pancreas and joints, because excess iron is toxic. This also increases the risk of hepatocellular carcinoma, especially in those with cirrhosis of the liver, with an estimate of 1 in 10 HH patients affected. The risk of developing type 2 diabetes is increased by 2.5-7.1 times compared with non-diabetic patients. Haemochromatosis is usually considered when elevated serum ferritin and transferrin saturation levels are found. Ferritin in excess of 300 ng/mL usually indicates iron overload. Genetic testing can identify the two most common mutations in the HFE gene - a positive result confirms the diagnosis of haemochromatosis - but there are also rare forms of the disease unrelated to HFE mutations. Liver biopsy can be used to ascertain iron accumulation and histological presence of fibrosis (cirrhosis). Assessment of the hepatic iron index is considered the gold standard for diagnosis of haemochromatosis. Magnetic resonance imaging has been used as a non-invasive alternative to accurately estimate iron deposition levels in the liver, heart, joints and pituitary gland. Population screening is not recommended; however, family members of identified people should be screened to determine their phenotypic or carrier potential. Early diagnosis enables preventative measures to be commenced. Routine treatment is by regular venesection of 500 mL of whole blood per session. An initiation phase of weekly or twice-weekly venesection is common until serum ferritin (SF) is reduced to normal. When SF and other markers are within normal range, regular venesections are usually scheduled 1-3 months apart, depending on the underlying cause and SF response. Dietary iron including red meat and fortified foods such as cereals should be avoided. Vitamin C promotes iron absorption, and supplementation should be avoided, as should alcohol, which can increase the risk of concomitant liver disease. John's story outlines a typical journey through diagnosis, treatment and care during HH while living on Arran, an island off the coast of Scotland. Subsequently, John developed hepatocellular carcinoma, and his treatment and palliative care are described. We wrote this article to give the reader an insight to this silent disorder and the value of recognising the signs and symptoms for early diagnosis and subsequent treatment.


Assuntos
Carcinoma Hepatocelular/complicações , Hemocromatose/complicações , Hemocromatose/diagnóstico , Neoplasias Hepáticas/complicações , Adulto , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/terapia , Hemocromatose/terapia , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/terapia , Masculino
6.
Nat Rev Dis Primers ; 4: 18016, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29620054

RESUMO

Haemochromatosis is defined as systemic iron overload of genetic origin, caused by a reduction in the concentration of the iron regulatory hormone hepcidin, or a reduction in hepcidin-ferroportin binding. Hepcidin regulates the activity of ferroportin, which is the only identified cellular iron exporter. The most common form of haemochromatosis is due to homozygous mutations (specifically, the C282Y mutation) in HFE, which encodes hereditary haemochromatosis protein. Non-HFE forms of haemochromatosis due to mutations in HAMP, HJV or TFR2 are much rarer. Mutations in SLC40A1 (also known as FPN1; encoding ferroportin) that prevent hepcidin-ferroportin binding also cause haemochromatosis. Cellular iron excess in HFE and non-HFE forms of haemochromatosis is caused by increased concentrations of plasma iron, which can lead to the accumulation of iron in parenchymal cells, particularly hepatocytes, pancreatic cells and cardiomyocytes. Diagnosis is noninvasive and includes clinical examination, assessment of plasma iron parameters, imaging and genetic testing. The mainstay therapy is phlebotomy, although iron chelation can be used in some patients. Hepcidin supplementation might be an innovative future approach.


