Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Dev Cell ; 58(20): 2112-2127.e4, 2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37586368

RESUMEN

Controlled release of promoter-proximal paused RNA polymerase II (RNA Pol II) is crucial for gene regulation. However, studying RNA Pol II pausing is challenging, as pause-release factors are almost all essential. In this study, we identified heterozygous loss-of-function mutations in SUPT5H, which encodes SPT5, in individuals with ß-thalassemia. During erythropoiesis in healthy human cells, cell cycle genes were highly paused as cells transition from progenitors to precursors. When the pathogenic mutations were recapitulated by SUPT5H editing, RNA Pol II pause release was globally disrupted, and as cells began transitioning from progenitors to precursors, differentiation was delayed, accompanied by a transient lag in erythroid-specific gene expression and cell cycle kinetics. Despite this delay, cells terminally differentiate, and cell cycle phase distributions normalize. Therefore, hindering pause release perturbs proliferation and differentiation dynamics at a key transition during erythropoiesis, identifying a role for RNA Pol II pausing in temporally coordinating the cell cycle and erythroid differentiation.


Asunto(s)
Regulación de la Expresión Génica , ARN Polimerasa II , Humanos , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Diferenciación Celular , Ciclo Celular , Transcripción Genética , Proteínas Nucleares/metabolismo , Factores de Elongación Transcripcional/genética
2.
medRxiv ; 2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36945604

RESUMEN

The controlled release of promoter-proximal paused RNA polymerase II (Pol II) into productive elongation is a major step in gene regulation. However, functional analysis of Pol II pausing is difficult because factors that regulate pause release are almost all essential. In this study, we identified heterozygous loss-of-function mutations in SUPT5H , which encodes SPT5, in individuals with ß-thalassemia unlinked to HBB mutations. During erythropoiesis in healthy human cells, cell cycle genes were highly paused at the transition from progenitors to precursors. When the pathogenic mutations were recapitulated by SUPT5H editing, Pol II pause release was globally disrupted, and the transition from progenitors to precursors was delayed, marked by a transient lag in erythroid-specific gene expression and cell cycle kinetics. Despite this delay, cells terminally differentiate, and cell cycle phase distributions normalize. Therefore, hindering pause release perturbs proliferation and differentiation dynamics at a key transition during erythropoiesis, revealing a role for Pol II pausing in the temporal coordination between the cell cycle and differentiation.

3.
Metabolites ; 12(10)2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36295822

RESUMEN

Mutations in the ferroportin (FPN) gene SLC40A1 alter iron recycling and cause disturbances in iron homeostasis. The variants of TMPRSS6 contribute to the development of iron deficiencies. In this study, we determined the role of FPN and TMPRSS6 gene polymorphisms in the modulation of iron homeostasis based on biochemical parameters. PCR analysis and sequencing were performed to determine the single nucleotide polymorphisms (SNPs) SLC40A1 c.44−24G>C (rs1439816), SLC40A1 c.663T>C (rs2304704), and TMPRSS6 c.2207T>C (rs855791). Hemoglobin concentration and iron status were determined by standard procedures. We studied 79 iron-loaded individuals for SLC40A1 polymorphisms. Interestingly, 35/79 individuals with SLC40A1 SNPs also carried a TMPRSS6 c.2207T>C polymorphism. The biochemical values of the iron overloaded individuals were compared to those of the individuals carrying TMPRSS6 SNPs and the healthy individuals (wild-type group). The ferritin concentration, transferrin saturation % (TS%), and hemoglobin concentration were significantly higher in the participants with FPN SNPs than in the other three groups. The ferritin concentration and TS% were higher in participants with both SLC40A1 and TMPRSS6 SNPs than in the TMPRSS6 and wild-type groups, while hemoglobin concentration was significantly higher than that in the TMPRSS6 SNP group only. The participants with TMPRSS6 SNPs had significantly lower ferritin concentration, TS%, and hemoglobin concentration than all the other groups. SLC40A1 and TMPRSS6 SNPs might act in the opposite direction, preventing the development of severe iron overload, and the modulation of the iron status by TMPRSS6 SNPs might provide protection.

