Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 104(2): 341-347, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30712775

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Péptidos de Penetración Celular/metabolismo , Células Precursoras Eritroides/metabolismo , Terapia Genética/métodos , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/terapia , Receptores de Transferrina/metabolismo , Antígenos CD/administración & dosificación , Antígenos CD34/metabolismo , Línea Celular , Péptidos de Penetración Celular/administración & dosificación , Eritroblastos/citología , Eritroblastos/metabolismo , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Humanos , Ligandos , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Protoporfirinas/metabolismo , ARN Mensajero , Receptores de Transferrina/administración & dosificación
2.
Blood ; 136(21): 2457-2468, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32678895

RESUMEN

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.


Asunto(s)
Anemia Hemolítica/tratamiento farmacológico , Deferiprona/uso terapéutico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Trastornos por Fotosensibilidad/tratamiento farmacológico , Porfiria Eritropoyética/tratamiento farmacológico , 5-Aminolevulinato Sintetasa/antagonistas & inhibidores , 5-Aminolevulinato Sintetasa/biosíntesis , 5-Aminolevulinato Sintetasa/genética , Adulto , Anemia Hemolítica/etiología , Animales , Sistemas CRISPR-Cas , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Femenino , Técnicas de Sustitución del Gen , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/etiología , Leucemia Eritroblástica Aguda/patología , Ratones , Células Madre de Sangre Periférica/efectos de los fármacos , Células Madre de Sangre Periférica/metabolismo , Trastornos por Fotosensibilidad/etiología , Porfiria Intermitente Aguda/metabolismo , Porfiria Eritropoyética/complicaciones , Porfirinas/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/farmacología
3.
Ann Hematol ; 101(12): 2633-2643, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36195681

RESUMEN

In low-risk myelodysplastic syndrome (LR-MDS), erythropoietin (EPO) is widely used for the treatment of chronic anemia. However, initial response to EPO has time-limited effects. Luspatercept reduces red blood cell transfusion dependence in LR-MDS patients. Here, we investigated the molecular action of luspatercept (RAP-536) in an in vitro model of erythroid differentiation of MDS, and also in a in vivo PDX murine model with primary samples of MDS patients carrying or not SF3B1 mutation. In our in vitro model, RAP-536 promotes erythroid proliferation by increasing the number of cycling cells without any impact on apoptosis rates. RAP-536 promoted late erythroid precursor maturation while decreasing intracellular reactive oxygen species level. RNA sequencing of erythroid progenitors obtained under RAP-536 treatment showed an enrichment of genes implicated in positive regulation of response to oxidative stress and erythroid differentiation. In our PDX model, RAP-536 induces a higher hemoglobin level. RAP-536 did not modify variant allele frequencies in vitro and did not have any effect against leukemic burden in our PDX model. These results suggest that RAP-536 promotes in vivo and in vitro erythroid cell differentiation by decreasing ROS level without any remarkable impact on iron homeostasis and on mutated allele burden.


Asunto(s)
Síndromes Mielodisplásicos , Humanos , Ratones , Animales , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Mutación , Estrés Oxidativo
4.
Blood ; 133(12): 1358-1370, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30700418

RESUMEN

Diamond-Blackfan anemia (DBA) is a congenital erythroblastopenia that is characterized by a blockade in erythroid differentiation related to impaired ribosome biogenesis. DBA phenotype and genotype are highly heterogeneous. We have previously identified 2 in vitro erythroid cell growth phenotypes for primary CD34+ cells from DBA patients and following short hairpin RNA knockdown of RPS19, RPL5, and RPL11 expression in normal human CD34+ cells. The haploinsufficient RPS19 in vitro phenotype is less severe than that of 2 other ribosomal protein (RP) mutant genes. We further documented that proteasomal degradation of HSP70, the chaperone of GATA1, is a major contributor to the defect in erythroid proliferation, delayed erythroid differentiation, increased apoptosis, and decreased globin expression, which are all features of the RPL5 or RPL11 DBA phenotype. In the present study, we explored the hypothesis that an imbalance between globin and heme synthesis may be involved in pure red cell aplasia of DBA. We identified disequilibrium between the globin chain and the heme synthesis in erythroid cells of DBA patients. This imbalance led to accumulation of excess free heme and increased reactive oxygen species production that was more pronounced in cells of the RPL5 or RPL11 phenotype. Strikingly, rescue experiments with wild-type HSP70 restored GATA1 expression levels, increased globin synthesis thereby reducing free heme excess and resulting in decreased apoptosis of DBA erythroid cells. These results demonstrate the involvement of heme in DBA pathophysiology and a major role of HSP70 in the control of balanced heme/globin synthesis.


