Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Int J Mol Sci ; 22(15)2021 Jul 22.
Article in English | MEDLINE | ID: mdl-34360586

ABSTRACT

Copper, manganese, and iron are vital elements required for the appropriate development and the general preservation of good health. Additionally, these essential metals play key roles in ensuring proper brain development and function. They also play vital roles in the central nervous system as significant cofactors for several enzymes, including the antioxidant enzyme superoxide dismutase (SOD) and other enzymes that take part in the creation and breakdown of neurotransmitters in the brain. An imbalance in the levels of these metals weakens the structural, regulatory, and catalytic roles of different enzymes, proteins, receptors, and transporters and is known to provoke the development of various neurological conditions through different mechanisms, such as via induction of oxidative stress, increased α-synuclein aggregation and fibril formation, and stimulation of microglial cells, thus resulting in inflammation and reduced production of metalloproteins. In the present review, the authors focus on neurological disorders with psychiatric signs associated with copper, iron, and manganese excess and the diagnosis and potential treatment of such disorders. In our review, we described diseases related to these metals, such as aceruloplasminaemia, neuroferritinopathy, pantothenate kinase-associated neurodegeneration (PKAN) and other very rare classical NBIA forms, manganism, attention-deficit/hyperactivity disorder (ADHD), ephedrone encephalopathy, HMNDYT1-SLC30A10 deficiency (HMNDYT1), HMNDYT2-SLC39A14 deficiency, CDG2N-SLC39A8 deficiency, hepatic encephalopathy, prion disease and "prion-like disease", amyotrophic lateral sclerosis, Huntington's disease, Friedreich's ataxia, and depression.


Subject(s)
Ceruloplasmin/deficiency , Copper/adverse effects , Iron Metabolism Disorders/pathology , Iron/adverse effects , Manganese/adverse effects , Metabolic Diseases/pathology , Neuroaxonal Dystrophies/pathology , Neurodegenerative Diseases/pathology , Humans , Iron Metabolism Disorders/chemically induced , Iron Metabolism Disorders/etiology , Manganese Poisoning/complications , Metabolic Diseases/chemically induced , Metalloproteins/metabolism , Neuroaxonal Dystrophies/chemically induced , Neurodegenerative Diseases/etiology , Oxidative Stress
2.
Acta Neuropathol ; 142(4): 729-759, 2021 10.
Article in English | MEDLINE | ID: mdl-34292399

ABSTRACT

Neuronal dysfunction due to iron accumulation in conjunction with reactive oxygen species (ROS) could represent an important, yet underappreciated, component of the epileptogenic process. However, to date, alterations in iron metabolism in the epileptogenic brain have not been addressed in detail. Iron-related neuropathology and antioxidant metabolic processes were investigated in resected brain tissue from patients with temporal lobe epilepsy and hippocampal sclerosis (TLE-HS), post-mortem brain tissue from patients who died after status epilepticus (SE) as well as brain tissue from the electrically induced SE rat model of TLE. Magnetic susceptibility of the presumed seizure-onset zone from three patients with focal epilepsy was compared during and after seizure activity. Finally, the cellular effects of iron overload were studied in vitro using an acute mouse hippocampal slice preparation and cultured human fetal astrocytes. While iron-accumulating neurons had a pyknotic morphology, astrocytes appeared to acquire iron-sequestrating capacity as indicated by prominent ferritin expression and iron retention in the hippocampus of patients with SE or TLE. Interictal to postictal comparison revealed increased magnetic susceptibility in the seizure-onset zone of epilepsy patients. Post-SE rats had consistently higher hippocampal iron levels during the acute and chronic phase (when spontaneous recurrent seizures are evident). In vitro, in acute slices that were exposed to iron, neurons readily took up iron, which was exacerbated by induced epileptiform activity. Human astrocyte cultures challenged with iron and ROS increased their antioxidant and iron-binding capacity, but simultaneously developed a pro-inflammatory phenotype upon chronic exposure. These data suggest that seizure-mediated, chronic neuronal iron uptake might play a role in neuronal dysfunction/loss in TLE-HS. On the other hand, astrocytes sequester iron, specifically in chronic epilepsy. This function might transform astrocytes into a highly resistant, pro-inflammatory phenotype potentially contributing to pro-epileptogenic inflammatory processes.