4.
Front Physiol ; 13: 886194, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35923227

RESUMEN

Background: The heme biosynthesis (HB) involves eight subsequent enzymatic steps. Erythropoietic protoporphyria (EPP) is caused by loss-of-function mutations in the ferrochelatase (FECH) gene, which in the last HB step inserts ferrous iron into protoporphyrin IX (PPIX) to form heme. Aim and method: The aim of this work was to for the first time analyze the mRNA expression of all HB genes in peripheral blood samples of patients with EPP having the same genotype FECH c.[215dupT]; [315-48T > C] as compared to healthy controls by highly sensitive and specific digital PCR assays (dPCR). Results: We confirmed a decreased FECH mRNA expression in patients with EPP. Further, we found increased ALAS2 and decreased ALAS1, CPOX, PPOX and HMBS mRNA expression in patients with EPP compared to healthy controls. ALAS2 correlated with FECH mRNA expression (EPP: r = 0.63, p = 0.03 and controls: r = 0.68, p = 0.02) and blood parameters like PPIX (EPP: r = 0.58 p = 0.06). Conclusion: Our method is the first that accurately quantifies HB mRNA from blood samples with potential applications in the monitoring of treatment effects of mRNA modifying therapies in vivo, or investigation of the HB pathway and its regulation. However, our findings should be studied in separated blood cell fractions and on the enzymatic level.

5.
Front Physiol ; 13: 841050, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309058

RESUMEN

Partial deficiency of the last enzyme of the heme biosynthetic pathway, namely, ferrochelatase (FECH), is responsible for erythropoietic protoporphyria (EPP) in humans. This disorder is characterized by painful skin photosensitivity, due to excessive protoporphyrin IX (PPIX) production in erythrocytes. Although several papers report the presence of iron deficiency anemia in about 50% of EPP patients, there is still no a conclusive explanation of the why this occurs. In the present work, we explored hematological indices and iron status in 20 unrelated Italian EPP patients in order to propose a new hypothesis. Our data show that microcytosis is present in EPP patients also in the absence of anemia and iron deficiency with a link between PPIX accumulation and reduced MCV, probably indicating an indirect condition of heme deficiency. Patients studied had a downward shift of iron parameters due to increased hepcidin concentrations only in a state of repleted iron stores. Interestingly, hemoglobin synthesis was not limited by iron supply except in cases with further iron loss, in which concomitantly increased soluble transferrin (Tf) receptor (sTfR) levels were detected. The mechanisms involved in the iron uptake downregulation in EPP remain unclear, and the role of PPIX accumulation in microcytosis.

6.
Front Physiol ; 12: 684569, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093240

RESUMEN

Congenital hemolytic anemias (CHAs) are heterogeneous and rare disorders caused by alterations in structure, membrane transport, metabolism, or red blood cell production. The pathophysiology of these diseases, in particular the rarest, is often poorly understood, and easy-to-apply tools for diagnosis, clinical management, and patient stratification are still lacking. We report the 3-years monocentric experience with a 43 genes targeted Next Generation Sequencing (t-NGS) panel in diagnosis of CHAs; 122 patients from 105 unrelated families were investigated and the results compared with conventional laboratory pathway. Patients were divided in two groups: 1) cases diagnosed with hematologic investigations to be confirmed at molecular level, and 2) patients with unexplained anemia after extensive hematologic investigation. The overall sensitivity of t-NGS was 74 and 35% for families of groups 1 and 2, respectively. Inside this cohort of patients we identified 26 new pathogenic variants confirmed by functional evidence. The implementation of laboratory work-up with t-NGS increased the number of diagnoses in cases with unexplained anemia; cytoskeleton defects are well detected by conventional tools, deserving t-NGS to atypical cases; the diagnosis of Gardos channelopathy, some enzyme deficiencies, familial siterosterolemia, X-linked defects in females and other rare and ultra-rare diseases definitely benefits of t-NGS approaches.

7.
Front Immunol ; 12: 615620, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33664746

RESUMEN

The homeostasis of tissues in a chronic disease is an essential function of the alternative pathway (AP) of the complement system (CS). However, if not controlled, it may also be detrimental to healthy cells with a consequent aggravation of symptoms. The protoporphyria (PP) is a rare chronic disease that causes phototoxicity in visible light with local skin pain and general malaise. In order to establish if there is a systemic involvement of the CS during sun exposure, we designed a non-invasive method with a serum collection in winter and summer from 19 PP and 13 controls to detect the levels of CS protein: Properdin, Factor H (FH), and C5. Moreover, the global radiation data were collected from the regional agency of environmental protection (ARPA). The results show growing values for every protein in patients with PP, compared to control, in both seasons, in particular in summer compared to winter. To reinforce the evidence, we have estimated the personal exposure of patients based on the global radiation data. The main factors of the AP increased over the season, confirming the involvement of the AP in relation to light exposure. The systemic response could justify the general malaise of patients after long light exposure and can be exploited to elucidate new therapeutic approaches.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Vía Alternativa del Complemento/efectos de la radiación , Proteínas del Sistema Complemento/inmunología , Susceptibilidad a Enfermedades , Protoporfiria Eritropoyética/etiología , Luz Solar/efectos adversos , Adulto , Biomarcadores , Complemento C5/inmunología , Complemento C5/metabolismo , Factor H de Complemento/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Properdina/inmunología , Properdina/metabolismo , Protoporfiria Eritropoyética/diagnóstico , Protoporfiria Eritropoyética/metabolismo , Estaciones del Año
8.
Mediterr J Hematol Infect Dis ; 12(1): e2020075, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194149

RESUMEN

ß-thalassemia is a hereditary disorder caused by defective production of ß-globin chains of hemoglobin (Hb) that leads to an increased α/ß globins ratio with subsequent free α-globins. Alpha globin excess causes oxidative stress, red blood cells membrane damage, premature death of late-stage erythroid precursors, resulting in ineffective erythropoiesis. The transforming growth factor ß (TGF-ß) superfamily signaling acts on biological processes, such as cell quiescence, apoptosis, proliferation, differentiation, and migration, and plays an essential role in regulating the hematopoiesis. This pathway can lose its physiologic regulation in pathologic conditions, leading to anemia and ineffective erythropoiesis. Activin receptor-ligand trap molecules such as Sotatercept and Luspatercept downregulate the TGF-ß pathway, thus inhibiting the Smad2/3 cascade and alleviating anemia in patients with ß-thalassemia and myelodysplastic syndromes. In this review, we describe in extenso the TGF-ß pathway, as well as the molecular and biological basis of activin receptors ligand traps, focusing on their role in various ß-thalassemia experimental models. The most recent results from clinical trials on sotatercept and luspatercept will also be reviewed.

9.
Eur J Dermatol ; 30(5): 532-540, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33021473

RESUMEN

BACKGROUND: Erythropoietic protoporphyria (EPP) is a rare inherited disease associated with heme metabolism, characterized by severe life-long photosensitivity and liver involvement. OBJECTIVE: To provide epidemiological data of EPP in Italy. MATERIALS & METHODS: Prospective/retrospective data of EPP patients were collected by an Italian network of porphyria specialist centres (Gruppo Italiano Porfiria, GrIP) over a 20-year period (1996-2017). RESULTS: In total, 179 patients (79 females) with a clinical and biochemical diagnosis of EPP were assessed, revealing a prevalence of 3.15 cases per million persons and an incidence of 0.13 cases per million persons/year. Incidence significantly increased after 2009 (due to the availability of alfa-melanotide, which effectively limits skin photosensitivity). Mean age at diagnosis was 28 years, with only 22 patients (12.2%) diagnosed ≤10 years old. Gene mutations were assessed in 173 (96.6%) patients; most (164; 91.3%) were FECH mutations on one allele in association with the hypomorphic variant, c.315-48C, on the other (classic EPP), and nine (5.2%) were ALAS2 mutations (X-linked EPP). Only one case of autosomal recessive EPP was observed. Of the 42 different FECH mutations, 15 are novel, three mutations collectively accounted for 45.9% (75/164) of the mutations (c.215dupT [27.2%], c.901_902delTG [11.5%] and c.67 + 5G > A [7.2%]), and frameshift mutations were prevalent (33.3%). A form of light protection was used by 109/179 (60.8%) patients, and 100 (56%) had at least one α-melanotide implant. Three cases of severe acute liver involvement, requiring OLT, were observed. CONCLUSION: These data define, for the first time, the clinical and molecular epidemiology of EPP in Italy.