Asunto(s)
Anemia de Diamond-Blackfan/patología , Diferenciación Celular , Células Eritroides/patología , Factor de Transcripción GATA1/metabolismo , Globinas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Hemo/metabolismo , Anemia de Diamond-Blackfan/metabolismo , Proliferación Celular , Células Cultivadas , Células Eritroides/metabolismo , Femenino , Estudios de Seguimiento , Haploinsuficiencia , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Fenotipo , Pronóstico , ARN Interferente Pequeño , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
5.
Hum Mol Genet ; 27(7): 1164-1173, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29360981

RESUMEN

Acute intermittent porphyria (AIP) is a disease affecting the heme biosynthesis pathway caused by mutations of the hydroxymethylbilane synthase (HMBS) gene. AIP is thought to display autosomal dominant inheritance with incomplete penetrance. We evaluated the prevalence, penetrance and heritability of AIP, in families with the disease from the French reference center for porphyria (CFP) (602 overt patients; 1968 relatives) and the general population, using Exome Variant Server (EVS; 12 990 alleles) data. The pathogenicity of the 42 missense variants identified was assessed in silico, and in vitro, by measuring residual HMBS activity of the recombinant protein. The minimal estimated prevalence of AIP in the general population was 1/1299. Thus, 50 000 subjects would be expected to carry the AIP genetic trait in France. Penetrance was estimated at 22.9% in families with AIP, but at only 0.5-1% in the general population. Intrafamily correlation studies showed correlations to be strong overall and modulated by kinship and the area in which the person was living, demonstrating strong influences of genetic and environmental modifiers on inheritance. Null alleles were associated with a more severe phenotype and a higher penetrance than for other mutant alleles. In conclusion, the striking difference in the penetrance of HMBS mutations between the general population and the French AIP families suggests that AIP inheritance does not follow the classical autosomal dominant model, instead of being modulated by strong environmental and genetic factors independent from HMBS. An oligogenic inheritance model with environmental modifiers might better explain AIP penetrance and heritability.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Interacción Gen-Ambiente , Hidroximetilbilano Sintasa/genética , Mutación Missense , Penetrancia , Porfiria Intermitente Aguda/genética , Femenino , Francia/epidemiología , Humanos , Masculino , Porfiria Intermitente Aguda/enzimología , Porfiria Intermitente Aguda/epidemiología , Prevalencia
6.
Proc Natl Acad Sci U S A ; 114(38): E8045-E8052, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874591

RESUMEN

Loss-of-function mutations in genes for heme biosynthetic enzymes can give rise to congenital porphyrias, eight forms of which have been described. The genetic penetrance of the porphyrias is clinically variable, underscoring the role of additional causative, contributing, and modifier genes. We previously discovered that the mitochondrial AAA+ unfoldase ClpX promotes heme biosynthesis by activation of δ-aminolevulinate synthase (ALAS), which catalyzes the first step of heme synthesis. CLPX has also been reported to mediate heme-induced turnover of ALAS. Here we report a dominant mutation in the ATPase active site of human CLPX, p.Gly298Asp, that results in pathological accumulation of the heme biosynthesis intermediate protoporphyrin IX (PPIX). Amassing of PPIX in erythroid cells promotes erythropoietic protoporphyria (EPP) in the affected family. The mutation in CLPX inactivates its ATPase activity, resulting in coassembly of mutant and WT protomers to form an enzyme with reduced activity. The presence of low-activity CLPX increases the posttranslational stability of ALAS, causing increased ALAS protein and ALA levels, leading to abnormal accumulation of PPIX. Our results thus identify an additional molecular mechanism underlying the development of EPP and further our understanding of the multiple mechanisms by which CLPX controls heme metabolism.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Endopeptidasa Clp , Mutación Missense , Porfiria Eritropoyética , Protoporfirinas/biosíntesis , 5-Aminolevulinato Sintetasa/genética , Adolescente , Sustitución de Aminoácidos , Endopeptidasa Clp/genética , Endopeptidasa Clp/metabolismo , Estabilidad de Enzimas/genética , Femenino , Humanos , Masculino , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/metabolismo , Porfiria Eritropoyética/patología , Protoporfirinas/genética
7.
Biochem Biophys Res Commun ; 520(2): 297-303, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31601421