Subject(s)
Epilepsy, Temporal Lobe/complications , Hippocampus/metabolism , Iron Metabolism Disorders/etiology , Iron/metabolism , Status Epilepticus/complications , Adult , Aged , Aged, 80 and over , Animals , Astrocytes/metabolism , Astrocytes/pathology , Case-Control Studies , Cell Culture Techniques , Disease Models, Animal , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Female , Humans , Iron Metabolism Disorders/pathology , Male , Middle Aged , Oxidative Stress/physiology , Rats , Status Epilepticus/metabolism , Status Epilepticus/pathology
3.
Article in English | MEDLINE | ID: mdl-33373681

ABSTRACT

Exacerbation of cognitive, motor and nonmotor symptoms have been described in critically ill COVID-19 patients, indicating that, like prior pandemics, neurodegenerative sequelae may mark the aftermath of this viral infection. Moreover, SARS-CoV-2, the causative agent of COVID-19 disease, was associated with hyperferritinemia and unfavorable prognosis in older individuals, suggesting virus-induced ferrosenescence. We have previously defined ferrosenescence as an iron-associated disruption of both the human genome and its repair mechanisms, leading to premature cellular senescence and neurodegeneration. As viruses replicate more efficiently in iron-rich senescent cells, they may have developed the ability to induce this phenotype in host tissues, predisposing to both immune dysfunction and neurodegenerative disorders. In this mini-review, we summarize what is known about the SARS-CoV-2-induced cellular senescence and iron dysmetabolism. We also take a closer look at immunotherapy with natural killer cells, angiotensin II receptor blockers ("sartans"), iron chelators and dipeptidyl peptidase 4 inhibitors ("gliptins") as adjunct treatments for both COVID-19 and its neurodegenerative complications.


Subject(s)
COVID-19/complications , COVID-19/physiopathology , Cellular Senescence , Iron Metabolism Disorders/etiology , Iron Metabolism Disorders/physiopathology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/physiopathology , Humans , Iron/metabolism , Iron/physiology
4.
Pediatrics ; 146(1)2020 07.
Article in English | MEDLINE | ID: mdl-32503934

ABSTRACT

BACKGROUND: Teenagers aged 16 to 18 are at increased risk for iron deficiency, exacerbated by losses with whole blood (WB) or double red blood cell (2RBC) donations. Required 56-day (WB) or 112-day (2RBC) interdonation intervals (IDIs) are too short for many to replace lost iron without supplements. METHODS: Teenagers donating WB or 2RBCs at Vitalant, a national blood provider, had serum ferritin measured at their first and immediately subsequent successful donation from December 2016 to 2018. We modeled postindex log-ferritin as a function of IDI to estimate the shortest intervals that corresponded with 50% to 95% prevalence of adequate donor iron stores (ferritin ≥20 ng/mL female donors, ≥30 ng/mL male donors) at the subsequent donation. RESULTS: Among 30 806 teenagers, 11.4% of female and 9.7% of male donors had inadequate iron stores at index donation. Overall, 92.6% had follow-up ferritin values within 13 months. Approximately 12 months after WB index donations, >60% of female and >80% of male donors had adequate iron stores (>50% and >70% after 2RBC donations). Follow-up-donation iron stores were highly dependent on index ferritin. Less than half of WB donors with low ferritin at index achieved adequate stores within 12 months. Achieving a ≥90% prevalence of adequate ferritin at 12 months required index values >50 ng/mL. CONCLUSIONS: These findings suggest that postdonation low-dose iron supplements should be strongly encouraged in teenagers with borderline or low iron stores to permit donation without increased risk for symptoms of mild iron depletion. Increasing the minimum recommended IDI to allow time for replacing donation-related iron losses may be desirable for teenagers.


Subject(s)
Blood Donors/statistics & numerical data , Ferritins/blood , Iron/metabolism , Adolescent , Cohort Studies , Dietary Supplements , Female , Humans , Iron Compounds/administration & dosage , Iron Deficiencies , Iron Metabolism Disorders/etiology , Iron Metabolism Disorders/prevention & control , Male , Time Factors
5.
Medicina (Kaunas) ; 55(7)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269687

ABSTRACT

Background and objectives: The etiology of anemia associated with heart failure is not fully understood, but there are data suggesting the involvement of multiple mechanisms, including various drug therapies used in patients with heart failure. Our primary objective was to evaluate the impact of beta blockers, angiotensin-converting enzyme inhibitors, and calcium-channel blockers on iron metabolism in patients with heart failure. Materials and Methods: This was a prospective observational study that included patients diagnosed with heart failure and iron deficiency (defined by ferritin <100 µg/L, or 100-300 µg/L with transferrin saturation <20%). Patients with anemia secondary to a known cause were excluded. Results: We found a statistically significant correlation between beta-blocker treatment and ferritin values (p = 0.02). Iron, hemoglobin, and hematocrit levels were significantly lower in the patients using calcium-channel blockers than those who were not. We also found a statistically significant indirect correlation (p = 0.04) between the use of angiotensin-converting enzyme inhibitors and hematocrit levels. Conclusion: The contribution of our study arises from the additional data regarding the drug-induced etiology of iron deficiency. Practitioners should be aware of the potential impact of therapeutic recommendations and this should imply a close monitoring of the biochemical parameters of iron deficiency in this category of patients.


Subject(s)
Anemia/etiology , Heart Failure/drug therapy , Iron Metabolism Disorders/etiology , Adrenergic beta-Antagonists/adverse effects , Adrenergic beta-Antagonists/therapeutic use , Aged , Anemia/blood , Anemia/complications , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/therapeutic use , Female , Heart Failure/blood , Humans , Iron/analysis , Iron/blood , Iron Metabolism Disorders/blood , Male , Middle Aged
6.
Nutrients ; 11(5)2019 May 20.
Article in English | MEDLINE | ID: mdl-31137583

ABSTRACT

BACKGROUND: Since haemodialysis is a lifesaving therapy, adequate control measures are necessary to evaluate its adequacy and to constantly adjust the dose to reduce hospitalisation and prolong patient survival. Malnutrition is common in haemodialysis patients and closely related to morbidity and mortality. Patients undergoing haemodialysis have a high prevalence of protein-energy malnutrition and inflammation, along with abnormal iron status. The haemodialysis dose delivered is an important predictor of patient outcome. AIM: To evaluate through haemodialysis adequacy, which parameter(s), if any, better predict Kt/V, among those used to assess nutritional status, inflammation response, and iron status. METHODS: We retrospectively studied 78 patients undergoing haemodialysis due to end-stage renal disease. As parameters of nutritional status, geriatric nutritional risk index (GNRI), transferrin levels, lymphocyte count, and albumin concentration were analysed. As signs of inflammation, C reactive protein (CRP) levels and ferritin concentrations were studied as well. Iron status was evaluated by both transferrin and ferritin levels, as well as by haemoglobin (Hb) concentration. RESULTS: The core finding of our retrospective study is that transferrin levels predict the adequacy of haemodialysis expressed as Kt/V; the latter is the only predictor (P = 0.001) when adjusting for CRP concentrations, a solid marker of inflammation, and for ferritin levels considered an iron-storage protein, but also a parameter of inflammatory response. DISCUSSION AND CONCLUSION: In keeping with the results of this study, we underline that the use of transferrin levels to assess haemodialysis quality combine into a single test the evaluation of the three most important factors of protein-energy wasting.


Subject(s)
Inflammation/blood , Iron Metabolism Disorders/blood , Kidney Failure, Chronic/therapy , Protein-Energy Malnutrition/blood , Renal Dialysis , Transferrin/metabolism , Aged , Biomarkers/blood , Female , Geriatric Assessment , Humans , Inflammation/diagnosis , Inflammation/etiology , Inflammation Mediators/blood , Iron/blood , Iron Deficiencies , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/etiology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Male , Middle Aged , Nutritional Status , Predictive Value of Tests , Protein-Energy Malnutrition/diagnosis , Protein-Energy Malnutrition/etiology , Renal Dialysis/adverse effects , Retrospective Studies , Risk Factors , Treatment Outcome
8.
Blood ; 133(1): 51-58, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30401707

ABSTRACT

ß-Thalassemia (BT) is an inherited genetic disorder that is characterized by ineffective erythropoiesis (IE), leading to anemia and abnormal iron metabolism. IE is an abnormal expansion of the number of erythroid progenitor cells with unproductive synthesis of enucleated erythrocytes, leading to anemia and hypoxia. Anemic patients affected by BT suffer from iron overload, even in the absence of chronic blood transfusion, suggesting the presence of ≥1 erythroid factor with the ability to modulate iron metabolism and dietary iron absorption. Recent studies suggest that decreased erythroid cell differentiation and survival also contribute to IE, aggravating the anemia in BT. Furthermore, hypoxia can also affect and increase iron absorption. Understanding the relationship between iron metabolism and IE could provide important insights into the BT condition and help to develop novel treatments. In fact, genetic or pharmacological manipulations of iron metabolism or erythroid cell differentiation and survival have been shown to improve IE, iron overload, and anemia in animal models of BT. Based on those findings, new therapeutic approaches and drugs have been proposed; clinical trials are underway that have the potential to improve erythrocyte production, as well as to reduce the iron overload and organ toxicity in BT and in other disorders characterized by IE.


Subject(s)
Erythropoiesis , Iron Metabolism Disorders/etiology , Iron Metabolism Disorders/pathology , Iron/metabolism , beta-Thalassemia/complications , Animals , Humans , beta-Thalassemia/pathology
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 967-973, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29317336

ABSTRACT

Understandings of the disturbed iron metabolism in Parkinson's disease (PD) are largely from the perspectives of neurons. Neurodegenerative processes in PD trigger universal and conserved astroglial dysfunction and microglial activation. In this review, we start with astroglia and microglia in PD with an emphasis on their roles in spreading α-synuclein pathology, and then focus on their contributions in iron metabolism under normal conditions and the diseased state of PD. Elevated iron in the brain regions affects glial features, meanwhile, glial effects on neuronal iron metabolism are largely dependent on their releasing factors. These advances might be valuable for better understanding and modulating iron metabolism disturbance in PD.


Subject(s)
Astrocytes/physiology , Iron Metabolism Disorders/etiology , Iron/metabolism , Microglia/physiology , Parkinson Disease/complications , Parkinson Disease/metabolism , Animals , Astrocytes/pathology , Humans , Iron Metabolism Disorders/metabolism , Microglia/pathology
10.
Liver Int ; 38(1): 164-173, 2018 01.
Article in English | MEDLINE | ID: mdl-28679028

ABSTRACT

BACKGROUND AND AIMS: Elevated serum ferritin is common in NAFLD, and is associated with more advanced disease and increased mortality. Hyperferritinaemia in NAFLD is often attributed to inflammation, while in other conditions ferritin closely reflects body iron stores. The aim of this study was to clarify the underlying cause of hyperferritinaemia in NAFLD. METHODS: Ferritin levels were examined with markers of iron status, inflammation and liver injury across the clinical spectrum of NAFLD using blood, tissue and magnetic resonance (MR) imaging. A separate larger group of NAFLD patients with hepatic iron staining and quantification were used for validation. RESULTS: Serum ferritin correlated closely with the iron regulatory hormone hepcidin, and liver iron levels determined by MR. Furthermore, ferritin levels reflected lower serum adiponectin, a marker of insulin resistance, and liver fat, but not cytokine or CRP levels. Ferritin levels differed according to fibrosis stage, increasing from early to moderate disease, and declining in cirrhosis. A similar pattern was found in the validation cohort of NAFLD patients, where ferritin levels were highest in those with macrophage iron deposition. Multivariate analysis revealed liver iron and hepcidin levels as the major determinants of serum ferritin. CONCLUSIONS: While hyperferritinaemia is associated with markers of liver injury and insulin resistance, serum hepcidin and hepatic iron are the strongest predictors of ferritin levels. These findings highlight the role of disordered iron homeostasis in the pathogenesis of NAFLD, suggesting that therapies aimed at correcting iron metabolism may be beneficial.


Subject(s)
Ferritins/blood , Iron Metabolism Disorders/blood , Iron/analysis , Liver/chemistry , Non-alcoholic Fatty Liver Disease/blood , Adiponectin/blood , Adult , Aged , Biomarkers/blood , Female , Hepcidins/analysis , Humans , Insulin Resistance , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/etiology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/diagnosis , Prospective Studies , Up-Regulation
11.
Mutat Res Rev Mutat Res ; 774: 25-32, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29173496

ABSTRACT

Iron, as an essential nutrient, and the DNA, as the carrier of genetic information which is physically compacted into chromosomes, are both needed for normal life and well-being. Therefore, it is not surprising that close interactions exist between iron and the genome. On the one hand, iron, especially when present in excess, may alter genome stability through oxidative stress, and may favor cell cycle abnormalities and the development of malignant diseases. The genome also receives a feedback signal from the systemic iron status, leading to promotion of expression of genes that regulate iron metabolism. Conversely, on the other hand, DNA mutations may cause genetic iron-related diseases such as hemochromatosis, archetype of iron-overload diseases, or refractory iron deficiency anemia (IRIDA).


Subject(s)
Genomic Instability , Iron Metabolism Disorders/etiology , Iron Overload/etiology , Iron/metabolism , Mutation , Humans , Iron Metabolism Disorders/metabolism , Iron Overload/metabolism
12.
Med Clin (Barc) ; 148(5): 218-224, 2017 Mar 03.
Article in English, Spanish | MEDLINE | ID: mdl-28073521

ABSTRACT

Hepcidin is the main regulator of iron metabolism and a pathogenic factor in iron disorders. Hepcidin deficiency causes iron overload, whereas hepcidin excess causes or contributes to the development of iron-restricted anaemia in chronic inflammatory diseases. We know the mechanisms involved in the synthesis of hepcidin and, under physiological conditions, there is a balance between activating signals and inhibitory signals that regulate its synthesis. The former include those related to plasmatic iron level and also those related to chronic inflammatory diseases. The most important inhibitory signals are related to active erythropoiesis and to matriptase-2. Knowing how hepcidin is synthesised has helped design new pharmacological treatments whose main target is the hepcidin. In the near future, there will be effective treatments aimed at correcting the defect of many of these iron metabolism disorders.


Subject(s)
Hepcidins/metabolism , Iron Metabolism Disorders/metabolism , Iron/metabolism , Biomarkers/metabolism , Cation Transport Proteins/metabolism , Homeostasis , Humans , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/etiology , Iron Metabolism Disorders/therapy
13.
Am J Hematol ; 92(4): 338-343, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28052375

ABSTRACT

Although hyperferritinemia may be reflective of elevated total body iron stores, there are conditions in which ferritin levels are disproportionately elevated relative to iron status. Autosomal dominant forms of hyperferritinemia due to mutations in the L-ferritin IRE or in A helix of L-ferritin gene have been described, however cases of isolated hyperferritinemia still remain unsolved. We describe 12 Italian subjects with unexplained isolated hyperferritinemia (UIH). Four probands have affected siblings, but no affected parents or offspring. Sequencing analyses did not identify casual mutations in ferritin gene or IRE regions. These patients had normal levels of intracellular ferritin protein and mRNA in peripheral blood cells excluding pathological ferritin production at transcriptional and post-transcriptional level. In contrast with individuals with benign hyperferritinemia caused by mutations affecting the ferritin A helix, low rather than high glycosylation of serum ferritin was observed in our UIH subjects compared with controls. These findings suggest that subjects with UIH have a previously undescribed form of hyperferritinemia possibly attributable to increased cellular ferritin secretion and/or decreased serum ferritin clearance. The cause remains to be defined and we can only speculate the existence of mutations in gene/s not directly implicated in iron metabolism that could affect ferritin turnover including ferritin secretion.


Subject(s)
Ferritins/blood , Iron Metabolism Disorders/etiology , Adult , Case-Control Studies , Female , Glycosylation , Humans , Iron Overload , Italy , Male , Middle Aged , Pedigree , RNA, Messenger/blood , Siblings , Young Adult
16.
Medicine (Baltimore) ; 95(14): e3150, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27057839

ABSTRACT

Iron is required for most forms of organisms, and it is the most essential element for the functions of many iron-containing proteins involved in oxygen transport, cellular respiration, DNA replication, and so on. Disorders of iron metabolism are associated with diverse diseases, including anemias (e.g., iron-deficiency anemia and anemia of chronic diseases) and iron overload diseases, such as hereditary hemochromatosis and ß-thalassemia. Hepcidin (encoded by Hamp gene) is a peptide hormone synthesized by hepatocytes, and it plays an important role in regulating the systematic iron homeostasis. As the systemic iron regulator, hepcidin, not only controls dietary iron absorption and iron egress out of iron storage cells, but also induces iron redistribution in various organs. Deregulated hepcidin is often seen in a variety of iron-related diseases including anemias and iron overload disorders. In the case of iron overload disorders (e.g., hereditary hemochromatosis and ß-thalassemia), hepatic hepcidin concentration is significantly reduced.Since hepcidin deregulation is responsible for iron disorder-associated diseases, the purpose of this review is to summarize the recent findings on therapeutics targeting hepcidin.Continuous efforts have been made to search for hepcidin mimics and chemical compounds that could be used to increase hepcidin level. Here, a literature search was conducted in PubMed, and research papers relevant to hepcidin regulation or hepcidin-centered therapeutic work were reviewed. On the basis of literature search, we recapitulated recent findings on therapeutic studies targeting hepcidin, including agonists and antagonists to modulate hepcidin expression or its downstream signaling. We also discussed the molecular mechanisms by which hepcidin level and iron metabolism are modulated.Elevating hepcidin concentration is an optimal strategy to ameliorate iron overload diseases, and also to relieve ß-thalassemia phenotypes by improving ineffective erythropoiesis. Relative to the current conventional therapies, such as phlebotomy and blood transfusion, therapeutics targeting hepcidin would open a new avenue for treatment of iron-related diseases.


Subject(s)
Hepcidins/drug effects , Iron Metabolism Disorders/drug therapy , Hepcidins/physiology , Homeostasis , Humans , Iron/physiology , Iron Metabolism Disorders/etiology
17.
Bone Marrow Transplant ; 51(1): 89-95, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26367238

ABSTRACT

Elevated serum ferritin contributes to treatment-related morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). The multicenter DE02 trial assessed the safety, efficacy and impact of deferasirox on iron homeostasis after allogeneic HSCT. Deferasirox was administered at a starting dose of 10 mg/kg per day to 76 recipients of allogeneic HSCT, with subsequent dose adjustments based on efficacy and safety. Deferasirox was initiated at a median of 168 days after HSCT, with 84% of patients still on immunosuppression. Baseline serum ferritin declined from 2045 to 957 ng/mL. Deferasirox induced a negative iron balance in 84% of patients. Hemoglobin increased in the first 3 months, and trough serum cyclosporine levels were stable. Median exposure was 330 days, with a median compliance rate of >80%. The most common investigator-reported drug-related adverse events (AEs) were increased blood creatinine (26.5%), nausea (9.0%) and abdominal discomfort (8.3%). Fifty-four (71.1%) patients experienced drug-related AEs, which occasionally resulted in discontinuation (gastrointestinal (n=6), skin (n=3), elevated transaminases (n=1) and creatinine (n=1)). The incidence of AEs appeared to be dose related, with 7.5 mg/kg per day being the best-tolerated dose. Low-dose deferasirox is an effective chelation therapy after allogeneic HSCT, with a manageable safety profile, even in patients receiving cyclosporine.


Subject(s)
Benzoates/administration & dosage , Benzoates/pharmacokinetics , Ferritins/blood , Hematopoietic Stem Cell Transplantation , Iron Metabolism Disorders , Iron/blood , Triazoles/administration & dosage , Triazoles/pharmacokinetics , Adult , Aged , Allografts , Benzoates/adverse effects , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Cyclosporine/blood , Deferasirox , Female , Humans , Iron Metabolism Disorders/blood , Iron Metabolism Disorders/drug therapy , Iron Metabolism Disorders/etiology , Male , Middle Aged , Prospective Studies , Triazoles/adverse effects
20.
Clin Rheumatol ; 34(11): 1989-92, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26233722

ABSTRACT

There are four medical conditions characterized by high levels of ferritin, the macrophage activation syndrome (MAS), adult onset Still' s disease (AOSD), catastrophic antiphospholipid syndrome (CAPS), and septic shock, that share similar clinical and laboratory features, suggesting a common pathogenic mechanism. This common syndrome entity is termed "the hyperferritinemic syndrome." Here, we describe two different cases of hyperferritinemic syndrome triggered by Chikungunya fever virus infection: a 21-year-old female with SLE and a 32-year-old male patient who developed AOSD after the coinfection of dengue and Chikungunya viruses.


Subject(s)
Antiphospholipid Syndrome/etiology , Cataract/congenital , Chikungunya Fever/complications , Dengue/complications , Iron Metabolism Disorders/congenital , Still's Disease, Adult-Onset/etiology , Adult , Antiphospholipid Syndrome/diagnosis , Cataract/diagnosis , Cataract/etiology , Female , Ferritins/blood , Fever , Humans , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/etiology , Lupus Erythematosus, Systemic , Male , Still's Disease, Adult-Onset/diagnosis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...