Assuntos
Hemocromatose/complicações , Hemocromatose/terapia , Proteínas de Transporte de Cátions/toxicidade , Terapia por Quelação/métodos , Hemocromatose/epidemiologia , Humanos , Ferro/toxicidade , Imageamento por Ressonância Magnética/métodos , Programas de Rastreamento/métodos , Flebotomia/métodos , Polimorfismo Genético/genética , Qualidade de Vida/psicologia
7.
Int Heart J ; 59(2): 427-430, 2018 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-29563373

RESUMO

A 31-year-old man was admitted to our hospital with atrial tachycardia and cardiogenic shock. He had been diagnosed with hereditary spherocytosis (HS) during childhood, but he never received any red blood cell transfusions. Right ventricular endomyocardial biopsy revealed multiple myocardial hemosiderin deposits, and he was diagnosed with cardiac hemochromatosis. In addition to the iron deposition in the heart, the loss of myocyte and severe interstitial fibrosis were present. His cardiac function did not improve even after the cardioversion for atrial tachycardia, and he suffered from recurrent heart failure. Despite intensive medical treatment for heart failure and arrhythmias in combination with iron chelation therapy, he eventually died of progressive and refractory heart failure. Hemochromatosis is a systemic disorder characterized by the excessive deposition of iron in multiple organs. The occurrence of hemochromatosis in HS is extremely rare, and previous reports have shown that the coexistence of heterozygosity for the HFE gene mutation in HS patients causes excess iron storage. The prognosis is poor due to progressive congestive heart failure and refractory arrhythmias. Here we report a rare case of fatal cardiac hemochromatosis associated with HS. The possibility of cardiac hemochromatosis needs to be considered in cases of heart failure or arrhythmia in patients with HS.


Assuntos
Insuficiência Cardíaca/etiologia , Hemocromatose/complicações , Esferocitose Hereditária/complicações , Adulto , Evolução Fatal , Humanos , Masculino
8.
J Med Case Rep ; 12(1): 18, 2018 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-29373985

RESUMO

BACKGROUND: Juvenile hemochromatosis is the most severe form of iron overloading phenotype. Although rare, it should be suspected in patients who present with hypogonadotropic hypogonadism, diabetes mellitus, or cardiomyopathy without a clear cause. CASE PRESENTATION: A young Serbian male presenting with end-stage heart failure was referred for extracorporeal membrane oxygenation. An endomyocardial biopsy revealed cytoplasmic iron deposits in myocytes. His condition was stabilized with biventricular assist devices and he was listed for heart transplantation. Iron chelation therapy was commenced and resulted in rapid removal of iron burden. Serial outpatient echocardiograms demonstrated myocardial recovery such that a successful biventricular assist device explant occurred 131 days after initial implant. Targeted gene sequencing revealed a loss-of-function mutation within the HJV gene, which is consistent with juvenile hemochromatosis. CONCLUSIONS: This rare case of a patient with juvenile hemochromatosis associated with a HJV mutation provides histologic evidence documenting the reversal of associated end-stage heart failure, requiring emergent mechanical circulatory support, with iron chelation therapy.


Assuntos
Terapia por Quelação , Desferroxamina/uso terapêutico , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/terapia , Hemocromatose/terapia , Quelantes de Ferro/uso terapêutico , Adulto , Biópsia , Ecocardiografia , Ferritinas/sangue , Insuficiência Cardíaca/etiologia , Ventrículos do Coração/patologia , Hemocromatose/complicações , Hemocromatose/diagnóstico , Hemocromatose/genética , Proteína da Hemocromatose , Humanos , Fígado/patologia , Mutação com Perda de Função , Masculino , Tomografia Computadorizada por Raios X
9.
World J Gastroenterol ; 23(40): 7337-7342, 2017 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-29142481

RESUMO

We report a case involving a rescued low birth weight infant (LBWI) with acute liver failure. CASE: The patient was 1594 g and 323/7 gestational wk at birth. At the age of 11 d, she developed acute liver failure due to gestational alloimmune liver disease. Exchange transfusion and high-dose gamma globulin therapy were initiated, and body weight increased with enteral nutrition. Exchange transfusion was performed a total of 33 times prior to living donor liver transplantation (LDLT). Her liver dysfunction could not be treated by medications alone. At 55 d old and a body weight of 2946 g, she underwent LDLT using an S2 monosegment graft from her mother. Three years have passed with no reports of intellectual disability or liver dysfunction. LBWIs with acute liver failure may be rescued by LDLT after body weight has increased to over 2500 g.


Assuntos
Hemocromatose/terapia , Recém-Nascido de Baixo Peso , Recém-Nascido Prematuro , Hipertensão Intra-Abdominal/terapia , Falência Hepática Aguda/terapia , Transplante de Fígado/efeitos adversos , Biópsia , Nutrição Enteral , Transfusão Total , Feminino , Rejeição de Enxerto/tratamento farmacológico , Hemocromatose/sangue , Hemocromatose/complicações , Humanos , Terapia de Imunossupressão/métodos , Lactente , Recém-Nascido , Hipertensão Intra-Abdominal/etiologia , Fígado/patologia , Fígado/cirurgia , Falência Hepática Aguda/sangue , Falência Hepática Aguda/etiologia , Falência Hepática Aguda/patologia , Transplante de Fígado/métodos , Doadores Vivos , Insuficiência Respiratória/etiologia , Insuficiência Respiratória/terapia , Resultado do Tratamento , gama-Globulinas/uso terapêutico
10.
Swiss Med Wkly ; 147: w14550, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29120023

RESUMO

Hyperferritinaemia is a frequent clinical problem. Elevated serum ferritin levels can be detected in different genetic and acquired diseases and can occur with or without anaemia. It is therefore important to determine whether hyperferritinaemia is due to iron overload or due to a secondary cause. The main causes of iron overload are intestinal iron hyperabsorption disorders and transfusion-dependent disorders. Iron homeostasis and iron overload are quantified by different diagnostic approaches. The evaluation of serum ferritin and transferrin saturation is the first diagnostic step to identify the cause of hyperferritinaemia. The assessment of liver iron concentration by liver biopsy or magnetic resonance imaging (MRI) may guide the further diagnostic and therapeutic workup. Liver biopsy is invasive and poorly accepted by patients and should only be carried out in selected patients with hereditary haemochromatosis. As a non-invasive approach, MRI is considered the standard method to diagnose and to monitor both hepatic iron overload and the effectiveness of iron chelation therapy in many clinical conditions such as thalassaemia and myelodysplastic syndromes. Accurate evaluation and monitoring of iron overload has major implications regarding adherence, quality of life and prognosis. There are different technical MRI approaches to measuring the liver iron content. Of these, T2 and T2* relaxometry are considered the standard of care. MRI with cardiac T2* mapping is also suitable for the assessment of cardiac iron. Currently there is no consensus which technique should be preferred. The choice depends on local availability and patient population. However, it is important to use the same MRI technique in subsequent visits in the same patient to get comparable results. Signal intensity ratio may be a good adjunct to R2 and R2* methods as it allows easy visual estimation of the liver iron concentration. In this review a group of Swiss haematologists and radiologists give an overview of different conditions leading to primary or secondary iron overload and on diagnostic methods to assess hyperferritinaemia with a focus on the role of liver MRI. They summarise the standard practice in Switzerland on the use of liver iron concentration MRI as well as disease-specific guideline recommendations.


Assuntos
Ferritinas/efeitos adversos , Sobrecarga de Ferro/diagnóstico , Imageamento por Ressonância Magnética/métodos , Biópsia , Feminino , Ferritinas/sangue , Hemocromatose/sangue , Hemocromatose/complicações , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/etiologia , Fígado/patologia , Masculino , Suíça , Talassemia/sangue , Talassemia/complicações
11.
Drug Discov Ther ; 10(3): 177-80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27301711