RESUMEN

Clinical severity is heterogeneous among patients suffering from congenital erythropoietic porphyria (CEP) suggesting a modulation of the disease (UROS deficiency) by environmental factors and modifier genes. A KI model of CEP due to a missense mutation of UROS gene present in human has been developed on 3 congenic mouse strains (BALB/c, C57BL/6, and 129/Sv) in order to study the impact of genetic background on disease severity. To detect putative modifiers of disease expression in congenic mice, hematologic data, iron parameters, porphyrin content and tissue samples were collected. Regenerative hemolytic anemia, a consequence of porphyrin excess in RBCs, had various expressions: 129/Sv mice were more hemolytic, BALB/c had more regenerative response to anemia, C57BL/6 were less affected. Iron status and hemolysis level were directly related: C57BL/6 and BALB/c had moderate hemolysis and active erythropoiesis able to reduce iron overload in the liver, while, 129/Sv showed an imbalance between iron release due to hemolysis and erythroid use. The negative control of hepcidin on the ferroportin iron exporter appeared strain specific in the CEP mice models tested. Full repression of hepcidin was observed in BALB/c and 129/Sv mice, favoring parenchymal iron overload in the liver. Unchanged hepcidin levels in C57BL/6 resulted in retention of iron predominantly in reticuloendothelial tissues. These findings open the field for potential therapeutic applications in the human disease, of hepcidin agonists and iron depletion in chronic hemolytic anemia.


Asunto(s)
Hepcidinas/metabolismo , Hierro/metabolismo , Porfiria Eritropoyética/genética , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Femenino , Hemólisis , Hepcidinas/genética , Sobrecarga de Hierro/genética , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Porfiria Eritropoyética/etiología , Porfiria Eritropoyética/metabolismo , Porfirinas/metabolismo , Uroporfirinógeno III Sintetasa/genética
8.
Mol Genet Metab ; 128(3): 236-241, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30413387

RESUMEN

Porphyrias are inherited diseases with low penetrance affecting the heme biosynthesis pathway. Acute intermittent porphyria (AIP), variegate porphyria (VP) and hereditary coproporphyria (HCP) together constitute the acute hepatic porphyrias (AHP). These diseases have been identified as risk factors for primary liver cancers (PLC), mainly hepatocellular carcinoma (HCC: range 87-100%) but also cholangiocarcinoma, alone or combination with HCC. In AHP, HCC annual incidence rates range from 0.16 to 0.35% according to the populations studied. Annual incidence rates are higher in Swedish and Norwegian patients, due to a founder effect. It increases above age 50. The pathophysiology could include both direct toxic effects of heme precursors, particularly δ-aminolevulinic acid (ALA), compound heterozygosity for genes implied in heme biosynthesis pathway or the loss of oxidative stress homeostasis due to a relative lack of heme. The high HCC incidence justifies radiological surveillance in AHP patients above age 50. Efforts are made to find new biological non-invasive markers. In this respect, we describe here the first report of PIVKA-II clinical utility in the follow-up of an AIP patient that develop an HCC. In this manuscript we reviewed the epidemiology, the physiopathology, and the screening strategy of HCC in AHP.


Asunto(s)
Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/fisiopatología , Neoplasias Hepáticas/etiología , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/complicaciones , Biomarcadores , Femenino , Hemo/biosíntesis , Humanos , Incidencia , Neoplasias Hepáticas/fisiopatología , Masculino , Persona de Mediana Edad , Noruega/epidemiología , Porfiria Intermitente Aguda/complicaciones , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/epidemiología , Factores de Riesgo , Suecia/epidemiología
9.
Mol Genet Metab ; 128(3): 190-197, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30737140

RESUMEN

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.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Ácido Aminolevulínico/metabolismo , Anemia Sideroblástica/genética , Regulación de la Expresión Génica , Porfirias/genética , 5-Aminolevulinato Sintetasa/metabolismo , Anemia Sideroblástica/enzimología , Animales , Sitios de Unión , Factor de Transcripción GATA1/genética , Hemo/biosíntesis , Humanos , Hígado/metabolismo , Ratones , Mutación Missense , Porfirias/enzimología , Regiones Promotoras Genéticas
10.
Mol Genet Metab ; 128(3): 342-351, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30660387

RESUMEN

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.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/genética , Ferroquelatasa/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Glutarredoxinas/genética , Hemo/biosíntesis , Mutación Missense , 5-Aminolevulinato Sintetasa/metabolismo , Aconitato Hidratasa/metabolismo , Adolescente , Secuencia de Aminoácidos , Anemia Sideroblástica/enzimología , Línea Celular Transformada , Femenino , Ferroquelatasa/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Glutatión/metabolismo , Humanos , Mitocondrias/enzimología , Estrés Oxidativo , Linaje , Estructura Terciaria de Proteína
11.
Haematologica ; 104(3): 497-504, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30287621

RESUMEN

Erythropoiesis-stimulating agents are generally the first line of treatment of anemia in patients with lower-risk myelodysplastic syndrome. We prospectively investigated the predictive value of somatic mutations, and biomarkers of ineffective erythropoiesis including the flow cytometry RED score, serum growth-differentiation factor-15, and hepcidin levels. Inclusion criteria were no prior treatment with erythropoiesis-stimulating agents, low- or intermediate-1-risk myelodysplastic syndrome according to the International Prognostic Scoring System, and a hemoglobin level <10 g/dL. Patients could be red blood cell transfusion-dependent or not and were given epoetin zeta 40 000 IU/week. Serum erythropoietin level, iron parameters, hepcidin, flow cytometry Ogata and RED scores, and growth-differentiation factor-15 levels were determined at baseline, and molecular analysis by next-generation sequencing was also conducted. Erythroid response (defined according to the International Working Group 2006 criteria) was assessed at week 12. Seventy patients, with a median age of 78 years, were included in the study. There were 22 patients with refractory cytopenia with multilineage dysplasia, 19 with refractory cytopenia with unilineage dysplasia, 14 with refractory anemia with ring sideroblasts, four with refractory anemia with excess blasts-1, six with chronic myelomonocytic leukemia, two with del5q-and three with unclassifiable myelodysplastic syndrome. According to the revised International Prognostic Scoring System, 13 had very low risk, 47 had low risk, nine intermediate risk and one had high-risk disease. Twenty patients were transfusion dependent. Forty-eight percent had an erythroid response and the median duration of the response was 26 months. At baseline, non-responders had significantly higher RED scores and lower hepcidin:ferritin ratios. In multivariate analysis, only a RED score >4 (P=0.05) and a hepcidin:ferritin ratio <9 (P=0.02) were statistically significantly associated with worse erythroid response. The median response duration was shorter in patients with growth-differentiation factor-15 >2000 pg/mL and a hepcidin:ferritin ratio <9 (P=0.0008 and P=0.01, respectively). In multivariate analysis, both variables were associated with shorter response duration. Erythroid response to epoetin zeta was similar to that obtained with other erythropoiesis-stimulating agents and was correlated with higher baseline hepcidin:ferritin ratio and lower RED score. ClinicalTrials.gov registration: NCT 03598582.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Eritropoyetina/uso terapéutico , Ferritinas/sangre , Hepcidinas/sangre , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Biomarcadores , Eritropoyetina/administración & dosificación , Eritropoyetina/efectos adversos , Femenino , Citometría de Flujo , Humanos , Hierro/metabolismo , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/etiología , Pronóstico , Curva ROC , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
12.
Nephrology (Carlton) ; 24(7): 751-757, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30175513

RESUMEN

AIM: Relatively few haemodialysis (HD) patients remain independent of recombinant human erythropoietin ('rHU-EPO free patients'). We investigated the role of EPO and hepcidin, two key hormones involved in anaemia. METHODS: We report a monocentric case-control series. Iron status, EPO and hepcidin levels were analysed in 15 Adult HD (Age > 18 years) with a stable haemoglobin (Hb) level that have not received rHU-EPO for at least 6 months (=rHU-EPO free patients); and in 60 controls with a stable rHU-EPO dose and Hb level. RESULTS: The rHU-EPO free patients had a higher Hb level compared to controls (12.1 ± 0.99 g/dL vs 11.1 ± 0.73, P = 0.0014), and a lower ferritin level (183 ± 102 vs 312 ± 166 ng/mL, P = 0.001). Hepcidin levels were lower in the rHU-EPO free patients (12.53 ± 10.46 ng/mL) compared to the controls (37.95 ± 34.33 ng/mL), P = 0.0033. Hepcidin levels correlated significantly with ferritin levels; but neither with transferrin saturation, C-reactive protein nor EPO levels. Unsupervised analysis revealed that rHU-EPO free patients had a specific clinical/biological profile (presence of renal cyst, longer dialysis vintage, lower ferritin, and EPO and hepcidin levels compared to the control group). Finally, we showed that a lower ferritin level might be a surrogate marker of a lower hepcidin status in this population. CONCLUSION: Recombinant human erythropoietin free patients seem to restore the EPO-hepcidin axis that is critical for erythropoiesis. A specific combination of clinical and biological parameters may help to detect future rHU-EPO free patients.


Asunto(s)
Anemia/tratamiento farmacológico , Eritropoyetina/fisiología , Hepcidinas/fisiología , Diálisis Renal , Insuficiencia Renal Crónica/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Anemia/etiología , Eritropoyetina/uso terapéutico , Femenino , Ferritinas/sangre , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , Insuficiencia Renal Crónica/sangre
13.
J Biol Chem ; 292(39): 16109-16121, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28768767

RESUMEN

The chloroquine resistance transporter of the human malaria parasite Plasmodium falciparum, PfCRT, is an important determinant of resistance to several quinoline and quinoline-like antimalarial drugs. PfCRT also plays an essential role in the physiology of the parasite during development inside erythrocytes. However, the function of this transporter besides its role in drug resistance is still unclear. Using electrophysiological and flux experiments conducted on PfCRT-expressing Xenopus laevis oocytes, we show here that both wild-type PfCRT and a PfCRT variant associated with chloroquine resistance transport both ferrous and ferric iron, albeit with different kinetics. In particular, we found that the ability to transport ferrous iron is reduced by the specific polymorphisms acquired by the PfCRT variant as a result of chloroquine selection. We further show that iron and chloroquine transport via PfCRT is electrogenic. If these findings in the Xenopus model extend to P. falciparum in vivo, our data suggest that PfCRT might play a role in iron homeostasis, which is essential for the parasite's development in erythrocytes.


Asunto(s)
Antimaláricos/metabolismo , Cloroquina/metabolismo , Hierro/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Sustitución de Aminoácidos , Animales , Transporte Biológico , Hierro/química , Cinética , Proteínas de Transporte de Membrana/genética , Mutación , Oocitos/metabolismo , Oxidación-Reducción , Técnicas de Placa-Clamp , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
14.
Glia ; 66(8): 1709-1723, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29624734

RESUMEN

Neuroinflammation and iron accumulation are hallmarks of a variety of adult neurodegenerative diseases. In Sanfilippo syndrome (mucopolysaccharidosis type III, MPSIII, a pediatric neurodegenerative disease that shares some features with adult neurodegenerative diseases), the progressive accumulation of heparan sulfate oligosaccharides (HSOs) induces microglia and astrocytes to produce pro-inflammatory cytokines leading to severe neuroinflammation. The objectives of the present study were (1) to measure the local iron concentration and to assess iron metabolism in the brain of a MPSIIIB murine model and (2) to identify the brain cells involved in this accumulation. We found that iron accumulation in MPSIIIB mice primarily affected the cerebral cortex where hepcidin levels were higher than in wild-type mice, and increased with aging. This increase was correlated with low expression of ferroportin 1 (FPN1), and thus brain iron retention. Moreover, we showed in vitro that HSOs are directly responsible for the production of hepcidin and the relative decrease in FPN1 expression when added to cultures of microglia and, to a lesser extent, to cultures of astrocytes. In contrast, no significant differences were observed in neurons. Hepcidin induction results from activation of the TLR4 pathway and STAT3 signaling, and leads to iron retention within microglia. Our results show that microglia have a key role in cerebral hepcidin overexpression and thus in the brain iron accumulation observed in the MPSIIIB model.


Asunto(s)
Encéfalo/metabolismo , Hierro/metabolismo , Microglía/metabolismo , Mucopolisacaridosis III/metabolismo , Animales , Astrocitos/metabolismo , Ratones Noqueados , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo
15.
Haematologica ; 103(4): 587-596, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29305416

RESUMEN

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.


Asunto(s)
Enfermedad de Gaucher/metabolismo , Hepcidinas/sangre , Hierro/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteínas de Transporte de Catión/metabolismo , Niño , Preescolar , Terapia de Reemplazo Enzimático/métodos , Femenino , Ferritinas/sangre , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
16.
J Am Soc Nephrol ; 28(6): 1924-1932, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28031405

RESUMEN

CKD occurs in most patients with acute intermittent porphyria (AIP). During AIP, δ-aminolevulinic acid (ALA) accumulates and promotes tubular cell death and tubulointerstitial damage. The human peptide transporter 2 (PEPT2) expressed by proximal tubular cells mediates the reabsorption of ALA, and variants of PEPT2 have different affinities for ALA. We tested the hypothesis that PEPT2 genotypes affect the severity and prognosis of porphyria-associated kidney disease. We analyzed data from 122 individuals with AIP who were followed from 2003 to 2013 and genotyped for PEPT2 At last follow-up, carriers of the PEPT2*1*1 genotype (higher affinity variant) exhibited worse renal function than carriers of the lower affinity variants PEPT2*1/*2 and PEPT2*2/*2 (mean±SD eGFR: 54.4±19.1, 66.6±23.8, and 78.1±19.9 ml/min per 1.73 m2, respectively). Change in eGFR (mean±SD) over the 10-year period was -11.0±3.3, -2.4±1.9, and 3.4±2.6 ml/min per 1.73 m2 for PEPT2*1/*1, PEPT2*1*2, and PEPT*2*2*2 carriers, respectively. At the end of follow-up, 68% of PEPT2*1*1 carriers had an eGFR<60 ml/min per 1.73 m2, compared with 37% of PEPT2*1*2 carriers and 15% of PEPT2*2*2 carriers. Multiple regression models including all confounders indicated that the PEPT2*1*1 genotype independently associated with an eGFR<60 ml/min per 1.73 m2 (odds ratio, 6.85; 95% confidence interval, 1.34 to 46.20) and an annual decrease in eGFR of >1 ml/min per 1.73 m2 (odds ratio, 3.64; 95% confidence interval, 1.37 to 9.91). Thus, a gene variant is predictive of the severity of a chronic complication of AIP. The therapeutic value of PEPT2 inhibitors in preventing porphyria-associated kidney disease warrants investigation.


Asunto(s)
Porfirias/complicaciones , Porfirias/genética , Insuficiencia Renal Crónica/genética , Simportadores/genética , Enfermedad Aguda , Anciano , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Índice de Severidad de la Enfermedad
17.
Hum Mol Genet ; 24(17): 5015-23, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071363

RESUMEN

Acute intermittent porphyria (AIP), an autosomal dominant metabolic disease (MIM #176000), is due to a deficiency of hydroxymethylbilane synthase (HMBS), which catalyzes the third step of the heme biosynthetic pathway. The clinical expression of the disease is mainly neurological, involving the autonomous, central and peripheral nervous systems. We explored mitochondrial oxidative phosphorylation (OXPHOS) in the brain and skeletal muscle of the Hmbs(-/-) mouse model first in the basal state (BS), and then after induction of the disease with phenobarbital and treatment with heme arginate (HA). The modification of the respiratory parameters, determined in mice in the BS, reflected a spontaneous metabolic energetic adaptation to HMBS deficiency. Phenobarbital induced a sharp alteration of the oxidative metabolism with a significant decrease of ATP production in skeletal muscle that was restored by treatment with HA. This OXPHOS defect was due to deficiencies in complexes I and II in the skeletal muscle whereas all four respiratory chain complexes were affected in the brain. To date, the pathogenesis of AIP has been mainly attributed to the neurotoxicity of aminolevulinic acid and heme deficiency. Our results show that mitochondrial energetic failure also plays an important role in the expression of the disease.


Asunto(s)
Encéfalo/metabolismo , Hidroximetilbilano Sintasa/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Músculos/metabolismo , Porfiria Intermitente Aguda/genética , Porfiria Intermitente Aguda/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos , Músculos/efectos de los fármacos , Fenobarbital/farmacología
18.
Am J Hum Genet ; 94(4): 611-7, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24680888

RESUMEN

In 90% of people with erythropoietic protoporphyria (EPP), the disease results from the inheritance of a common hypomorphic FECH allele, encoding ferrochelatase, in trans to a private deleterious FECH mutation. The activity of the resulting FECH enzyme falls below the critical threshold of 35%, leading to the accumulation of free protoporphyrin IX (PPIX) in bone marrow erythroblasts and in red cells. The mechanism of low expression involves a biallelic polymorphism (c.315-48T>C) localized in intron 3. The 315-48C allele increases usage of the 3' cryptic splice site between exons 3 and 4, resulting in the transcription of an unstable mRNA with a premature stop codon, reducing the abundance of wild-type FECH mRNA, and finally reducing FECH activity. Through a candidate-sequence approach and an antisense-oligonucleotide-tiling method, we identified a sequence that, when targeted by an antisense oligonucleotide (ASO-V1), prevented usage of the cryptic splice site. In lymphoblastoid cell lines derived from symptomatic EPP subjects, transfection of ASO-V1 reduced the usage of the cryptic splice site and efficiently redirected the splicing of intron 3 toward the physiological acceptor site, thereby increasing the amount of functional FECH mRNA. Moreover, the administration of ASO-V1 into developing human erythroblasts from an overtly EPP subject markedly increased the production of WT FECH mRNA and reduced the accumulation of PPIX to a level similar to that measured in asymptomatic EPP subjects. Thus, EPP is a paradigmatic Mendelian disease in which the in vivo correction of a common single splicing defect would improve the condition of most affected individuals.


Asunto(s)
Ferroquelatasa/genética , Oligonucleótidos Antisentido/uso terapéutico , Protoporfiria Eritropoyética/terapia , Línea Celular , Femenino , Humanos , Masculino , Linaje , Polimorfismo Genético , Protoporfirinas/metabolismo , Empalme del ARN , ARN Mensajero/genética
19.
Gastroenterology ; 150(3): 672-683.e4, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26582087

RESUMEN

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.


Asunto(s)
Proteína Morfogenética Ósea 6/genética , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hepcidinas/biosíntesis , Heterocigoto , Hierro/metabolismo , Hígado/metabolismo , Mutación Missense , Anciano , Animales , Biopsia , Proteína Morfogenética Ósea 6/metabolismo , Estudios de Casos y Controles , Línea Celular , Cromatografía Liquida , Análisis Mutacional de ADN , Femenino , Ferritinas/sangre , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Hemocromatosis/sangre , Hepcidinas/sangre , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Zarigüeyas , Fenotipo , Proteínas Smad Reguladas por Receptores/metabolismo , Espectrometría de Masas en Tándem , Transfección
20.
Haematologica ; 102(2): 260-270, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28143953

RESUMEN

Hemolysis occurring in hematologic diseases is often associated with an iron loading anemia. This iron overload is the result of a massive outflow of hemoglobin into the bloodstream, but the mechanism of hemoglobin handling has not been fully elucidated. Here, in a congenital erythropoietic porphyria mouse model, we evaluate the impact of hemolysis and regenerative anemia on hepcidin synthesis and iron metabolism. Hemolysis was confirmed by a complete drop in haptoglobin, hemopexin and increased plasma lactate dehydrogenase, an increased red blood cell distribution width and osmotic fragility, a reduced half-life of red blood cells, and increased expression of heme oxygenase 1. The erythropoiesis-induced Fam132b was increased, hepcidin mRNA repressed, and transepithelial iron transport in isolated duodenal loops increased. Iron was mostly accumulated in liver and spleen macrophages but transferrin saturation remained within the normal range. The expression levels of hemoglobin-haptoglobin receptor CD163 and hemopexin receptor CD91 were drastically reduced in both liver and spleen, resulting in heme- and hemoglobin-derived iron elimination in urine. In the kidney, the megalin/cubilin endocytic complex, heme oxygenase 1 and the iron exporter ferroportin were induced, which is reminiscent of significant renal handling of hemoglobin-derived iron. Our results highlight ironbound hemoglobin urinary clearance mechanism and strongly suggest that, in addition to the sequestration of iron in macrophages, kidney may play a major role in protecting hepatocytes from iron overload in chronic hemolysis.


Asunto(s)
Anemia Hemolítica/metabolismo , Hepatocitos/metabolismo , Hepcidinas/metabolismo , Hierro/metabolismo , Anemia Hemolítica/sangre , Anemia Hemolítica/complicaciones , Anemia Hemolítica/genética , Animales , Apoptosis , Transporte Biológico , Biomarcadores , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Eritropoyesis , Expresión Génica , Hemo/metabolismo , Hepcidinas/sangre , Hepcidinas/genética , Humanos , Hierro/orina , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Macrófagos , Ratones , Ratones Noqueados , Ratones Transgénicos , Bazo/fisiología , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA