Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Metabolites ; 12(2)2022 Jan 18.
Article En | MEDLINE | ID: mdl-35208164

Iron absorption requires an acidic environment that is generated by the activity of the proton pump gastric H(+)/K(+)ATPase (ATP4), expressed in gastric parietal cells. However, hepcidin, the iron regulatory peptide that inhibits iron absorption, unexpectedly upregulates ATP4 and increases gastric acidity. Thus, a concept of link between acidosis and alterations in iron metabolism, needs to be explored. We investigated this aspect in-vivo using experimental models of NH4Cl-induced acidosis and of an iron-rich diet. Under acidosis, gastric ATP4 was augmented. Serum hepcidin was induced and its mRNA level was increased in the liver but not in the stomach, a tissue where hepcidin is also expressed. mRNA and protein levels of intestinal DMT1(Divalent Metal Transporter 1) and ferroportin were downregulated. Serum iron level and transferrin saturation remained unchanged, but serum ferritin was significantly increased. Under iron-rich diet, the protein expression of ATP4A was increased and serum, hepatic and gastric hepcidin were all induced. Taken together, these results provide evidence of in-vivo relationship between iron metabolism and acidosis. For clinical importance, we speculate that metabolic acidosis may contribute in part to the pathologic elevation of serum hepcidin levels seen in patients with chronic kidney disease. The regulation of ATP4 by iron metabolism may also be of interest for patients with hemochromatosis.

3.
Blood ; 136(21): 2457-2468, 2020 11 19.
Article En | MEDLINE | ID: mdl-32678895

Congenital erythropoietic porphyria (CEP) is an inborn error of heme synthesis resulting from uroporphyrinogen III synthase (UROS) deficiency and the accumulation of nonphysiological porphyrin isomer I metabolites. Clinical features are heterogeneous among patients with CEP but usually combine skin photosensitivity and chronic hemolytic anemia, the severity of which is related to porphyrin overload. Therapeutic options include symptomatic strategies only and are unsatisfactory. One promising approach to treating CEP is to reduce the erythroid production of porphyrins through substrate reduction therapy by inhibiting 5-aminolevulinate synthase 2 (ALAS2), the first and rate-limiting enzyme in the heme biosynthetic pathway. We efficiently reduced porphyrin accumulation after RNA interference-mediated downregulation of ALAS2 in human erythroid cellular models of CEP disease. Taking advantage of the physiological iron-dependent posttranscriptional regulation of ALAS2, we evaluated whether iron chelation with deferiprone could decrease ALAS2 expression and subsequent porphyrin production in vitro and in vivo in a CEP murine model. Treatment with deferiprone of UROS-deficient erythroid cell lines and peripheral blood CD34+-derived erythroid cultures from a patient with CEP inhibited iron-dependent protein ALAS2 and iron-responsive element-binding protein 2 expression and reduced porphyrin production. Furthermore, porphyrin accumulation progressively decreased in red blood cells and urine, and skin photosensitivity in CEP mice treated with deferiprone (1 or 3 mg/mL in drinking water) for 26 weeks was reversed. Hemolysis and iron overload improved upon iron chelation with full correction of anemia in CEP mice treated at the highest dose of deferiprone. Our findings highlight, in both mouse and human models, the therapeutic potential of iron restriction to modulate the phenotype in CEP.


Anemia, Hemolytic/drug therapy , Deferiprone/therapeutic use , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Photosensitivity Disorders/drug therapy , Porphyria, Erythropoietic/drug therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/biosynthesis , 5-Aminolevulinate Synthetase/genetics , Adult , Anemia, Hemolytic/etiology , Animals , CRISPR-Cas Systems , Cell Line , Cell Line, Tumor , Disease Models, Animal , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Iron/metabolism , Iron Overload/etiology , Leukemia, Erythroblastic, Acute/pathology , Mice , Peripheral Blood Stem Cells/drug effects , Peripheral Blood Stem Cells/metabolism , Photosensitivity Disorders/etiology , Porphyria, Acute Intermittent/metabolism , Porphyria, Erythropoietic/complications , Porphyrins/biosynthesis , RNA Interference , RNA, Small Interfering/pharmacology
4.
World J Clin Cases ; 7(15): 1926-1936, 2019 Aug 06.
Article En | MEDLINE | ID: mdl-31423425

Hepcidin is the hyposideremic hormone regulating iron metabolism. It is a defensin-like disulfide-bonded peptide with antimicrobial activity. The main site of hepcidin production is the liver where its synthesis is modulated by iron, inflammation and erythropoietic signaling. However, hepcidin locally produced in several peripheral organs seems to be an important actor for the maintenance of iron homeostasis in these organs. This review highlights the presence of peripheral hepcidin and its potential functions. Understanding the role of extrahepatic hepcidin could be of great physiological and therapeutic importance for several specific pathologies.

5.
Am J Hum Genet ; 104(2): 341-347, 2019 02 07.
Article En | MEDLINE | ID: mdl-30712775

Erythropoietic protoporphyria (EPP) is a hereditary disease characterized by a deficiency in ferrochelatase (FECH) activity. FECH activity is responsible for the accumulation of protoporphyrin IX (PPIX). Without etiopathogenic treatment, EPP manifests as severe photosensitivity. 95% of affected individuals present a hypomorphic FECH allele trans to a loss-of-function (LOF) FECH mutation, resulting in a reduction in FECH activity in erythroblasts below a critical threshold. The hypomorphic allele promotes the use of a cryptic acceptor splice site, generating an aberrant FECH mRNA, which is responsible for the reduced level of wild-type FECH mRNA and, ultimately, FECH activity. We have previously identified an antisense oligonucleotide (AON), AON-V1 (V1), that redirects splicing to the physiological acceptor site and reduces the accumulation of PPIX. Here, we developed a specific strategy that uses transferrin receptor 1 (TRF1) as a Trojan horse to deliver V1 to erythroid progenitors. We designed a bifunctional peptide (P1-9R) including a TFR1-targeting peptide coupled to a nine-arginine cell-penetrating peptide (CPP) that facilitates the release of the AON from TFR1 in endosomal vesicles. We demonstrated that the P1-9R/V1 nanocomplex promotes the efficient and prolonged redirection of splicing towards the physiological splice site and subsequent normalization of WT FECH mRNA and protein levels. Finally, the P1-9R/V1 nanocomplex increases WT FECH mRNA production and significantly decreases PPIX accumulation in primary cultures of differentiating erythroid progenitors from an overt EPP-affected individual. P1-9R is a method designed to target erythroid progenitors and represents a potentially powerful tool for the in vivo delivery of therapeutic DNA in many erythroid disorders.


Antigens, CD/metabolism , Cell-Penetrating Peptides/metabolism , Erythroid Precursor Cells/metabolism , Genetic Therapy/methods , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy , Receptors, Transferrin/metabolism , Antigens, CD/administration & dosage , Antigens, CD34/metabolism , Cell Line , Cell-Penetrating Peptides/administration & dosage , Erythroblasts/cytology , Erythroblasts/metabolism , Ferrochelatase/genetics , Ferrochelatase/metabolism , Humans , Ligands , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protoporphyrins/metabolism , RNA, Messenger , Receptors, Transferrin/administration & dosage
6.
Mol Genet Metab ; 128(3): 190-197, 2019 11.
Article En | MEDLINE | ID: mdl-30737140

Recently, new genes and molecular mechanisms have been identified in patients with porphyrias and sideroblastic anemias (SA). They all modulate either directly or indirectly the δ-aminolevulinic acid synthase (ALAS) activity. ALAS, is encoded by two genes: the erythroid-specific (ALAS2), and the ubiquitously expressed (ALAS1). In the liver, ALAS1 controls the rate-limiting step in the production of heme and hemoproteins that are rapidly turned over in response to metabolic needs. Several heme regulatory targets have been identified as regulators of ALAS1 activity: 1) transcriptional repression via a heme-responsive element, 2) post-transcriptional destabilization of ALAS1 mRNA, 3) post-translational inhibition via a heme regulatory motif, 4) direct inhibition of the activity of the enzyme and 5) breakdown of ALAS1 protein via heme-mediated induction of the protease Lon peptidase 1. In erythroid cells, ALAS2 is a gatekeeper of production of very large amounts of heme necessary for hemoglobin synthesis. The rate of ALAS2 synthesis is transiently increased during the period of active heme synthesis. Its gene expression is determined by trans-activation of nuclear factor GATA1, CACC box and NF-E2-binding sites in the promoter areas. ALAS2 mRNA translation is also regulated by the iron-responsive element (IRE)/iron regulatory proteins (IRP) binding system. In patients, ALAS enzyme activity is affected in most of the mutations causing non-syndromic SA and in several porphyrias. Decreased ALAS2 activity results either directly from loss-of-function ALAS2 mutations as seen in X-linked sideroblastic anemia (XLSA) or from defect in the availability of one of its two mitochondrial substrates: glycine in SLC25A38 mutations and succinyl CoA in GLRX5 mutations. Moreover, ALAS2 gain of function mutations is responsible for X-linked protoporphyria and increased ALAS1 activity lead to acute attacks of hepatic porphyrias. A missense dominant mutation in the Walker A motif of the ATPase binding site in the gene coding for the mitochondrial protein unfoldase CLPX also contributes to increasing ALAS and subsequently protoporphyrinemia. Altogether, these recent data on human ALAS have informed our understanding of porphyrias and sideroblastic anemias pathogeneses and may contribute to new therapeutic strategies.


5-Aminolevulinate Synthetase/genetics , Aminolevulinic Acid/metabolism , Anemia, Sideroblastic/genetics , Gene Expression Regulation , Porphyrias/genetics , 5-Aminolevulinate Synthetase/metabolism , Anemia, Sideroblastic/enzymology , Animals , Binding Sites , GATA1 Transcription Factor/genetics , Heme/biosynthesis , Humans , Liver/metabolism , Mice , Mutation, Missense , Porphyrias/enzymology , Promoter Regions, Genetic
7.
Mol Genet Metab ; 128(3): 342-351, 2019 11.
Article En | MEDLINE | ID: mdl-30660387

Non-syndromic microcytic congenital sideroblastic anemia (cSA) is predominantly caused by defective genes encoding for either ALAS2, the first enzyme of heme biosynthesis pathway or SLC25A38, the mitochondrial importer of glycine, an ALAS2 substrate. Herein we explored a new case of cSA with two mutations in GLRX5, a gene for which only two patients have been reported so far. The patient was a young female with biallelic compound heterozygous mutations in GLRX5 (p.Cys67Tyr and p.Met128Lys). Three-D structure analysis confirmed the involvement of Cys67 in the coordination of the [2Fe2S] cluster and suggested a potential role of Met128 in partner interactions. The protein-level of ferrochelatase, the terminal-enzyme of heme process, was increased both in patient-derived lymphoblastoid and CD34+ cells, however, its activity was drastically decreased. The activity of ALAS2 was found altered and possibly related to a defect in the biogenesis of its co-substrate, the succinyl-CoA. Thus, the patient exhibits both a very low ferrochelatase activity without any accumulation of porphyrins precursors in contrast to what is reported in erythropoietic protoporphyria with solely impaired ferrochelatase activity. A significant oxidative stress was evidenced by decreased reduced glutathione and aconitase activity, and increased MnSOD protein expression. This oxidative stress depleted and damaged mtDNA, decreased complex I and IV activities and depleted ATP content. Collectively, our study demonstrates the key role of GLRX5 in modulating ALAS2 and ferrochelatase activities and in maintaining mitochondrial function.


5-Aminolevulinate Synthetase/genetics , Anemia, Sideroblastic/genetics , Ferrochelatase/genetics , Genetic Diseases, X-Linked/genetics , Glutaredoxins/genetics , Heme/biosynthesis , Mutation, Missense , 5-Aminolevulinate Synthetase/metabolism , Aconitate Hydratase/metabolism , Adolescent , Amino Acid Sequence , Anemia, Sideroblastic/enzymology , Cell Line, Transformed , Female , Ferrochelatase/metabolism , Genetic Diseases, X-Linked/enzymology , Glutathione/metabolism , Humans , Mitochondria/enzymology , Oxidative Stress , Pedigree , Protein Structure, Tertiary
8.
Neuromuscul Disord ; 28(7): 564-571, 2018 07.
Article En | MEDLINE | ID: mdl-29776718

Duchenne muscular dystrophy (DMD) is an X-linked disease caused by mutations in the dystrophin gene leading to the absence of the normal dystrophin protein. The efforts of many laboratories brought new treatments of DMD to the reality, but ongoing and forthcoming clinical trials suffer from absence of valuable biomarkers permitting to follow the outcome of the treatment day by day and to adjust the treatment if needed. In the present study the levels of 128 urinary proteins including growth factors, cytokines and chemokines were compared in urine of DMD patients and age related control subjects by antibody array approach. Surprisingly, statistically significant difference was observed only for urinary ferritin whose level was 50 times higher in young DMD patients. To explain the observed high urinary ferritin content we analysed the levels of iron, iron containing proteins and proteins involved in regulation of iron metabolism in serum and urine of DMD patients and their age-matched healthy controls. Obtained data strongly suggest that elevated level of urinary ferritin is functionally linked to the renal management of myoglobin iron derived from leaky muscles of DMD patients. This first observation of the high level of ferritin in urine of DMD patients permits to consider this protein as a new urinary biomarker in muscular dystrophies and sheds light on the mechanisms of iron metabolism and kidney functioning in DMD.


Ferritins/urine , Iron/metabolism , Muscular Dystrophy, Duchenne/urine , Myoglobin/metabolism , Adolescent , Biomarkers/metabolism , Child , Child, Preschool , Cytokines/urine , Humans , Male , Young Adult
9.
Haematologica ; 103(4): 587-596, 2018 04.
Article En | MEDLINE | ID: mdl-29305416

Gaucher disease (GD) is an inherited deficiency of glucocerebrosidase leading to accumulation of glucosylceramide in tissues such as the spleen, liver, and bone marrow. The resulting lipid-laden macrophages lead to the appearance of "Gaucher cells". Anemia associated with an unexplained hyperferritinemia is a frequent finding in GD, but whether this pathogenesis is related to an iron metabolism disorder has remained unclear. To investigate this issue, we explored the iron status of a large cohort of 90 type I GD patients, including 66 patients treated with enzyme replacement therapy. Ten of the patients treated with enzyme replacement were followed up before and during treatment. Serum levels of hepcidin, the iron regulatory peptide, remained within the physiological range, while the transferrin saturation was slightly decreased in children. Inflammation-independent hyperferritinemia was found in 65% of the patients, and Perl's staining of the spleen and marrow smear revealed iron accumulation in Gaucher cells. Treated patients exhibited reduced hyperferritinemia, increased transferrin saturation and transiently increased systemic hepcidin. In addition, the hepcidin and ferritin correlation was markedly improved, and, in most patients, the hemoglobin level was normalized. To further explore eventual iron sequestration in macrophages, we produce a Gaucher cells model by treating the J774 macrophage cell line with a glucocerebrosidase inhibitor and showed induced local hepcidin and membrane retrieval of the iron exporter, ferroportin. These data reveal the involvement of Gaucher cells in abnormal iron sequestration, which may explain the mechanism of hyperferritinemia in GD patients. Local hepcidin-ferroportin interaction was involved in this pathogenesis.


Gaucher Disease/metabolism , Hepcidins/blood , Iron/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cation Transport Proteins/metabolism , Child , Child, Preschool , Enzyme Replacement Therapy/methods , Female , Ferritins/blood , Humans , Macrophages/metabolism , Male , Mice , Middle Aged , Retrospective Studies , Young Adult
10.
Presse Med ; 46(12 Pt 2): e272-e278, 2017 Dec.
Article En | MEDLINE | ID: mdl-29129410

Although iron is vital, its free form is likely to be involved in oxidation-reduction reactions, leading to the formation of free radicals and oxidative stress. Living organisms have developed protein systems to transport free iron through the cell membranes and biological fluids and store it in a non-toxic and readily mobilizable form to avoid iron toxicity. Hepcidin plays a crucial role in maintaining iron homeostasis. Hepcidin expression is directly regulated by variations in iron intake and its repression leads to an increase in bioavailable serum iron level. However, in pathological situations, prolonged repression often leads to pathological iron overload. In this review, we describe the different molecular mechanisms responsible for the maintenance of iron metabolism and the consequences of iron overload. Indeed, genetic hemochromatosis and post-transfusional siderosis are the two main conditions responsible for iron overload. Long-term iron overload is deleterious, and treatment relies on venesection therapy for genetic hemochromatosis and chelation therapy for iron overload resulting from multiple transfusions.


Hepcidins/physiology , Iron Overload/etiology , Iron/metabolism , Hemochromatosis , Humans
11.
Gastroenterology ; 150(3): 672-683.e4, 2016 Mar.
Article En | MEDLINE | ID: mdl-26582087

BACKGROUND & AIMS: Hereditary hemochromatosis is a heterogeneous group of genetic disorders characterized by parenchymal iron overload. It is caused by defective expression of liver hepcidin, the main regulator of iron homeostasis. Iron stimulates the gene encoding hepcidin (HAMP) via the bone morphogenetic protein (BMP)6 signaling to SMAD. Although several genetic factors have been found to cause late-onset hemochromatosis, many patients have unexplained signs of iron overload. We investigated BMP6 function in these individuals. METHODS: We sequenced the BMP6 gene in 70 consecutive patients with a moderate increase in serum ferritin and liver iron levels who did not carry genetic variants associated with hemochromatosis. We searched for BMP6 mutations in relatives of 5 probands and in 200 healthy individuals (controls), as well as in 2 other independent cohorts of hyperferritinemia patients. We measured serum levels of hepcidin by liquid chromatography-tandem mass spectrometry and analyzed BMP6 in liver biopsy specimens from patients by immunohistochemistry. The functions of mutant and normal BMP6 were assessed in transfected cells using immunofluorescence, real-time quantitative polymerase chain reaction, and immunoblot analyses. RESULTS: We identified 3 heterozygous missense mutations in BMP6 (p.Pro95Ser, p.Leu96Pro, and p.Gln113Glu) in 6 unrelated patients with unexplained iron overload (9% of our cohort). These mutations were detected in less than 1% of controls. p.Leu96Pro also was found in 2 patients from the additional cohorts. Family studies indicated dominant transmission. Serum levels of hepcidin were inappropriately low in patients. A low level of BMP6, compared with controls, was found in a biopsy specimen from 1 patient. In cell lines, the mutated residues in the BMP6 propeptide resulted in defective secretion of BMP6; reduced signaling via SMAD1, SMAD5, and SMAD8; and loss of hepcidin production. CONCLUSIONS: We identified 3 heterozygous missense mutations in BMP6 in patients with unexplained iron overload. These mutations lead to loss of signaling to SMAD proteins and reduced hepcidin production. These mutations might increase susceptibility to mild-to-moderate late-onset iron overload.


Bone Morphogenetic Protein 6/genetics , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hepcidins/biosynthesis , Heterozygote , Iron/metabolism , Liver/metabolism , Mutation, Missense , Aged , Animals , Biopsy , Bone Morphogenetic Protein 6/metabolism , Case-Control Studies , Cell Line , Chromatography, Liquid , DNA Mutational Analysis , Female , Ferritins/blood , Genetic Association Studies , Genetic Predisposition to Disease , Hemochromatosis/blood , Hepcidins/blood , Humans , Immunohistochemistry , Male , Middle Aged , Opossums , Phenotype , Smad Proteins, Receptor-Regulated/metabolism , Tandem Mass Spectrometry , Transfection
12.
J Am Soc Nephrol ; 27(3): 835-46, 2016 Mar.
Article En | MEDLINE | ID: mdl-26293821

The iron-regulatory peptide hepcidin exhibits antimicrobial activity. Having previously shown hepcidin expression in the kidney, we addressed its role in urinary tract infection (UTI), which remains largely unknown. Experimental UTI was induced in wild-type (WT) and hepcidin-knockout (Hepc-/-) mice using the uropathogenic Escherichia coli CFT073 strain. Compared with infected WT mice, infected Hepc-/- mice showed a dramatic increase in renal bacterial load. Moreover, bacterial invasion was significantly dampened by the pretreatment of WT mice with hepcidin. Infected Hepc-/- mice exhibited decreased iron accumulation in the renal medulla and significant attenuation of the renal inflammatory response. Notably, we demonstrated in vitro bacteriostatic activity of hepcidin against CFT073. Furthermore, CFT073 repressed renal hepcidin, both in vivo and in cultured renal cells, and reduced phosphorylation of SMAD kinase in vivo, suggesting a bacterial strategy to escape the antimicrobial activities of hepcidin. In conclusion, we provide new mechanisms by which hepcidin contributes to renal host defense and suggest that targeting hepcidin offers a strategy to prevent bacterial invasion.


Anti-Infective Agents/pharmacology , Escherichia coli Infections/metabolism , Escherichia coli/drug effects , Hepcidins/metabolism , Hepcidins/pharmacology , Urinary Tract Infections/metabolism , Animals , Anti-Infective Agents/metabolism , Bacterial Load/genetics , Cells, Cultured , Colony Count, Microbial , Cytokines/metabolism , Escherichia coli Infections/microbiology , Female , Hepcidins/genetics , Iron/metabolism , Kidney Medulla/cytology , Kidney Medulla/metabolism , Kidney Medulla/microbiology , Mice , Mice, Inbred CBA , Mice, Knockout , Nephritis/metabolism , Nephritis/microbiology , Nephritis/pathology , Neutrophils , Phosphorylation , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Urinary Tract Infections/microbiology
...