RESUMO

Non-arteritic posterior ischemic optic neuropathy (NA-PION) is a disorder involving reduced blood flow to the retrobulbar portion of the optic nerve. This disorder usually develops acutely, and research has suggested that high-dose steroid therapy soon after the onset of visual loss can result in significant visual improvement. This treatment, however, is not universally successful. The addition of a potent vasodilator could help to restore ocular blood flow. This case report describes the use of prostaglandin E1 (PGE1), a powerful vasodilator of the microcirculation, to treat three separate episodes of NA-PION over five years in the same patient. A 68-year-old white male was first seen in June 2009 with NA-PION in the left eye, and the condition was treated with steroids and PGE1. The patient had a subsequent episode in July 2010 that was treated with steroids and PGE1 and another in May 2014 that was treated with PGE1 alone. Visual acuity improved from 4/10 to 11/10 in 2009, from 4/10 to 11/10 in 2010, and from 5/10 to 10/10 in 2014. No complications due to the use of PGE1 were noted. PGE1 should be considered as a treatment for NA-PION to immediately restore blood flow and potentially improve vision.


Assuntos
Alprostadil/efeitos adversos , Alprostadil/uso terapêutico , Neuropatia Óptica Isquêmica/induzido quimicamente , Vasodilatadores/efeitos adversos , Vasodilatadores/uso terapêutico , Idoso , Hemocromatose/complicações , Hemocromatose/genética , Humanos , Masculino , Microcirculação/efeitos dos fármacos , Neuropatia Óptica Isquêmica/diagnóstico por imagem , Artéria Retiniana/efeitos dos fármacos , Oclusão da Artéria Retiniana/tratamento farmacológico , Ultrassonografia Doppler em Cores , Acuidade Visual/efeitos dos fármacos
13.
Singapore Med J ; 56(7): e124-6, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26243982

RESUMO

A 39-year-old woman with end-stage renal disease, which was maintained on haemodialysis, developed secondary haemochromatosis after receiving blood transfusions and intravenous iron supplementation without sufficient serum ferritin concentration monitoring. The patient received intravenous deferoxamine three times a week, combined with high-dose recombinant human erythropoietin therapy and haemodialysis. After three months, improvements in biochemical indicators and iron overload were noted.


Assuntos
Ferritinas/sangue , Hemocromatose/complicações , Falência Renal Crônica/complicações , Diálise Renal/efeitos adversos , Adulto , Quelantes/química , Eritropoetina/uso terapêutico , Feminino , Hemoglobinas/análise , Humanos , Falência Renal Crônica/terapia , Proteínas Recombinantes/uso terapêutico , Análise de Sequência de DNA , Tomografia Computadorizada por Raios X , Transferrina/química , Reação Transfusional , Resultado do Tratamento
14.
Eur J Haematol ; 95(6): 545-50, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25684349

RESUMO

This open-label, prospective, phase 2 study evaluated the safety and efficacy of deferasirox (10 ± 5 mg/kg/d) in patients with hereditary hemochromatosis (HH) and iron overload refractory to or intolerant of phlebotomy. Ten patients were enrolled and all completed the 12-month treatment period. There were significant decreases from baseline to end of study (i.e., 12 months) in median serum ferritin (P < 0.001), mean transferrin saturation (P < 0.05), median liver iron concentration (P < 0.001), and mean alanine aminotransferase (P < 0.05). The median time to achieve serum ferritin reduction ≥50% compared to baseline was 7.53 months. The most common adverse events were mild, transient diarrhea (n = 5) and nausea (n = 2). No patient experienced an increase in serum creatinine that exceeded the upper limit of normal. These data confirm that deferasirox was well tolerated and effective in reducing iron burden in patients with hereditary hemochromatosis and could be a safe alternative to phlebotomy in selected patients.


Assuntos
Benzoatos/uso terapêutico , Hemocromatose/complicações , Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/etiologia , Triazóis/uso terapêutico , Adulto , Idoso , Benzoatos/administração & dosagem , Benzoatos/efeitos adversos , Biomarcadores , Deferasirox , Índices de Eritrócitos , Feminino , Hemocromatose/diagnóstico , Hemocromatose/genética , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Quelantes de Ferro/administração & dosagem , Quelantes de Ferro/efeitos adversos , Sobrecarga de Ferro/diagnóstico , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Mutação , Fatores de Tempo , Resultado do Tratamento , Triazóis/administração & dosagem , Triazóis/efeitos adversos
15.
Clin Nucl Med ; 40(6): 538-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25608173

RESUMO

Hereditary hemochromatosis is an autosomal recessive disorder of iron metabolism resulting in toxic accumulation of iron in vital organs. We present a 64-year-old white man with non-Hodgkin lymphoma treated with high-dose chemotherapy and stem cell transplant that was subsequently diagnosed with hereditary hemochromatosis. F-FDG PET/CT was performed as routine follow-up and showed a pathological finding of homogeneous increased liver glucose metabolism. Increased FDG avidity in the liver suggested the presence of damage caused by hemochromatosis.


Assuntos
Fluordesoxiglucose F18 , Hemocromatose/diagnóstico por imagem , Fígado/diagnóstico por imagem , Linfoma não Hodgkin/diagnóstico por imagem , Compostos Radiofarmacêuticos , Hemocromatose/complicações , Humanos , Linfoma não Hodgkin/complicações , Masculino , Pessoa de Meia-Idade , Imagem Multimodal , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X
16.
Am J Physiol Gastrointest Liver Physiol ; 308(4): G251-61, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25501544

RESUMO

Hemojuvelin (Hjv) is a membrane protein that controls body iron metabolism by enhancing signaling to hepcidin. Hjv mutations cause juvenile hemochromatosis, a disease of systemic iron overload. Excessive iron accumulation in the liver progressively leads to inflammation and disease, such as fibrosis, cirrhosis, or hepatocellular cancer. Fatty liver (steatosis) may also progress to inflammation (steatohepatitis) and liver disease, and iron is considered as pathogenic cofactor. The aim of this study was to investigate the pathological implications of parenchymal iron overload due to Hjv ablation in the fatty liver. Wild-type (WT) and Hjv(-/-) mice on C57BL/6 background were fed a standard chow, a high-fat diet (HFD), or a HFD supplemented with 2% carbonyl iron (HFD+Fe) for 12 wk. The animals were analyzed for iron and lipid metabolism. As expected, all Hjv(-/-) mice manifested higher serum and hepatic iron and diminished hepcidin levels compared with WT controls. The HFD reduced iron indexes and promoted liver steatosis in both WT and Hjv(-/-) mice. Notably, steatosis was attenuated in Hjv(-/-) mice on the HFD+Fe regimen. Hjv(-/-) animals gained less body weight and exhibited reduced serum glucose and cholesterol levels. Histological and ultrastructural analysis revealed absence of iron-induced inflammation or liver fibrosis despite early signs of liver injury (expression of α-smooth muscle actin). We conclude that parenchymal hepatic iron overload does not suffice to trigger progression of liver steatosis to steatohepatitis or fibrosis in C57BL/6 mice.


Assuntos
Dieta Hiperlipídica , Fígado Gorduroso/complicações , Hemocromatose/congênito , Ferro/metabolismo , Cirrose Hepática/etiologia , Fígado/metabolismo , Proteínas de Membrana/deficiência , Actinas/metabolismo , Animais , Glicemia/metabolismo , Colesterol/sangue , Modelos Animais de Doenças , Progressão da Doença , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/prevenção & controle , Proteínas Ligadas por GPI , Genótipo , Hemocromatose/complicações , Hemocromatose/genética , Hemocromatose/metabolismo , Proteína da Hemocromatose , Hepcidinas/metabolismo , Ferro/sangue , Compostos de Ferro/farmacologia , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fatores de Risco , Fatores de Tempo , Aumento de Peso
17.
Med Hypotheses ; 83(2): 238-46, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24857772

RESUMO

Iron overload is a big challenge when treating thalassemia (TM), hemochromatosis and sideroblastic anemia. It persists even after cure of TM with bone marrow transplantation. Iron overload results from increased iron absorption and repeated blood transfusions causing increased iron in plasma and interstitial fluids. Iron deposition in tissues e.g. heart, liver, endocrine glands and others leads to tissue damage and organ dysfunction. Iron chelation therapy and phlebotomy for iron overload have treatment difficulties, side effects and contraindications. As mean iron level in skin of TM patients increases by more than 200%, percutaneous iron excretion may be beneficial. Wet cupping therapy (WCT) is a simple, safe and economic treatment. WCT is a familiar treatment modality in some European countries and in Chinese hospitals in treating different diseases. WCT was reported to clear both blood plasma and interstitial spaces from causative pathological substances (CPS). Standard WCT method is Al-hijamah (cupping, puncturing and cupping, CPC) method of WCT that was reported to clear blood and interstitial fluids better than the traditional WCT (puncturing and cupping method, PC method of WCT). In other word, traditional WCT may be described as scarification and suction method (double S technique), while Al-hijamah may be described as suction, scarification and suction method (triple S technique). Al-hijamah is a more comprehensive treatment modality that includes all steps and therapeutic benefits of traditional dry cupping therapy and WCT altogether according to the evidence-based Taibah mechanism (Taibah theory). During the first cupping step of Al-hijamah, a fluid mixture is collected inside skin uplifting due to the effect of negative pressure inside sucking cups. This fluid mixture contains collected interstitial fluids with CPS (iron, ferritin and hemolyzed RBCs in thalassemia), filtered fluids (from blood capillaries) with iron and hemolyzed blood cells (hemolyzed RBCs, WBCs and platelets). That fluid mixture does not contain intact blood cells (having diameters in microns) that are too big to pass through pores of skin capillaries (6-12nm in diameter) and cannot be filtered. Puncturing skin upliftings and applying second cupping step excrete collected fluids. Skin scarifications (shartat mihjam in Arabic) should be small, superficial (0.1mm in depth), short (1-2mm in length), multiple, evenly distributed and confined to skin upliftings. Sucking pressure inside cups (-150 to -420mmHg) applied to skin is transmitted to around skin capillaries to be added to capillary hydrostatic pressure (-33mmHg at arterial end of capillaries and -13mmHg at venous end of capillaries) against capillary osmotic pressure (+20mmHg). This creates a pressure gradient and a traction force across skin and capillaries and increases filtration at arterial end of capillaries at net pressure of -163 to -433mmHg and at venous end of capillaries at net pressure of -143 to -413mmHg resulting in clearance of blood from CPS (iron, ferritin and hemolyzed blood cells). Net filtration pressure at renal glomeruli is 10mmHg i.e. Al-hijamah exerts a more pressure-dependent filtration than renal glomeruli. Al-hijamah may benefit patients through inducing negative iron balance. Interestingly, Al-hijamah was reported to decrease serum ferritin significantly (by about 22%) in healthy subjects while excessive traditional WCT was reported to cause iron deficiency anemia. Al-hijamah is a highly recommended treatment in prophetic medicine. In conclusion, Al-hijamah may be a promising adjuvant treatment for iron overload in TM, hemochromatosis and sideroblastic anemia.


Assuntos
Anemia Sideroblástica/complicações , Sangria/métodos , Eliminação Cutânea/fisiologia , Hemocromatose/complicações , Sobrecarga de Ferro/terapia , Sucção/métodos , Talassemia beta/complicações , Ferritinas/metabolismo , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/etiologia
18.
J Pediatr Hematol Oncol ; 36(4): 281-92, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-23887025

RESUMO

Iron overload in hereditary hemochromatosis and hematologic malignancy has unfavorable effects on morbidity. Herein, 53 children (age 108.4±58.3 mo, 25 girls and 28 boys) with acute myeloblastic and lymphoblastic leukemia, who received 4 different chemotherapy protocols, were evaluated for iron overload throughout chemotherapy. Iron overload arose: (1) before chemotherapy, which was dependent on neither chemotherapy nor packed red blood cell transfusions and (2) after chemotherapy, which was dependent on the duration and nature of chemotherapy and partially on transfusion of packed red blood cells. Iron overload was documented in 75% of patients with a ferritin level >1000 ng/mL, by liver and heart magnetic resonance imaging, and they were administered iron-chelation therapy with success. Three of 10 radiologically iron-overloaded patients were heterozygous for H63D mutation. Aminolevulinic acid and porphobilinogen levels were normal. Light microscopic examination of the bone marrow revealed increased iron granules in erythroblasts, platelets, and megakaryocytes, iron-laden macrophages, free iron in the matrix, dyshematopoiesis, and apoptotic cells. Electron microscopic examination revealed iron-laden secondary lysosomes and autolysosomes in normoblasts and iron-laden primary granules in promyelocytes, irrelevant to the ferritin level, implying autophagia due to chemotherapy as a source of the excess iron. We think that iron overload, which is an important complication of acute leukemia, should be evaluated separately from "transfusion overload," and the management principles specific to leukemia should be implemented.


Assuntos
Células da Medula Óssea , Medula Óssea , Hemocromatose , Quelantes de Ferro/administração & dosagem , Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Adolescente , Ácido Aminolevulínico/sangue , Medula Óssea/metabolismo , Medula Óssea/ultraestrutura , Células da Medula Óssea/metabolismo , Células da Medula Óssea/ultraestrutura , Criança , Feminino , Ferritinas/sangue , Hemocromatose/sangue , Hemocromatose/complicações , Hemocromatose/tratamento farmacológico , Hemocromatose/genética , Hemocromatose/patologia , Humanos , Ferro/sangue , Quelantes de Ferro/efeitos adversos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Masculino , Mutação de Sentido Incorreto , Porfobilinogênio/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia
20.
Vox Sang ; 97(3): 185-97, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19663936

RESUMO

BACKGROUND: The quantity of iron in body is carefully regulated, primarily by control of iron absorption, and excess total body iron can be extremely toxic. Since humans have no mechanism for elimination of excess iron, multiple transfusions of red blood cells, which are required for the management of a number of disorders, inevitably result in iron overload. Cumulative iron overload, in turn, leads to iron toxicity with organ dysfunction and damage. MATERIALS: This review examines the relationship between iron metabolism and hematologic disorders treated with multiple transfusions, with emphasis on the diagnosis and current methods of management of iron overload and toxicity in transfusion-dependent patients. Primarily using key words, we identified and reviewed more than 100 pertinent articles in English and other languages in the Medline database plus an additional number of abstracts of presentations at recent meetings of relevant scientific associations. RESULTS: Transfusion-dependent disorders include those characterized by decreased red blood cell production, increased red blood cell destruction, or chronic blood loss. Patients receiving chronic transfusion therapy should be screened and monitored for iron overload, yet in our opinion, this is not always done routinely. Once iron overload has been identified, it should be treated to reduce the risk of morbidity and mortality from iron toxicity, which particularly affects the liver and heart. CONCLUSION: Increased awareness of the risks of iron overload from chronic transfusion therapy should result in greater use of interventions such as iron chelation to reduce total body iron and the risk of long-term sequelae.


Assuntos
Sobrecarga de Ferro/etiologia , Reação Transfusional , Anemia/terapia , Cardiomiopatias/etiologia , Ceruloplasmina/deficiência , Terapia por Quelação , Doença Crônica , Hemocromatose/complicações , Humanos , Ferro/efeitos adversos , Ferro/metabolismo , Ferro/farmacocinética , Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/diagnóstico , Sobrecarga de Ferro/fisiopatologia , Sobrecarga de Ferro/prevenção & controle , Sobrecarga de Ferro/terapia , Hepatopatias/etiologia , Flebotomia , Risco , Transferrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA