Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 59
1.
J Chemother ; 35(5): 404-410, 2023 Sep.
Article En | MEDLINE | ID: mdl-36322121

Linezolid (LZD) is an effective drug in treating multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis. This study aimed to evaluate the safety of LZD in the treatment of patients with rifampicin resistant tuberculosis. This was a multicenter retrospective study. A total of 184 patients of the rifampicin resistant tuberculosis patients treated with LZD from Jan 2018 to Apr 2020 in three hospitals were involved, and their clinical symptoms were recorded and analyzed. Meanwhile, the types and incidence of adverse effects associated with LZD were evaluated. It showed that peripheral neuritis (51, 27.7%) and hemochromatosis (42, 22.8%) were the most common adverse effects observed among these patients. The median time of symptoms after LZD treatment was 45.5 and 120.0 days, respectively. Furthermore, female patients had a significantly higher risk for leukopenia (P = 0.002) and hemochromatosis (P = 0.033) when compared with male patients. History of underlying disease was the risk factor for thrombocytopenia (P = 0.022). Patients with long duration of medication (RR = 1.004, 95%CI: 1.002-1.006, P < 0.001) and daily dosage ≥600mg (RR = 3.059, 95%CI: 1.238-7.558, P = 0.015) were at higher risk of hemochromatosis. Age was the risk factor for rash (P = 0.008) and nausea and vomiting (P = 0.018). In addition, LZD administration time was the risk factor for optic neuritis (P < 0.001) and peripheral neuritis (P < 0.001). LZD can cause adverse symptoms in patients with rifampicin resistant tuberculosis. Gender, history of underlying disease, LZD use time, LZD dosage, and age are the risk factors in the LZD treatment of these patients. During medication, bone marrow suppression and neuropathy should be closely monitored. This study could potentially provide useful information for the clinical practice.


Drug-Related Side Effects and Adverse Reactions , Hemochromatosis , Neuritis , Tuberculosis, Multidrug-Resistant , Humans , Male , Female , Linezolid/adverse effects , Rifampin/adverse effects , Retrospective Studies , Hemochromatosis/chemically induced , Hemochromatosis/drug therapy , Tuberculosis, Multidrug-Resistant/drug therapy , Neuritis/chemically induced , Neuritis/drug therapy , Antitubercular Agents/adverse effects
2.
J Mol Med (Berl) ; 100(12): 1673-1689, 2022 12.
Article En | MEDLINE | ID: mdl-36214835

Alcohol-associated liver disease (ALD) is one of the most common chronic liver diseases. Its pathological spectrum includes the overlapping stages of hepatic steatosis/steatohepatitis that can progress to liver fibrosis and cirrhosis; both are risk factors for hepatocellular carcinoma. Moreover, ALD diagnosis and management pose several challenges. The early pathological stages are reversible by alcohol abstinence, but these early stages are often asymptomatic, and currently, there is no specific laboratory biomarker or diagnostic test that can confirm ALD etiology. Alcohol consumers frequently show dysregulation of iron and iron-related proteins. Examination of iron-related parameters in this group may aid in early disease diagnosis and better prognosis and management. For this, a coherent overview of the status of iron and iron-related proteins in alcohol consumers is essential. Therefore, here, we collated and reviewed the alcohol-induced alterations in iron and iron-related proteins. Reported observations include unaltered, increased, or decreased levels of hemoglobin and serum iron, increments in intestinal iron absorption (facilitated via upregulations of duodenal divalent metal transporter-1 and ferroportin), serum ferritin and carbohydrate-deficient transferrin, decrements in serum hepcidin, decreased or unaltered levels of transferrin, increased or unaltered levels of transferrin saturation, and unaltered levels of soluble transferrin receptor. Laboratory values of iron and iron-related proteins in alcohol consumers are provided for reference. The causes and mechanisms underlying these alcohol-induced alterations in iron parameters and anemia in ALD are explained. Notably, alcohol consumption by hemochromatosis (iron overload) patients worsens disease severity due to the synergistic effects of excess iron and alcohol.


