Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Sci Rep ; 12(1): 1967, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121800

ABSTRACT

Neutrophil dysfunction contributes to a high susceptibility to severe bacterial infection which is a leading cause of morbidity and mortality in ß-thalassaemia/HbE, especially in splenectomised patients. This study demonstrated another abnormality of neutrophil function, namely neutrophil extracellular trap (NET) formation in splenectomised and non-splenectomised ß-thalassaemia/HbE patients who had iron overload. A classification system of morphological NET formation using confocal microscopy was developed, and samples were categorized into early and late phases which were subdivided into web-like and non-web structures. At baseline, neutrophils from non-splenectomised patients (58 ± 4%) and splenectomised patients (65 ± 3%) had higher early phase NETs than those from normal subjects (33 ± 1%). As a mimic of iron overload and infection, haemin/PMA/LPS treatment led to a significant reduction of early NETs and an increase of late NETs in neutrophils from normal and non-splenectomised patients. Interestingly, neutrophils from splenectomised patients had impaired development of late NETs. This suggests that during infection bacteria might not be trapped and may spread from the site of infection resulting in higher susceptibility to severe bacterial infection in splenectomised patients.


Subject(s)
Bacterial Infections/genetics , Extracellular Traps/genetics , Neutrophils/microbiology , beta-Thalassemia/genetics , Bacterial Infections/microbiology , Bacterial Infections/pathology , Extracellular Traps/microbiology , Humans , Immunity, Innate/genetics , Iron/metabolism , Iron Overload/genetics , Iron Overload/microbiology , Iron Overload/pathology , Neutrophils/pathology , Splenectomy , beta-Thalassemia/microbiology , beta-Thalassemia/pathology
2.
J Mycol Med ; 30(1): 100905, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31706700

ABSTRACT

INTRODUCTION: Iron chelator has previously demonstrated fungicidal effects. This study aimed to investigate the antifungal activity of the iron chelators deferoxamine (DFO) and deferasirox (DSX) against Cryptococcus. MATERIALS AND METHODS: Cryptococcus neoformans and Cryptococcus gattii were used to determine the minimal inhibitory concentrations (MICs) of DFO and DSX, and the fractional inhibitory concentration index (FICI) of DFO and DSX when combined with amphotericin B (AMB). Expression of cryptococcal CFT1, CFT2, and CIR1 genes was determined using real-time polymerase chain reaction (PCR). RESULTS: Neither DFO nor DSX alone showed antifungal activity against Cryptococcus strains. When combined with AMB, the MICs of DFO and DSX decreased from>200µg/mL to 6.25 or 12.5µg/mL. The MIC of AMB decreased one-fold dilution in most strains when combined with iron chelators. The FICI of DFO+AMB and DSX+AMB was 0.5 and 1, respectively. C. neoformans showed significant growth retardation when incubated with a combination of sub-MIC concentrations of AMB and DFO; whereas, C. gattii demonstrated lesser growth retardation in DFO+AMB. No cryptococcal growth retardation was observed when DSX was combined with AMB. When C. neoformans was grown in DFO, the CFT1, CFT2, and CIR1 proteins were expressed 1.7, 2.0, and 0.9 times, respectively. When C. neoformans was grown in DSX, the CFT1, CFT2, and CIR1 genes were expressed 0.5, 0.6, and 0.3 times, respectively. CONCLUSION: Synergistic antifungal activity of combination DFO and AMB was observed in Cryptococcus. Relatively increased CFT1 and CFT2 expression may be associated with the effect of DFO that inhibits the growth of fungi.


Subject(s)
Cryptococcus/drug effects , Cryptococcus/growth & development , Cryptococcus/genetics , Iron Chelating Agents/pharmacology , Iron/metabolism , Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus/metabolism , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/genetics , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/metabolism , Deferasirox/pharmacology , Deferoxamine/pharmacology , Drug Synergism , Fungal Capsules/drug effects , Fungal Capsules/genetics , Fungal Capsules/metabolism , Gene Expression Regulation, Fungal/drug effects , Humans , Invasive Fungal Infections/complications , Invasive Fungal Infections/drug therapy , Invasive Fungal Infections/microbiology , Iron Overload/complications , Iron Overload/drug therapy , Iron Overload/microbiology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Microbial Sensitivity Tests , Virulence Factors/genetics , Virulence Factors/metabolism
3.
Pediatr Infect Dis J ; 39(4): e40-e41, 2020 04.
Article in English | MEDLINE | ID: mdl-31815828

ABSTRACT

Escherichia coli is an extremely unusual cause of monomicrobial necrotizing fasciitis of the extremities in children. We report a transfusion-dependent adolescent boy with iron-overload secondary to congenital dyserythropoietic anemia who developed severe E. coli monomicrobial necrotizing fasciitis of the leg following a minor trauma. Combined surgical, antimicrobial and supportive care resulted in a good outcome.


Subject(s)
Blood Transfusion , Escherichia coli Infections/complications , Escherichia coli/pathogenicity , Fasciitis, Necrotizing/diagnosis , Iron Overload/complications , Adolescent , Escherichia coli Infections/diagnosis , Fasciitis, Necrotizing/microbiology , Humans , Iron Overload/microbiology , Male
4.
Cell Microbiol ; 21(8): e13034, 2019 08.
Article in English | MEDLINE | ID: mdl-31013389

ABSTRACT

How Salmonella enterica serovar Typhi (S. Typhi), an important human pathogen, survives the stressful microenvironments inside the gastrointestinal tract and within macrophages remains poorly understood. We report here that S. Typhi has a bonafide stringent response (SR) system, which is mediated by (p)ppGpp and regulates multiple virulence-associated traits and the pathogenicity of the S. Typhi Ty2 strain. In an iron overload mouse model of S. Typhi infection, the (p)ppGpp0 (Ty2ΔRelAΔSpoT) strain showed minimal systemic spread and no mortality, as opposed to 100% death of the mice challenged with the isogenic wild-type strain. Ty2ΔRelAΔSpoT had markedly elongated morphology with incomplete septa formation and demonstrated severely attenuated motility and chemotaxis due to the loss of flagella. Absence of the Vi-polysaccharide capsule rendered the mutant strain highly susceptible to complement-mediated lysis. The phenotypes of Ty2ΔRelAΔSpoT was contributed by transcriptional repression of several genes, including fliC, tviA, and ftsZ, as found by reverse transcriptase quantitative polymerase chain reaction and gene complementation studies. Finally, Ty2ΔRelAΔSpoT had markedly reduced invasion into intestinal epithelial cells and significantly attenuated survival within macrophages. To the best of our knowledge, this was the first study that addressed SR in S. Typhi and showed that (p)ppGpp was essential for optimal pathogenic fitness of the organism.


Subject(s)
Bacterial Proteins/genetics , Guanosine Pentaphosphate/metabolism , Host-Pathogen Interactions/genetics , Salmonella typhi/genetics , Salmonella typhi/pathogenicity , Typhoid Fever/microbiology , Animals , Bacterial Proteins/metabolism , Caco-2 Cells , Disease Models, Animal , GTP Pyrophosphokinase/deficiency , GTP Pyrophosphokinase/genetics , Gene Expression Regulation, Bacterial , HT29 Cells , Humans , Iron Overload/metabolism , Iron Overload/microbiology , Iron Overload/mortality , Iron Overload/pathology , Liver/metabolism , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Polysaccharides, Bacterial/deficiency , Pyrophosphatases/deficiency , Pyrophosphatases/genetics , RAW 264.7 Cells , Salmonella typhi/growth & development , Salmonella typhi/metabolism , Signal Transduction , Spleen/metabolism , Spleen/microbiology , Spleen/pathology , Survival Analysis , THP-1 Cells , Typhoid Fever/metabolism , Typhoid Fever/mortality , Typhoid Fever/pathology , Virulence
5.
J Trace Elem Med Biol ; 47: 12-20, 2018 May.
Article in English | MEDLINE | ID: mdl-29544798

ABSTRACT

The aim of this study was to determine the toxicological effects of excess iron in a liquid iron preparation (especially on intestinal barrier function) and the possible etiology of side effects or diseases caused by the excess iron. In study 1, forty male Sprague-Dawley rats (4-5 wk old) were subjected to oral gavage with 1 ml vehicle (0.01 mol/L HCl) or 1 ml liquid iron preparation containing 8 mg, 16 mg or 24 mg of iron for 30 d. Iron status, oxidative stress, histology (H&E staining), ultrastructure (electron microscopy) and apoptosis (TUNEL assay) in the intestines and liver were assessed. The cecal microbiota was evaluated by 16S rRNA sequencing. In study 2, twenty rats with the same profile as above were subjected to oral gavage with 1 ml vehicle or 24 mg Fe for 30 d. The intestinal barrier function was determined by in vivo studies and an Ussing chamber assay; tight junction proteins and serum pro-inflammatory cytokines were observed by enzyme-linked immunosorbent assay. In study 1, the intestinal mucosa and liver showed apparent oxidative stress. In addition, iron concentration-dependent ultrastructural alterations to duodenal enterocytes and hepatocytes and histological damage to the colonic mucosa were detected. Notably, apoptosis was increased in duodenal enterocytes and hepatocytes. Impaired intestinal barrier function and lower expression of intestinal tight junction proteins were observed, and the phenotype was more severe in the colon than in the duodenum. A trend toward higher expression of serum pro-inflammatory cytokines might indicate systemic inflammation. Furthermore, the caecal microbiota showed a significant change, with increased Defluviitaleaceae, Ruminococcaceae, and Coprococcus and reduced Lachnospiraceae and Allobaculum, which could mediate the detrimental effects of excess iron on gut health. We concluded that excessive iron exposure from liquid iron preparation induces oxidative stress and histopathological alterations in the intestine and liver. Impaired intestinal barrier function could increase iron transportation, and inflammation along with oxidative stress-enhanced liver iron deposition may cause further liver injury in a vicious circle. These effects were accompanied by lower intestinal segment damage and altered gut microbial composition of rats toward a profile with an increased risk of gut disease.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Iron Overload/physiopathology , Iron/administration & dosage , Administration, Oral , Animals , Body Weight/drug effects , Chemical and Drug Induced Liver Injury/physiopathology , Intestines/physiopathology , Intestines/ultrastructure , Iron/adverse effects , Iron Overload/microbiology , Male , Oxidative Stress/drug effects , Rats, Sprague-Dawley
7.
Blood ; 130(3): 245-257, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28465342

ABSTRACT

The iron-regulatory hormone hepcidin is induced early in infection, causing iron sequestration in macrophages and decreased plasma iron; this is proposed to limit the replication of extracellular microbes, but could also promote infection with macrophage-tropic pathogens. The mechanisms by which hepcidin and hypoferremia modulate host defense, and the spectrum of microbes affected, are poorly understood. Using mouse models, we show that hepcidin was selectively protective against siderophilic extracellular pathogens (Yersinia enterocolitica O9) by controlling non-transferrin-bound iron (NTBI) rather than iron-transferrin concentration. NTBI promoted the rapid growth of siderophilic but not nonsiderophilic bacteria in mice with either genetic or iatrogenic iron overload and in human plasma. Hepcidin or iron loading did not affect other key components of innate immunity, did not indiscriminately promote intracellular infections (Mycobacterium tuberculosis), and had no effect on extracellular nonsiderophilic Y enterocolitica O8 or Staphylococcus aureus Hepcidin analogs may be useful for treatment of siderophilic infections.


Subject(s)
Catheter-Related Infections/immunology , Hemochromatosis/immunology , Hepcidins/immunology , Iron Overload/immunology , Iron/metabolism , Staphylococcal Infections/immunology , Animals , Binding, Competitive , Catheter-Related Infections/metabolism , Catheter-Related Infections/microbiology , Catheter-Related Infections/mortality , Disease Models, Animal , Disease Resistance , Gene Expression , Hemochromatosis/metabolism , Hemochromatosis/microbiology , Hemochromatosis/mortality , Hepcidins/agonists , Hepcidins/deficiency , Hepcidins/genetics , Humans , Iron/immunology , Iron Overload/metabolism , Iron Overload/microbiology , Iron Overload/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Oligopeptides/pharmacology , Protein Binding , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Staphylococcus aureus , Survival Analysis , Transferrin/genetics , Transferrin/metabolism , Yersinia enterocolitica/drug effects , Yersinia enterocolitica/growth & development , Yersinia enterocolitica/metabolism
8.
J Nutr Biochem ; 41: 25-33, 2017 03.
Article in English | MEDLINE | ID: mdl-27951517

ABSTRACT

Mammalian siderophores are believed to play a critical role in maintaining iron homeostasis. However, the properties and functions of mammalian siderophores have not been fully clarified. In this study, we have employed Chrome Azurol S (CAS) assay which is a well-established method for bacterial siderophores study, to detect and quantify mammalian siderophores in urine samples. Our study demonstrates that siderophores in urine can be altered by diet, gut microbiota and inflammation. C57BL/6 mice, fed on plant-based chow diets which contain numerous phytochemicals, have more siderophores in the urine compared to those fed on purified diets. Urinary siderophores were up-regulated in iron overload conditions, but not altered by other tested nutrients status. Further, germ-free mice displayed 50% reduced urinary siderophores, in comparison to conventional mice, indicating microbiota biotransformation is critical in generating or stimulating host metabolism to create more siderophores. Altered urinary siderophores levels during inflammation suggest that host health conditions influence systemic siderophores level. This is the first report to measure urinary siderophores as a whole, describing how siderophores levels are modulated under different physiological conditions. We believe that our study opens up a new field in mammalian siderophores research and the technique we used in a novel manner has the potential to be applied to clinical purpose.


Subject(s)
Anemia, Iron-Deficiency/urine , Colitis/urine , Diet/adverse effects , Gastrointestinal Microbiome , Iron Overload/urine , Siderophores/urine , Vitamin A Deficiency/urine , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/immunology , Anemia, Iron-Deficiency/microbiology , Animals , Biomarkers/blood , Biomarkers/urine , Colitis/chemically induced , Colitis/immunology , Colitis/microbiology , Crosses, Genetic , Diet, High-Fat/adverse effects , Female , Germ-Free Life , Hemochromatosis Protein/genetics , Hemochromatosis Protein/metabolism , Iron Overload/etiology , Iron Overload/immunology , Iron Overload/microbiology , Lipocalin-2/genetics , Lipocalin-2/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/urine , Selenium/deficiency , Selenium/immunology , Selenium/poisoning , Vitamin A Deficiency/etiology , Vitamin A Deficiency/immunology , Vitamin A Deficiency/microbiology
9.
Annu Rev Genet ; 50: 67-91, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27617971

ABSTRACT

Transition metals are required trace elements for all forms of life. Due to their unique inorganic and redox properties, transition metals serve as cofactors for enzymes and other proteins. In bacterial pathogenesis, the vertebrate host represents a rich source of nutrient metals, and bacteria have evolved diverse metal acquisition strategies. Host metal homeostasis changes dramatically in response to bacterial infections, including production of metal sequestering proteins and the bombardment of bacteria with toxic levels of metals. In response, bacteria have evolved systems to subvert metal sequestration and toxicity. The coevolution of hosts and their bacterial pathogens in the battle for metals has uncovered emerging paradigms in social microbiology, rapid evolution, host specificity, and metal homeostasis across domains. This review focuses on recent advances and open questions in our understanding of the complex role of transition metals at the host-pathogen interface.


Subject(s)
Bacteria/metabolism , Bacteria/pathogenicity , Host-Pathogen Interactions , Metals/metabolism , Animals , Bacterial Infections , Deficiency Diseases/microbiology , Diet , Heme/metabolism , Humans , Iron/metabolism , Iron Overload/microbiology , Siderophores/metabolism
10.
PLoS One ; 8(2): e55331, 2013.
Article in English | MEDLINE | ID: mdl-23390527

ABSTRACT

Salmonella, a ubiquitous Gram-negative intracellular bacterium, is a food borne pathogen that infects a broad range of hosts. Infection with Salmonella Typhimurium in mice is a broadly recognized experimental model resembling typhoid fever in humans. Using a N-ethyl-N-nitrosurea (ENU) mutagenesis recessive screen, we report the identification of Ity16 (Immunity to Typhimurium locus 16), a locus responsible for increased susceptibility to infection. The position of Ity16 was refined on chromosome 8 and a nonsense mutation was identified in the ankyrin 1 (Ank1) gene. ANK1 plays an important role in the formation and stabilization of the red cell cytoskeleton. The Ank1(Ity16/Ity16) mutation causes severe hemolytic anemia in uninfected mice resulting in splenomegaly, hyperbilirubinemia, jaundice, extramedullary erythropoiesis and iron overload in liver and kidneys. Ank1(Ity16/Ity16) mutant mice demonstrated low levels of hepcidin (Hamp) expression and significant increases in the expression of the growth differentiation factor 15 (Gdf15), erythropoietin (Epo) and heme oxygenase 1 (Hmox1) exacerbating extramedullary erythropoiesis, tissue iron deposition and splenomegaly. As the infection progresses in Ank1(Ity16/Ity16), the anemia worsens and bacterial load were high in liver and kidneys compared to wild type mice. Heterozygous Ank1(+/Ity16) mice were also more susceptible to Salmonella infection although to a lesser extent than Ank1(Ity16/Ity16) and they did not inherently present anemia and splenomegaly. During infection, iron accumulated in the kidneys of Ank1(+/Ity16) mice where bacterial loads were high compared to littermate controls. The critical role of HAMP in the host response to Salmonella infection was validated by showing increased susceptibility to infection in Hamp-deficient mice and significant survival benefits in Ank1(+/Ity16) heterozygous mice treated with HAMP peptide. This study illustrates that the regulation of Hamp and iron balance are crucial in the host response to Salmonella infection in Ank1 mutants.


Subject(s)
Anemia, Hemolytic/genetics , Ankyrins/genetics , Antimicrobial Cationic Peptides/genetics , Codon, Nonsense/drug effects , Ethylnitrosourea/toxicity , Iron Overload/genetics , Salmonella Infections/genetics , Anemia, Hemolytic/metabolism , Anemia, Hemolytic/microbiology , Anemia, Hemolytic/mortality , Animals , Ankyrins/metabolism , Antimicrobial Cationic Peptides/deficiency , Erythrocytes/metabolism , Erythrocytes/pathology , Erythropoietin/genetics , Erythropoietin/metabolism , Gene Expression , Genetic Predisposition to Disease , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hepcidins , Heterozygote , Homozygote , Iron/metabolism , Iron Overload/metabolism , Iron Overload/microbiology , Iron Overload/mortality , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/mortality , Salmonella typhimurium/physiology , Survival Analysis
11.
Transfus Apher Sci ; 48(1): 103-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22985533

ABSTRACT

Invasive fungal pneumonia (IFP) has become increasingly common in patients that previously underwent alloHSCT. The aim of this study was to determine the role of hyperferritinemia, via iron overload in invasive fungal pneumonia in patients that underwent alloHSCT. Medical records of 73 patients with pneumonia that underwent alloHSCT were studied retrospectively, whereby a pre-transplantation serum ferritin level measured up to 100 days prior to transplantation of patients with invasive fungal pneumonia (IFP) and non-fungal pneumonia (non-IFP) was compared. Patient records revealed 35 and 38 cases of IFP and non-IFP, respectively. In risk evaluation for IFP, age, gender, HLA status, conditioning regimen, smoking history, and underlying disease were not significantly different among groups (p>0.05). However, performance status (Karnofsky) was significantly lower in patients with IFP (p<0.05). The median ferritin levels were 1,705 ng/ml (41-7198) in the IFP group and 845 ng/ml (18-7099) in non-IFP group and the difference was found statistically significant (p=0.001). Elevated pretransplant serum ferritin level is associated with IFP in patients that underwent alloHSCT, in particular when values exceed 1550 ng/ml.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Iron Overload/etiology , Lung Diseases, Fungal/blood , Pneumonia/blood , Adolescent , Adult , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Iron Overload/blood , Iron Overload/microbiology , Lung Diseases, Fungal/diagnosis , Lung Diseases, Fungal/microbiology , Male , Middle Aged , Pneumonia/microbiology , Retrospective Studies , Risk Factors , Transplantation, Homologous , Young Adult
12.
Hemoglobin ; 33(5): 352-60, 2009.
Article in English | MEDLINE | ID: mdl-19814682

ABSTRACT

Infections are among the leading causes of death for thalassemia major patients. The known predisposing factors of infection include prior splenectomy, iron overload and use of iron chelator such as deferoxamine (DFO). While encapsulated organisms frequently found in splenectomized patients were readily controlled by prophylactic vaccination and vigilant antibiotic treatment, ferrophilic organisms such as Yersinia and Klebsiella remain common pathogens in thalassemic patients. Yersinia infections are more prevalent in temperate regions and Klebsiella infections are commonly found in tropical and subtropical areas. While the use of DFO further aggravates the risk of Yersinia infection, oral chelators such as deferiprone (L1) do not enhance the growth of Yersinia in vitro or in vivo. We found that the growth of Klebsiella was marginally enhanced by DFO in vitro when compared to Yersinia. Such an unfavorable effect was not found in either L1 or deferasirox (DFRA) in vitro. The growth of Aeromonas was not affected by the presence of all three forms of chelators. Therefore, we suggest that factors other than DFO may account for the increased prevalence of Klebsiella and Aeromonas infection in Asian thalassemic patients.


Subject(s)
Aeromonas hydrophila/drug effects , Deferoxamine/adverse effects , Gram-Negative Bacterial Infections/chemically induced , Iron Chelating Agents/adverse effects , Klebsiella Infections/chemically induced , Klebsiella pneumoniae/drug effects , Thalassemia/microbiology , Aeromonas hydrophila/growth & development , Aeromonas hydrophila/isolation & purification , Benzoates/adverse effects , Benzoates/pharmacology , Blood Transfusion , Deferasirox , Deferiprone , Deferoxamine/pharmacology , Gram-Negative Bacterial Infections/epidemiology , Humans , Iron Chelating Agents/pharmacology , Iron Overload/drug therapy , Iron Overload/microbiology , Kinetics , Klebsiella Infections/epidemiology , Klebsiella pneumoniae/growth & development , Klebsiella pneumoniae/isolation & purification , Prevalence , Pyridones/adverse effects , Pyridones/pharmacology , Thalassemia/drug therapy , Triazoles/adverse effects , Triazoles/pharmacology
13.
Blood ; 111(2): 924-31, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17938254

ABSTRACT

The liver peptide hepcidin regulates iron absorption and recycling. Hemojuvelin (HJV) has a key role in hepcidin regulation, and its inactivation causes severe iron overload both in humans and in mice. Membrane HJV (m-HJV) acts as a coreceptor for bone morphogenetic proteins (BMPs), whereas soluble HJV (s-HJV) may down-regulate hepcidin in a competitive way interfering with BMP signaling. s-HJV is decreased by iron in vitro and increased by iron deficiency in vivo. However, the mechanisms regulating the 2 HJV isoforms remain unclear. Here we show that s-HJV originates from a furin cleavage at position 332-335. s-HJV is reduced in the cleavage mutant R335Q as well as in cells treated with a furin inhibitor, and increased in cells overexpressing exogenous furin, but not in cells overexpressing an inactive furin variant. Furin is up-regulated by iron deficiency and hypoxia in association with the stabilization of HIF-1alpha. Increased s-HJV in response to HIF-1alpha occurs during differentiation of murine muscle cells expressing endogenous Hjv. Our data are relevant to the mechanisms that relate iron metabolism to the hypoxic response. The release of s-HJV might be a tissue-specific mechanism, signaling the local iron requests of hypoxic skeletal muscles independently of the oxygen status of the liver.


Subject(s)
Furin/metabolism , Homeostasis/physiology , Iron Overload/microbiology , Iron/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Amino Acid Substitution , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Cell Hypoxia/physiology , Down-Regulation/physiology , Furin/genetics , GPI-Linked Proteins , HeLa Cells , Hemochromatosis Protein , Hepcidins , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Iron Deficiencies , Iron Overload/genetics , Liver/cytology , Membrane Proteins/genetics , Mice , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Mutation, Missense , Organ Specificity/physiology , Oxygen/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/physiology
14.
Int J Antimicrob Agents ; 29(4): 465-70, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17241773

ABSTRACT

Mycobacterium tuberculosis, human immunodeficiency virus (HIV) and iron overload (dietary/hereditary) are very common in sub-Saharan Africa. The requirement for iron as a crucial factor for cellular processes is well established, as are the disadvantages of excess iron in the system. Mycobacterium tuberculosis and HIV are believed to have a reciprocal effect on each another. An in vitro model was evaluated where chronically HIV-infected cells were secondarily exposed to M. tuberculosis in the presence of iron overload. Co-infection alone caused cell type-specific reductions in host cell viability, more than doubled the number of viral particles and stimulated bacterial viability. Excess iron (in addition to co-infection) further decreased cell viability, with a marked increase in necrosis (rather than apoptosis) of cells, and was also found to enhance both HIV (26%; P<0.01) and M. tuberculosis (47%; P<0.01) replication. Chelation of excess iron with deferoxamine abrogated the enhanced replication of the pathogens, with a marginal restoration in host cell viability. These findings demonstrate that (i) increased levels of iron in HIV-infected patients secondarily co-infected with M. tuberculosis elevate viral replication, which could lead to rapid disease progression, and (ii) iron chelation may serve as a means to slow/decelerate these processes.


Subject(s)
HIV Infections/pathology , Iron Overload/microbiology , Tuberculosis/pathology , AIDS-Related Opportunistic Infections/microbiology , AIDS-Related Opportunistic Infections/pathology , Cells, Cultured , HIV/pathogenicity , HIV Infections/microbiology , Humans , Iron Overload/pathology , Leukocytes, Mononuclear/microbiology , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/virology , Mycobacterium tuberculosis , Necrosis , Tuberculosis/microbiology
15.
Prog Transplant ; 17(4): 332-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18240700

ABSTRACT

Iron is a critical nutrient source and contributes to staphylococcal pathogenesis. We assessed the role of hepatic explant iron overload as a risk factor for Staphylococcus aureus bacteremia in liver transplant recipients. Seven of 13 cases with S aureus bacteremia (53.8%) had hepatic explant iron concentrations that exceeded normal limits (grade > or = 2). Length of posttransplant intensive care unit stay (P= .013) and hepatocellular carcinoma as underlying liver disease (P = .04), but not hepatic explant iron concentration, correlated with a higher risk of S aureus bacteremia after transplantation. However, noncarriers (patients without S aureus nasal carriage) who developed S aureus bacteremia were more likely to have high hepatic iron content; 4 of 7 (57%) noncarriers with high-grade iron content developed S aureus bacteremia but no noncarriers with low-grade iron content did (P = .07). All noncarriers who became infected had high iron content (grade > or = 2) of the hepatic explant. A readily quantifiable assessment of hepatic iron at the time of transplantation can potentially identify patients without carriage who may be at risk for early S aureus bacteremia.


Subject(s)
Bacteremia/etiology , Iron Overload/microbiology , Liver Transplantation/adverse effects , Liver/metabolism , Staphylococcal Infections/etiology , Adult , Aged , Humans , Middle Aged , Proportional Hazards Models , Retrospective Studies , Risk Factors
16.
Infect Immun ; 68(3): 1724-6, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10678997

ABSTRACT

Killing of intracellular Penicillium marneffei conidia is demonstrated in gamma interferon-lipopolysaccharide-activated human THP1 and mouse J774 cells. Iron overload significantly reduces the antifungal activity of macrophages. Likewise, exogenous iron enhances and iron chelators inhibit the extracellular growth of P. marneffei. These results suggest that iron availability critically affects immunity to and the pathogenicity of P. marneffei.


Subject(s)
Iron/pharmacology , Penicillium/drug effects , Animals , Deferoxamine/pharmacology , Humans , Interferon-gamma/pharmacology , Iron Overload/microbiology , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide/physiology , Penicillium/growth & development
17.
Infect Immun ; 66(12): 5659-68, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9826339

ABSTRACT

Vibrio vulnificus expresses a number of potential virulence determinants that may contribute to its ability to cause a severe and rapidly disseminating septicemia in susceptible hosts. We have cloned and characterized two genes encoding products related to components of the type IV pilus biogenesis and general secretory (type II) pathways by complementation of a type IV peptidase/N-methyltransferase (PilD) mutant of Pseudomonas aeruginosa with a V. vulnificus genomic library. One of the genes (vvpD) encodes a protein homologous to PilD and other members of the type IV peptidase family that completely restores this activity in a P. aeruginosa mutant deficient in the expression of PilD. The other gene (vvpC) encodes a homolog of PilC from P. aeruginosa, where it is essential for assembly of type IV pili. Phenotypic characterization of a V. vulnificus vvpD mutant, constructed by allelic exchange, showed that VvpD is required for the expression of surface pili, suggesting that the pili observed on V. vulnificus are of the type IV class. This mutant was also unable to secrete at least three extracellular degradative enzymes, and the localization of one of these (the cytolysin/hemolysin) to the periplasmic space indicates that these proteins are normally exported via the type II secretion pathway. Loss of VvpD resulted in significant decreases in CHO cell cytotoxicity, adherence to HEp-2 cells, and virulence in a mouse model. Capsule formation and serum resistance were not affected in the vvpD mutant, indicating that in addition to capsule, virulence of V. vulnificus requires type IV pili and/or extracellular secretion of several exoenzymes.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/metabolism , Fimbriae Proteins , Fimbriae, Bacterial/metabolism , Iron Overload/microbiology , Vibrio/pathogenicity , Amino Acid Sequence , Animals , Bacterial Capsules , Bacterial Proteins/genetics , CHO Cells , Cloning, Molecular , Conserved Sequence , Cricetinae , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Genetic Complementation Test , Liver/cytology , Liver/microbiology , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Molecular Sequence Data , Pseudomonas aeruginosa , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Toxicity Tests , Vibrio/enzymology , Vibrio/ultrastructure
18.
J Infect Dis ; 175(6): 1467-76, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9180188

ABSTRACT

The effect of iron overload on susceptibility of mice to Candida albicans infection and on the type of T helper (Th) immunity elicited was investigated. Iron overload greatly increased susceptibility to disseminated infection with low-virulence C. albicans cells of exogenous origin. The candidacidal activity and the ability to release nitric oxide and bioactive interleukin (IL)-12 were greatly impaired in neutrophils and macrophages from infected mice. CD4 T cells from spleens of iron-overloaded mice were found to produce high levels of IL-4 and IL-10 and low levels of interferon-gamma. Treatment of iron-overloaded mice with the iron chelator, deferoxamine, resulted in the cure of mice from infection, restored the antifungal effector and immunomodulatory functions of the phagocytic cells, and allowed the occurrence of CD4 Th1 protective antifungal responses. These data indicate that iron overload may negatively affect CD4 Th1 development in mice with candidiasis, a function efficiently restored by therapy with deferoxamine.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Iron Overload/immunology , Th1 Cells/immunology , Animals , Candida albicans/drug effects , Candidiasis/metabolism , Cytokines/analysis , Cytokines/blood , Deferoxamine/pharmacology , Disease Susceptibility , Ferric Compounds/pharmacology , Gene Expression , Immunoglobulin E/blood , Iron/pharmacology , Iron Chelating Agents/pharmacology , Iron Overload/chemically induced , Iron Overload/microbiology , Kidney/immunology , Kidney/microbiology , Liver/microbiology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Nitrates/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , RNA/analysis
SELECTION OF CITATIONS
SEARCH DETAIL