Fatty Liver , Hemochromatosis , Iron Overload , Liver Diseases, Alcoholic , Humans , Iron/metabolism , Hepcidins , Iron Overload/etiology , Iron Overload/metabolism , Hemochromatosis/chemically induced , Hemochromatosis/complications , Hemochromatosis/metabolism , Receptors, Transferrin/metabolism , Liver Diseases, Alcoholic/metabolism , Fatty Liver/etiology , Ethanol
3.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G966-G979, 2020 05 01.
Article En | MEDLINE | ID: mdl-32308038

Iron overload induces intestinal-permeability defect (gut leakage), and gut translocation of organismal molecules might enhance systemic inflammation and sepsis severity in patients with thalassemia (Thal). Hence, iron administration in Hbbth3/+ mice, heterozygous ß-globin-deficient Thal mice, was explored. Oral iron administration induced more severe secondary hemochromatosis and gut leakage in Thal mice compared with wild-type (WT) mice. Gut leakage was determined by 1) FITC-dextran assay, 2) spontaneous serum elevation of endotoxin (LPS) and (1→3)-ß-d-glucan (BG), molecular structures of gut-organisms, and 3) reduction of tight-junction molecules with increased enterocyte apoptosis (activated caspase-3) by immunofluorescent staining. Iron overload also enhanced serum cytokines and increased Bacteroides spp. (gram-negative bacteria) in feces as analyzed by microbiome analysis. LPS injection in iron-overloaded Thal mice produced higher mortality and prominent cytokine responses. Additionally, stimulation with LPS plus iron in macrophage from Thal mice induced higher cytokines production with lower ß-globin gene expression compared with WT. Furthermore, possible gut leakage as determined by elevated LPS or BG (>60 pg/mL) in serum without systemic infection was demonstrated in 18 out of 41 patients with ß-thalassemia major. Finally, enhanced LPS-induced cytokine responses of mononuclear cells from these patients compared with cells from healthy volunteers were demonstrated. In conclusion, oral iron administration in Thal mice induced more severe gut leakage and increased fecal gram-negative bacteria, resulting in higher levels of endotoxemia and serum inflammatory cytokines compared with WT. Preexisting hyperinflammatory cytokines in iron-overloaded Thal enhanced susceptibility toward infection.NEW & NOTEWORTHY Although the impact of iron accumulation in several organs of patients with thalassemia is well known, the adverse effect of iron accumulation in gut is not frequently mentioned. Here, we demonstrated iron-induced gut-permeability defect, impact of organismal molecules from gut translocation of, and macrophage functional defect upon the increased sepsis susceptibility in thalassemia mice.


Cytokines/metabolism , Duodenum/metabolism , Gastrointestinal Microbiome , Hemochromatosis/metabolism , Inflammation Mediators/metabolism , Iron/metabolism , Macrophages/metabolism , Sepsis/metabolism , beta-Thalassemia/metabolism , Adult , Animals , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Duodenum/immunology , Duodenum/microbiology , Female , Ferric Oxide, Saccharated , Hemochromatosis/chemically induced , Hemochromatosis/immunology , Hemochromatosis/microbiology , Heterozygote , Humans , Lipopolysaccharides , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Permeability , Sepsis/chemically induced , Sepsis/immunology , Sepsis/microbiology , Young Adult , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/immunology , beta-Thalassemia/microbiology
4.
Environ Sci Pollut Res Int ; 25(3): 2007-2013, 2018 Jan.
Article En | MEDLINE | ID: mdl-27628916

Many environmental agents, such as excessive alcohol intake, xenobiotics, and virus, are able to damage the human body, targeting especially the liver. Metal excess may also assault the liver. Thus, chronic iron overload may cause, especially when associated with cofactors, diffuse organ damage that is a source of significant morbidity and mortality. Iron excess can be either of acquired (mostly transfusional) or of genetic origin. Hemochromatosis is the archetype of genetic iron overload diseases and represents a serious health problem. A better understanding of iron metabolism has deeply modified the hemochromatosis field which today benefits from much more efficient diagnostic and therapeutic approaches.


Hemochromatosis/chemically induced , Iron/toxicity , Hemochromatosis/metabolism , Humans , Iron/metabolism , Iron Overload , Liver/drug effects , Liver/metabolism
5.
Medicina (B Aires) ; 77(6): 458-464, 2017.
Article Es | MEDLINE | ID: mdl-29223935

Excess iron (Fe) intake in subjects carrying certain mutations in the HFE gene may result in Fe overload. To estimate risk of Fe overload, 166 male blood donors (19-65 years) from Buenos Aires city were investigated. Daily Fe intake (FeI), hem Fe intake, and Fe intake from SO4Fe enriched flours were estimated (SARA Computer Program and Food Composition Table, USDA). Serum ferritin and transferrin saturation were determined; criteria for Fe overload was serum ferritin > 300 ng/ml and transferrin saturation = 50%. HFE genotypes C282Y, H63D and S65C were analyzed by PCR-RFLP in blood samples. No participant presented FeI lower than the estimated average requirement (6 mg Fe/day) and 3.0% was over the upper level (45 mg Fe/day). Hem Fe and Fe from flour enrichment were 9.4% and 47.7% of daily Fe intake, respectively. A significant association was observed between the increase in serum ferritin (ng/ml) and the increase in FeI (p = 0.0472); 2.3% of the donors presented serum ferritin > 300 ng/ml and transferrin saturation = 50%. Genotypes associated with hereditary hemochromatosis (H63D, S65C and C282Y) were found in 29.3% of the donors. The percentage of transferrin saturation was higher in subjects carrying mutation than in wild type subjects (p = 0.0167). Although penetrance of hereditary hemochromatosis in the studied group was only 1.2%, an excessive Fe intake could enhance adverse effects in individuals unaware of any family history of Fe overload.


Blood Donors/statistics & numerical data , Ferritins/blood , Hemochromatosis Protein/genetics , Hemochromatosis/chemically induced , Hemochromatosis/genetics , Iron, Dietary/administration & dosage , Adult , Genotype , Humans , Iron/blood , Male , Mutation , Polymorphism, Restriction Fragment Length , Transferrin/analysis
6.
Medicina (B.Aires) ; 77(6): 458-464, dic. 2017. graf, tab
Article Es | LILACS | ID: biblio-894521

El consumo excesivo de hierro (Fe) en portadores de mutaciones en el gen HFE puede resultar en sobrecarga. Para evaluar el riesgo de sobrecarga de Fe fueron investigados 166 varones adultos donantes de sangre de la ciudad de Buenos Aires. Se estimó la ingesta diaria de Fe (IFe), de Fe hemínico y de Fe proveniente de harinas enriquecidas con SO4Fe. Se determinó ferritina sérica y porcentaje de saturación de transferrina (criterio de sobrecarga de Fe: ferritina sérica > 300 ng/ml y saturación de transferrina ≥ 50%). Las mutaciones C282Y, H63D y S65C fueron investigadas en sangre mediante PCR-RFLP. Todos los participantes cubrieron ampliamente el requerimiento estimado promedio de Fe (6 mg Fe/día) y 3.0% superó el máximo tolerable (45 mg Fe/día). El Fe hemínico correspondió al 9.4% de la IFe y el de harinas enriquecidas al 47.7%. Se observó una asociación entre el aumento de IFe y el de ferritina sérica (p = 0.0472), y el 2.3% de los donantes presentaron ferritina sérica > 300 ng/ml y saturación de transferrina ≥ 50%. El 29.3% de los donantes eran portadores de los genotipos H63D, S65C o C282Y, asociados a hemocromatosis hereditaria, y tenían valores de saturación de transferrina significativamente mayores a los de los donantes wild type (p = 0.0167). Si bien la incidencia clínica de hemocromatosis hereditaria fue baja en el grupo estudiado (1.2%), el consumo excesivo de Fe plantea un riesgo potencial para la salud de individuos que ignoran sus antecedentes familiares de sobrecarga de Fe.


Excess iron (Fe) intake in subjects carrying certain mutations in the HFE gene may result in Fe overload. To estimate risk of Fe overload, 166 male blood donors (19-65 years) from Buenos Aires city were investigated. Daily Fe intake (FeI), hem Fe intake, and Fe intake from SO4Fe enriched flours were estimated (SARA Computer Program and Food Composition Table, USDA). Serum ferritin and transferrin saturation were determined; criteria for Fe overload was serum ferritin > 300 ng/ml and transferrin saturation ≥ 50%. HFE genotypes C282Y, H63D and S65C were analyzed by PCR-RFLP in blood samples. No participant presented FeI lower than the estimated average requirement (6 mg Fe/day) and 3.0% was over the upper level (45 mg Fe/day). Hem Fe and Fe from flour enrichment were 9.4% and 47.7% of daily Fe intake, respectively. A significant association was observed between the increase in serum ferritin (ng/ml) and the increase in FeI (p = 0.0472); 2.3% of the donors presented serum ferritin > 300 ng/ml and transferrin saturation ≥ 50%. Genotypes associated with hereditary hemochromatosis (H63D, S65C and C282Y) were found in 29.3% of the donors. The percentage of transferrin saturation was higher in subjects carrying mutation than in wild type subjects (p = 0.0167). Although penetrance of hereditary hemochromatosis in the studied group was only 1.2%, an excessive Fe intake could enhance adverse effects in individuals unaware of any family history of Fe overload.


Humans , Male , Adult , Blood Donors/statistics & numerical data , Iron, Dietary/administration & dosage , Ferritins/blood , Hemochromatosis Protein/genetics , Hemochromatosis/genetics , Hemochromatosis/chemically induced , Polymorphism, Restriction Fragment Length , Transferrin/analysis , Genotype , Iron/blood , Mutation
7.
J Biol Chem ; 291(38): 19858-72, 2016 09 16.
Article En | MEDLINE | ID: mdl-27445333

Iron transfer across the basolateral membrane of an enterocyte into the circulation is the rate-limiting step in iron absorption and is regulated by various pathophysiological factors. Ferroportin (FPN), the only known mammalian iron exporter, transports iron from the basolateral surface of enterocytes, macrophages, and hepatocytes into the blood. Patients with genetic mutations in FPN or repeated blood transfusion develop hemochromatosis. In this study, non-mutagenic ribosomal inactivation was assessed as an etiological factor of FPN-associated hemochromatosis in enterocytes. Non-mutagenic chemical ribosomal inactivation disrupted iron homeostasis by regulating expression of the iron exporter FPN-1, leading to intracellular accumulation in enterocytes. Mechanistically, a xenobiotic insult stimulated the intracellular sentinel p38 MAPK signaling pathway, which was positively involved in FPN-1 suppression by ribosomal dysfunction. Moreover, ribosomal inactivation-induced iron accumulation in Caenorhabditis elegans as a simplified in vivo model for gut nutrition uptake was dependent on SEK-1, a p38 kinase activator, leading to suppression of FPN-1.1 expression and iron accumulation. In terms of gene regulation, ribosomal stress-activated p38 signaling down-regulated NRF2 and NF-κB, both of which were positive transcriptional regulators of FPN-1 transcription. This study provides molecular evidence for the modulation of iron bioavailability by ribosomal dysfunction as a potent etiological factor of non-mutagenic environmental hemochromatosis in the gut-to-blood axis.


Caenorhabditis elegans/metabolism , Cation Transport Proteins/metabolism , Hemochromatosis/metabolism , Iron/metabolism , MAP Kinase Signaling System , Ribosomes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cation Transport Proteins/genetics , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Hemochromatosis/chemically induced , Hemochromatosis/genetics , Hep G2 Cells , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Ribosomes/genetics , U937 Cells , Xenobiotics/adverse effects , Xenobiotics/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics
8.
Alcohol Clin Exp Res ; 37(10): 1619-31, 2013 Oct.
Article En | MEDLINE | ID: mdl-23742171

BACKGROUND: Combined iron overload and alcohol may promote synergistic chronic liver injury and toxicity. The role of specific dietary fats in influencing the development of co-toxic alcoholic liver disease needs further evaluation and is investigated in this study. METHODS: Wild-type (WT) and the iron-loaded Hfe-null (Hfe(-/-) ) mice were fed chow (CC), a AIN-93G standard control (SC), or a corn oil-modified, AIN-93G-based (CO) diet with or without the addition of 20% ethanol (EtOH) in the drinking water for 8 weeks and assessed for liver injury. RESULTS: WT mice on CC, SC, and CO diets had no liver injury, although mild steatosis developed in the SC and CO groups. The addition of EtOH resulted in mild steatohepatitis in WT mice fed SC but not those on a CO diet. EtOH administration in Hfe(-/-) animals on the CC and SC diets caused marked oxidative stress, inflammatory activity, and subsinusoidal and portal-portal tract linkage fibrosis with significant up-regulation of genes involved in cellular stress signaling and fibrogenic pathways. These effects were abrogated in the CO-fed mice, despite elevated serum EtOH levels and hepatic iron concentrations, reduced hepatic glutathione and mitochondrial superoxide dismutase activities. Feeding with the CO diet led to increased hepatic glutathione peroxidase and catalase activities and attenuated alcohol-induced hepatic steatosis in the Hfe(-/-) animals. Iron and EtOH feeding markedly reduced p-STAT3 and p-AMPK protein levels, but this effect was significantly attenuated when a CO diet was consumed. CONCLUSIONS: A CO-based diet is protective against combined EtOH- and iron-induced liver toxicity, likely via attenuation of hepatic steatosis and oxidative stress and may have a role in the prevention of fibrosis development in chronic liver disease.


Corn Oil/administration & dosage , Dietary Fats/administration & dosage , Disease Models, Animal , Ethanol/toxicity , Hemochromatosis/diet therapy , Iron/toxicity , Animals , Hemochromatosis/chemically induced , Hemochromatosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation
9.
Kidney Int ; 84(4): 756-66, 2013 Oct.
Article En | MEDLINE | ID: mdl-23615502

Hepcidin, the key regulatory hormone of iron homeostasis, and iron carriers such as transferrin receptor1 (TFR1), divalent metal transporter1 (DMT1), and ferroportin (FPN) are expressed in kidney. Whether hepcidin plays an intrinsic role in the regulation of renal iron transport is unknown. Here, we analyzed the renal handling of iron in hemochromatosis Hepc(-/-) and Hjv(-/-) mouse models, as well as in phenylhydrazine (PHZ)-treated mice. We found a marked medullary iron deposition in the kidneys of Hepc(-/-) mice, and iron leak in the urine. The kidneys of Hepc(-/-) mice exhibited a concomitant decrease in TFR1 and increase in ferritin and FPN expression. Increased FPN abundance was restricted to the thick ascending limb (TAL). DMT1 protein remained unaffected despite a significant decrease of its mRNA level, suggesting that DMT1 protein is stabilized in the absence of hepcidin. Treatment of kidney sections from Hepc(-/-) mice with hepcidin decreased DMT1 protein, an effect confirmed in renal cell lines where hepcidin markedly decreased (55)Fe transport. In the kidneys of Hjv(-/-) mice exhibiting low hepcidin expression, the iron overload was similar to that in the kidneys of Hepc(-/-) mice. However, in PHZ mice, iron accumulation resulting from hemoglobin leak was detected in the proximal tubule. Thus, kidneys exhibit a tissue-specific handling of iron that depends on the extra iron source. Hepcidin may control the expression of iron transporters to prevent renal iron overload.


Hemochromatosis/metabolism , Hepcidins/metabolism , Homeostasis/physiology , Iron/metabolism , Kidney Tubules, Distal/metabolism , Animals , Cation Transport Proteins/metabolism , Cell Line , Cells, Cultured , Disease Models, Animal , GPI-Linked Proteins , Hemochromatosis/chemically induced , Hemochromatosis/genetics , Hemochromatosis Protein , Hepcidins/deficiency , Hepcidins/genetics , In Vitro Techniques , Kidney Tubules, Distal/pathology , Loop of Henle/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Opossums , Phenylhydrazines/adverse effects , Receptors, Transferrin/metabolism
10.
J Lipid Res ; 54(5): 1484-92, 2013 May.
Article En | MEDLINE | ID: mdl-23471031

Hereditary hemochromatosis (HH) is characterized by accumulation of iron, oxidative stress, inflammation, and fibrogenesis in liver tissue. In this setting, research on the protection afforded by intracellular antioxidants is of clinical relevance. Paraoxonase-1 (PON1) is an enzyme that degrades lipid peroxides. This study investigates the alterations in serum PON1 status, PON1 gene polymorphisms, and PON1 hepatic expression in patients with HH. We performed a case-control study in 77 patients with HH (80.5% men, 22-70 years of age) and 408 healthy individuals (43.1% men, 26-74 years of age). Serum PON1 activities against different substrates and PON1192 and PON155 polymorphisms were analyzed. PON1 protein expression was investigated in 20 liver biopsies. HH patients had significantly lower serum PON1 activity, which was inversely correlated with ferritin (marker of iron stores) and serum 8-isoprostane concentrations (index of oxidative stress). PON1 protein expression in liver tissue was higher in patients and showed stronger staining in hepatocytes surrounding the areas of inflammation. Our study provides preliminary evidence that PON1 may play a role in protecting against iron-induced oxidative stress in hereditary hemochromatosis.


Aryldialkylphosphatase/blood , Hemochromatosis/genetics , Liver/enzymology , Oxidative Stress , Adult , Aged , Aryldialkylphosphatase/genetics , Biopsy , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Female , Ferritins/metabolism , Gene Expression Regulation , Hemochromatosis/blood , Hemochromatosis/chemically induced , Humans , Iron/toxicity , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Middle Aged
11.
J Biomed Sci ; 19: 83, 2012 Sep 21.
Article En | MEDLINE | ID: mdl-22998440

BACKGROUND: Iron homeostasis is chiefly regulated by hepcidin whose expression is tightly controlled by inflammation, iron stores, and hypoxia. Hemojuvelin (HJV) is a bone morphogenetic protein co-receptor that has been identified as a main upstream regulator of hepcidin expression; HJV mutations are associated with a severe form of iron overload (Juvenile haemochromatosis). Currently however, there is no information on how HJV is regulated by inflammation. METHODS: To study the regulation of Hjv expression by inflammation and whether Hfe has a role in that regulation, control and LPS-injected wild type and Hfe KO mice were used. Moreover, human hepatoma cells (HuH7) were used to study the effect of IL-6 and TNF-α on HJV mRNA expression. RESULTS: Here we show that LPS repressed hepatic Hjv and BMPs, while it induced hepcidin 1 expression in wild-type and Hfe KO mice with no effect on hepatic pSMAD 1, 5, 8 protein levels. In addition, exogenous TNF-α (20 ng/mL) decreased HJV mRNA and protein expression to 40% of control with no effect on hepcidin mRNA expression in 24 hours. On the other hand, IL-6 induced hepcidin mRNA and protein expression with no effect on HJV mRNA expression levels. Moreover, using the HJV promoter-luciferase reporter fusion construct (HJVP1.2-luc), we showed that the basal luciferase activity of HJVP1.2-luc was inhibited by 33% following TNF-α treatment of HuH7 transfected cells suggesting that the TNF-α down-regulation is exerted at the transcriptional level. Additionally, mutation of a canonical TNF- alpha responsive element (TNFRE) within HJVP1.2-luc abolished TNF-α response suggesting that this TNFRE is functional. CONCLUSIONS: From these results, we conclude that TNF-α suppresses HJV transcription possibly via a novel TNFRE within the HJV promoter. In addition, the results suggest that the proposed link between inflammation and BMP-SMAD signalling is independent of HJV and BMP ligands.


Hemochromatosis , Inflammation , Membrane Proteins , Tumor Necrosis Factor-alpha , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Cell Line, Tumor , GPI-Linked Proteins , Gene Expression Regulation/drug effects , Hemochromatosis/chemically induced , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hemochromatosis Protein , Hepcidins , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/metabolism , Interleukin-6/pharmacology , Iron/metabolism , Ligands , Lipopolysaccharides/toxicity , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Phosphorylation , Promoter Regions, Genetic , Response Elements/genetics , Smad Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Neth J Med ; 67(6): 234-6, 2009 Jun.
Article En | MEDLINE | ID: mdl-19749394

We present a case with sudden onset of throat pain, dysphagia and hoarseness. On endoscopic examination, supraglottic swelling and a brown covering of the mucous membranes were seen. The diagnosis of haemochromatosis was made on laryngeal biopsy. The patient admitted to long-term iron treatment for anaemia. Haemochromatosis can affect many different organs. If the larynx is involved, the airway may be endangered.


Airway Obstruction/etiology , Hemochromatosis/complications , Laryngeal Diseases/etiology , Larynx/injuries , Aged , Anemia/complications , Hemochromatosis/chemically induced , Humans , Iron Compounds/adverse effects , Male , Risk Factors
16.
Am J Hematol ; 81(10): 760-7, 2006 Oct.
Article En | MEDLINE | ID: mdl-16838333

We evaluated and treated four white adults (one man, three women) who had iron overload associated with daily ingestion of iron supplements for 7, 15, 35, and 61 years, respectively. We performed HFE mutation analysis to detect C282Y, H63D, and S65C in each patient; in two patients, HFE exons were sequenced. In two patients, direct sequencing was performed to detect coding region mutations of TFR2, HAMP, FPN1, HJV, and ALAS2. Patients 1-4 ingested 153, 547, 1,341, and 4,898 g of inorganic iron as supplements. Patient 1 had hemochromatosis, HFE C282Y homozygosity, and beta-thalassemia minor. Patient 2 had spherocytosis and no HFE coding region mutations. Patient 3 had no anemia, a normal HFE genotype, and no coding region mutations in HAMP, FPN1, HJV, or ALAS2; she was heterozygous for the TFR2 coding region mutation V583I (nt 1,747 G-->A, exon 15). Patient 4 had no anemia and no coding region mutations in HFE, TFR2, HAMP, FPN1, HJV, or ALAS2. Iron removed by phlebotomy was 32.4, 10.4, 15.2, and 4.0 g, respectively. There was a positive correlation of log(10) serum ferritin and the quantity of iron removed by phlebotomy (P = 0.0371). Estimated absorption of iron from supplements in patients 1-4 was 20.9%, 1.9%, 1.1%, and 0.08%. We conclude that the clinical phenotypes and hemochromatosis genotypes of adults who develop iron overload after ingesting iron supplements over long periods are heterogeneous. Therapeutic phlebotomy is feasible and effective, and would prevent complications of iron overload.


Dietary Supplements/adverse effects , Hemochromatosis/chemically induced , Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Iron Overload/diagnosis , Iron/adverse effects , Membrane Proteins/genetics , Adult , Aged , Drug Administration Schedule , Female , Genotype , Hemochromatosis/diagnosis , Hemochromatosis Protein , Humans , Iron/administration & dosage , Iron Overload/chemically induced , Iron Overload/genetics , Male , Mutation
17.
Nutrition ; 20(7-8): 615-9, 2004.
Article En | MEDLINE | ID: mdl-15212743

Although it generally does not improve performance, iron is often used by elite athletes. The physiologic changes induced by exercise can mimic iron deficiency and decrease hemoglobin and ferritin concentrations. Determination of serum transferrin receptor concentrations may identify true iron deficiency, which occurs particularly in young athletes. In contrast, increased iron stores in the body are a frequent finding in elite athletes who have used long-term iron supplementation. Elite runners have increased intestinal blood loss, but this usually can be compensated by enhanced absorption of dietary iron. The combination of exercise-induced hemolysis with enhanced intestinal blood loss in various endurance sports leads to severe abnormalities of routine tests, and extreme physical activity may be responsible for positive fecal occult blood determinations. Indiscriminate iron supplementation carries the risk of inducing hemochromatosis in individuals homozygous for the widespread C282Y allele of the HFE gene. This polymorphism is common and can be found in about 1% of individuals of Northern European descent; moreover, iron supplementation can modify the presentation of important underlying diseases such as celiac disease or colon carcinoma. In conclusion, iron supplements should be prescribed for athletes with iron-deficiency anemia and carefully monitored if given for prophylaxis; unless a therapeutic response occurs, investigations to establish the cause of iron deficiency should be initiated.


Iron, Dietary/administration & dosage , Sports/physiology , Adaptation, Physiological , Dietary Supplements , Exercise/physiology , Ferritins/blood , Hemochromatosis/chemically induced , Hemoglobins/analysis , Humans , Intestinal Absorption , Iron Deficiencies , Iron, Dietary/adverse effects , Iron, Dietary/pharmacokinetics , Risk Factors
18.
Z Gastroenterol ; 39(6): 453-5, 458, 2001 Jun.
Article En | MEDLINE | ID: mdl-11475000

Patients with porphyria cutanea tarda (PCT) develop hepatocellular carcinoma as a late consequence. Pre-loading of C57BL/10ScSn mice with iron greatly sensitizes them to the induction of hepatic porphyria caused by hexachlorobenzene (HCB). HCB will also cause liver tumors in experimental animals. Elevated liver iron stores are implicated in the development of some human liver cancers in connection with its known catalytic role in generation of highly reactive activated oxygen species. The aim of this study was to determine the lipid and DNA oxidative damage in iron and HCB-induced porphyric mice. C57BL/10ScSn mice received i.p. injections of dextran sulfate (control), iron (Imferon) or combined iron and HCB. 6 weeks after treatment plasma ALT levels and hepatic free iron, porphyrin, lipid peroxides and 8-hydroxyguanosine (8-OHdG) levels were analyzed. Hepatic porphyrin level was significantly (p < 0.001) increased following combined iron/HCB treatment as compared to control mice. The level of lipid peroxides increased 9-fold (p = 0.001) and 35-fold (p < 0.001) after iron and iron/HCB treatment respectively, whereas the level of 8-OHdG was increased 2.5-fold (p = 0.002) and 7.5-fold (p < 0.001) after iron and iron/HCB treatment respectively as compared to control mice. The authors conclude that iron overload in conjugation with HCB induce lipid and DNA oxidative damage in C57BL/10ScSn mice. DNA oxidative damage may be important in the early events of hepatic carcinogenesis in experimental porphyria.


DNA Damage/physiology , Hemochromatosis/pathology , Lipid Peroxidation/physiology , Liver Neoplasms, Experimental/pathology , Porphyria Cutanea Tarda/pathology , Porphyrias, Hepatic/pathology , Animals , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Hemochromatosis/chemically induced , Hexachlorobenzene/toxicity , Iron-Dextran Complex/toxicity , Liver/drug effects , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mice, Inbred C57BL , Porphyria Cutanea Tarda/chemically induced , Porphyrias, Hepatic/chemically induced
19.
Tidsskr Nor Laegeforen ; 121(4): 460-2, 2001 Feb 10.
Article Nor | MEDLINE | ID: mdl-11255863

Many persons associate fatigue and lassitude with iron deficiency and take extra iron "to be on the safe side". This is an unfortunate practice, as the early symptoms of iron deficiency anaemia and of hereditary iron overload (homozygous primary haemochromatosis) are similar. Primary haemochromatosis is considerably more prevalent than earlier believed. As many as 5 per 1,000 of the Norwegian population may have two mutated genes for haemochromatosis, while up to 15% may be carriers of a single mutated gene, and for these an extra intake of iron may be hazardous. The condition is highly underdiagnosed. In Norway at present, iron preparations of 60-100 mg are sold over the counter in pharmacies without prescription and often by self-service. However, no one should use iron tablets until iron deficiency and its cause has been ascertained. To avoid uncritical use of iron, iron preparations should be available only by doctor's prescription. Prolonged abuse of iron tablets may result in secondary haemochromatosis.


Iron/administration & dosage , Adult , Aged , Drug Prescriptions , Female , Hemochromatosis/chemically induced , Hemochromatosis/diagnosis , Humans , Iron/adverse effects , Iron/metabolism , Nonprescription Drugs , Norway , Risk Factors , Self Medication
20.
Pathol Res Pract ; 196(1): 47-54, 2000.
Article En | MEDLINE | ID: mdl-10674272

The purpose of this study was to estimate the iron concentration in the liver, spleen and brain of control rats and rats overloaded with iron and to determine the distribution of iron and of transferrin (TF). Iron was administered to Wistar rats by food supplemented with 3% carbonyl iron for 3 months, or intraperitoneally, or intraveneously as iron polymaltose for 4 months (total administered dose: 300 or 350 mg/rat, respectively). Iron concentration was estimated by atomic absorption spectrophotometry and iron- and TF-distribution histochemically and immunohistochemically, respectively. In control rats the organ with the highest iron content was the spleen, followed by the liver and brain. After iron loading the increase of iron in the liver was greater than that of the spleen; iron concentration in the brain did not change significantly. Distribution of iron in the liver was in Kupffer cells throughout the lobule and in hepatocytes at its periphery. No difference in the number of positive cells or staining intensity for TF was observed between control rats and iron overloaded animals in the liver or central nervous system (CNS); the spleen was negative for TF. Distribution of TF in the liver showed a centrilobular localisation in hepatocytes. TF reaction in the brain occurred in oligodendrocytes, vessel walls, choroid plexus epithelial cells and some neurons. In conclusion, experimental iron overload in rats leads to iron uptake mainly by reticuloendothelial (RE) cells and hepatocytes, indicating that hepatocytes are of particular importance for iron metabolism. Iron uptake by the brain was not significant, probably because the brain is protected against iron overload. Iron overload did not influence location and quantity of TF in the liver and CNS, whereas the visualisation of iron and TF did not coincide. This indicates that TF may have other functions beyond iron transport.


Central Nervous System/metabolism , Hemochromatosis/metabolism , Iron/metabolism , Liver/metabolism , Spleen/metabolism , Transferrin/metabolism , Animals , Central Nervous System/pathology , Hemochromatosis/chemically induced , Hemochromatosis/pathology , Immunoenzyme Techniques , Iron, Dietary/administration & dosage , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/pathology , Male , Rats , Rats, Wistar , Spleen/pathology
...