Asunto(s)
Protoporfiria Eritropoyética/epidemiología , Protoporfiria Eritropoyética/genética , 5-Aminolevulinato Sintetasa/genética , Adulto , Estudios Transversales , Femenino , Ferroquelatasa/genética , Genes Recesivos , Genes Ligados a X , Humanos , Incidencia , Italia , Masculino , Epidemiología Molecular , Mutación , Prevalencia , Estudios Prospectivos , Estudios Retrospectivos
10.
Genet Med ; 22(1): 35-43, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31273344

RESUMEN

PURPOSE: Existing data do not explain the reason why some individuals homozygous for the hypomorphic FECH allele develop erythropoietic protoporphyria (EPP) while the majority are completely asymptomatic. This study aims to identify novel possible genetic variants contributing to this variable phenotype. METHODS: High-throughput resequencing of the FECH gene, qualitative analysis of RNA, and quantitative DNA methylation examination were performed on a cohort of 72 subjects. RESULTS: A novel deep intronic variant was found in four homozygous carriers developing a clinically overt disease. We demonstrate that this genetic variant leads to the insertion of a pseudo-exon containing a stop codon in the mature FECH transcript by the abolition of an exonic splicing silencer site and the concurrent institution of a new methylated CpG dinucleotide. Moreover, we show that the hypomorphic FECH allele is linked to a single haplotype of about 20 kb in size that encompasses three noncoding variants that were previously associated with expression quantitative trait loci (eQTLs). CONCLUSION: This study confirms that intronic variants could explain the variability in the clinical manifestations of EPP. Moreover, it supports the hypothesis that the control of the FECH gene expression can be mediated through a methylation-dependent modulation of the precursor messenger RNA (pre-mRNA) splicing pattern.


Asunto(s)
Sustitución de Aminoácidos , Ferroquelatasa/genética , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Protoporfiria Eritropoyética/genética , Empalme Alternativo , Codón de Terminación , Metilación de ADN , Regulación hacia Abajo , Epigénesis Genética , Humanos , Intrones , Sitios de Carácter Cuantitativo , Análisis de Secuencia de ADN/métodos , Análisis de Secuencia de ARN/métodos
11.
Immunol Res ; 67(4-5): 382-389, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31760565

RESUMEN

Phototoxic reaction is a known feature of EPP at least in part triggered by the oxidative status, complement system activation, and mast cell response. The aim of this study was to verify some aspects involved in phototoxic reaction during a season. The complement system was evaluated by C3 assay, alternative pathway by factor-B, and classical pathway by C1q; oxidative status was tested with malondialdehyde (MDA) and mast cell by IL-10 assay. The serum samples were collected in winter and summer from 19 EPP patients and 13 controls. The reaction to sun exposure within each group was monitored without any invasive treatment. In summer, C3 and factor B were higher in patients than in controls (p = 0.002 and < 0.0001 respectively), while no change was detected for C1q. The oxidative stress was increased in summer in comparison with the control group (p = 0.04), and IL-10 an assay was normal in both seasons. The correlation between the C3 and factor-B in summer was significant. This study shows that the phototoxic reaction is not limited to the dermis but can also exert a systemic response, which could affect the general health of a patient. The knowledge of the pathophysiology of phototoxic reaction is essential for identifying new disease markers useful for improving clinical studies of known and future drugs.


Asunto(s)
Proteínas del Sistema Complemento , Dermatitis Fototóxica , Interleucina-10 , Malondialdehído , Mastocitos , Protoporfiria Eritropoyética , Adulto , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Dermatitis Fototóxica/sangre , Dermatitis Fototóxica/inmunología , Dermatitis Fototóxica/patología , Femenino , Humanos , Interleucina-10/sangre , Interleucina-10/inmunología , Masculino , Malondialdehído/sangre , Malondialdehído/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Mastocitos/patología , Persona de Mediana Edad , Estrés Oxidativo/inmunología , Protoporfiria Eritropoyética/sangre , Protoporfiria Eritropoyética/inmunología , Protoporfiria Eritropoyética/patología , Estaciones del Año , Luz Solar/efectos adversos
12.
Ann Hematol ; 98(4): 833-840, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30506348

RESUMEN

Beta-thalassemia can present with a wide spectrum of phenotypes determined by the coinheritance of α-thalassemia, hereditary persistence of fetal hemoglobin, and polymorphic variants in the BCL11A, HMIP, and HBB clusters. The codon 29 (cd29) mutation in the beta gene has been associated with a broad diversity of thalassemia phenotypes, possibly through genetic modifiers determining the genotype-phenotype relationship. In this study, we evaluated the effect of 10 single nucleotide polymorphisms (SNPs) on ß-thalassemia severity in a group of 21 Lebanese patients bearing the cd29 mutation. Hematological parameters and clinical characteristics were evaluated according to transfusion dependence. The proportions and absolute concentrations of HbF were found to be higher in non-transfusion-dependent (NTD) patients than in transfusion-dependent (TD) ones. Iron parameters were found to be higher in TD patients. The SNPs that were evaluated included the XmnI-158 polymorphism in the HBG gene and SNPs in the BCL11A and HMIP loci. It was noted that individuals homozygous or heterozygous for the effect allele in the BCL11A and HMIP SNPs had higher HbF levels, lower ferritin concentrations, and lower liver iron content and were less likely to be transfusion dependent. Our results showed that HbF production variants may have an important impact on the severity of ß-thalassemia, which might provide a severity prediction tool that can help in the anticipation of patients' phenotypes and therefore in future therapeutic decision making.


Asunto(s)
Codón , Hemoglobina Fetal/genética , Sitios Genéticos , Polimorfismo de Nucleótido Simple , Globinas beta/genética , Talasemia beta/genética , Adolescente , Adulto , Transfusión Sanguínea , Proteínas Portadoras/genética , Femenino , Hemoglobina Fetal/metabolismo , Humanos , Líbano , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Represoras , Globinas beta/metabolismo , Talasemia beta/sangre , Talasemia beta/terapia
13.
Mol Genet Metab ; 125(3): 295-301, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30201327

RESUMEN

Genetic variants in promoters and alternative-splicing lesions require to be experimentally tested in order to validate them as causatives of a disease. The digital PCR (dPCR) approach, which is an alternative to the classical qPCR, is an innovative and a more sensitive method for the detection and quantification of nucleic acids. In the present study, we identified four HMBS gene mutations affecting the ubiquitous isoform of porphobilinogen deaminase (PBGD) and established a dPCR protocol which would be able to detect the different transcripts of this gene. With the application of this method, we were able to characterize the functional roles of these four genetic variants, demonstrating that all these mutations were causatives of the non-erythroid variant of the acute intermittent porphyria (AIP) disease.


Asunto(s)
Hidroximetilbilano Sintasa/genética , Porfiria Intermitente Aguda/genética , Isoformas de Proteínas/genética , Sitios de Empalme de ARN/genética , Adulto , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Reacción en Cadena de la Polimerasa , Porfiria Intermitente Aguda/patología , Regiones Promotoras Genéticas/genética , Adulto Joven
14.
Mol Genet Metab ; 124(4): 287-296, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29941360

RESUMEN

Alterations in the ferrochelatase gene (FECH) are the basis of the phenotypic expressions in erythropoietic protoporphyria. The phenotype is due to the presence of a mutation in the FECH gene associated in trans to the c.315-48 T > C variant in the intron 3. The latter is able to increase the physiological quota of alternative splicing events in the intron 3. Other two variants in the FECH gene (c.1-252A > G and c.68-23C > T) have been found to be associated to the intron 3 variant in some populations and together, they constitute a haplotype (ACT/GTC), but eventually, their role in the alternative splicing event has never been elucidated. The absolute number of the aberrantly spliced FECH mRNA molecules and the absolute expression of the FECH gene were evaluated by digital PCR technique in a comprehensive cohort. The number of splicing events that rose in the presence of the c.315-48 T > C variant, both in the heterozygous and homozygous condition was reported for the first time. Also, the percentage of the inserted FECH mRNA increased, even doubled in the T/C cases, compared to T/T cases. The constant presence of variants in the promoter and intron 2 did not influence or modulate the aberrant splicing. The results of FECH gene expression suggested that the homozygosity for the c.315-48 T > C variant could be considered pathological. Thus, this study identified the homozygotes for the c.315-48 T > C variant as pathological. By extension, when the samples were categorised according to the haplotypes, the GTC haplotype in homozygosis was pathological.


Asunto(s)
Ferroquelatasa/genética , Predisposición Genética a la Enfermedad , Protoporfiria Eritropoyética/genética , Empalme Alternativo/genética , Femenino , Haplotipos/genética , Heterocigoto , Homocigoto , Humanos , Masculino , Linaje , Polimorfismo de Nucleótido Simple/genética , Protoporfiria Eritropoyética/enzimología , Protoporfiria Eritropoyética/patología
16.
Blood Cells Mol Dis ; 61: 48-53, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27667166

RESUMEN

The PPOX gene encodes for the protoporphyrinogen oxidase, which is involved in heme production. The partial deficiency of protoporphyrinogen oxidase causes variegate porphyria. The tissue-specific regulation of other heme biosynthetic enzymes is extensively studied, but the information concerning transcriptional and post-transcriptional regulation of PPOX gene expression is scarcely available. In this study, we characterized functions of three variants identified in the regulatory regions of the PPOX gene, which show a novel role for the 5' untranslated exon 1. Using luciferase assays and RNA analysis, we demonstrated that only c.1-883G>C promoter variant causes a significant loss in the transcriptional activity of PPOX gene whereas c.1-413G>T 5' UTR variant inhibits translation of PPOX mRNA and c.1-176G>A splicing variant causes 4bp deletion in 5' UTR of PPOX mRNA variant 2. These observations indicate that the regulation of PPOX gene expression can also occur through a post-transcriptional modulation of the amount of gene product and that this modulation can be mediated by 5' untranslated exon 1. Moreover this study confirms that these regulatory regions represent an important molecular target for the pathogenesis of variegate porphyria.


Asunto(s)
Epigénesis Genética , Flavoproteínas/genética , Proteínas Mitocondriales/genética , Mutación , Porfiria Variegata/genética , Protoporfirinógeno-Oxidasa/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Regiones no Traducidas 5'/genética , Recolección de Muestras de Sangre , Línea Celular Tumoral , Exones , Regulación de la Expresión Génica , Humanos , Porfiria Variegata/etiología
17.
Br J Haematol ; 173(3): 365-79, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26969896

RESUMEN

Congenital erythropoietic porphyria (CEP) is a rare genetic disease resulting from the remarkable deficient activity of uroporphyrinogen III synthase, the fourth enzyme of the haem biosynthetic pathway. This enzyme defect results in overproduction of the non-physiological and pathogenic porphyrin isomers, uroporphyrin I and coproporphyrin I. The predominant clinical characteristics of CEP include bullous cutaneous photosensitivity to visible light from early infancy, progressive photomutilation and chronic haemolytic anaemia. The severity of clinical manifestations is markedly heterogeneous among patients; and interdependence between disease severity and porphyrin amount in the tissues has been pointed out. A more pronounced endogenous production of porphyrins concomitant to activation of ALAS2, the first and rate-limiting of the haem synthesis enzymes in erythroid cells, has also been reported. CEP is inherited as autosomal recessive or X-linked trait due to mutations in UROS or GATA1 genes; however an involvement of other causative or modifier genes cannot be ruled out.


Asunto(s)
Porfiria Eritropoyética/patología , Factor de Transcripción GATA1/genética , Hemo/biosíntesis , Humanos , Mutación , Fenotipo , Porfiria Eritropoyética/etiología , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/metabolismo , Porfirinas/biosíntesis , Porfirinas/metabolismo , Uroporfirinógeno III Sintetasa
18.
Hemoglobin ; 39(3): 178-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25902180

RESUMEN

ß-Thalassemia intermedia (ß-TI) is a clinical term describing a range of clinical phenotypes that are intermediate in severity between the carrier state and ß-thalassemia major (ß-TM). To characterize the molecular basis of ß-TI in Erbil Province, Northern Iraq, 83 unrelated patients were investigated. Detection of ß-globin gene mutations was carried out by reverse hybridization assay and direct gene sequencing. All patients were screened for the XmnI polymorphism by direct sequencing of HBG2 ((G)γ promoter gene). Detection of α-globin gene deletions and triplication was carried out using the reverse hybridization assay. Four main molecular patterns were identified in association with the ß-TI phenotype, namely: ß(+)/ß(+) (38.5%), ß(+)/ß(0) (21.6%), ß(0)/ß(0) (31.3%), and ß(0)/wild type (8.4%). IVS-I-6 (T > C) was the most frequently encountered mutation (55 alleles, 34.6%), followed by IVS-II-1 (G > A) and codon 8 (-AA); furthermore, we report for the first time from Iraq two ß(+) mutations, -87 (C > G) and 5' untranslated region (5'UTR) +22 (G > A). The XmnI polymorphism was detected in 47.0% of patients, mainly in association with the ß(0)/ß(0) genotype. The α-globin gene deletions were encountered in four cases, including one case with (- -(FIL)) double gene deletion, a report that is the first from our country. The α-globin gene triplication was detected in five of the seven heterozygous ß-thalassemia (ß-thal) patients. Similar to other Mediterranean countries, inheritance of mild ß-globin mutations was the main molecular pattern underlying ß-TI in our patients followed by the ameliorating effect of the XmnI polymorphism.


Asunto(s)
Etnicidad/genética , Mutación , Globinas beta/genética , Talasemia beta/epidemiología , Talasemia beta/genética , Adolescente , Adulto , Alelos , Niño , Preescolar , Codón , Índices de Eritrocitos , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Irak/epidemiología , Masculino , Persona de Mediana Edad , Fenotipo , Adulto Joven , Talasemia beta/diagnóstico
19.
Eur J Haematol ; 94(6): 491-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25251786

RESUMEN

Congenital erythropoietic porphyria (CEP) is a rare genetic disease that is characterized by a severe cutaneous photosensitivity causing unrecoverable deformities, chronic hemolytic anemia requiring blood transfusion program, and by fatal systemic complications. A correct and early diagnosis is required to develop a management plan that is appropriate to the patient's needs. Recently only one case of X-linked CEP had been reported, describing the trans-acting GATA1-R216W mutation. Here, we have characterized two novel X-linked CEP patients, both with misleading hematological phenotypes that include dyserythropoietic anemia, thrombocytopenia, and hereditary persistence of fetal hemoglobin. We compare the previously reported case to ours and propose a diagnostic paradigm for this variant of CEP. Finally, a correlation between phenotype variability and the presence of modifier mutations in loci related to disease-causing gene is described.


Asunto(s)
Sustitución de Aminoácidos , Factor de Transcripción GATA1/genética , Estudios de Asociación Genética , Mutación , Porfiria Eritropoyética/diagnóstico , Porfiria Eritropoyética/genética , Biopsia , Médula Ósea/patología , Niño , Preescolar , Análisis Mutacional de ADN , Índices de Eritrocitos , Genes Ligados a X , Humanos , Masculino , Linaje , Fenotipo , Porfirinas/sangre , Porfirinas/orina
20.
Blood Cells Mol Dis ; 49(3-4): 147-51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22748422

RESUMEN

Acute intermittent porphyria (AIP) is an autosomal dominant disorder caused by molecular abnormalities in the HMBS gene. This gene is transcribed from two promoters to produce ubiquitous and erythroid specific isoforms of porphobilinogen deaminase (PBGD). In the classical form of AIP, both isoforms are deficient, but about 5% of families have the non-erythroid variant in which only the ubiquitous isoform is affected. Only one mutation sited in the housekeeping promoter has been previously reported as causative for this form of AIP. In this study, we identified one small deletion and six nucleotide substitutions within the 5'UTR and the housekeeping promoter of HMBS gene: c.1-440_-427del14bp; c.1-421G>A; c.1-331C>T; c.1-270G>A; c.1-122T>A; c.1-103C>T; c.1-28A>C. Using luciferase reporter assays and quantitative PCR experiments, we characterized the functional role of these seven novel genetic variants demonstrating that all mutations cause a significant loss of transcriptional activity. Our investigations suggest that these nucleotide substitutions may alter critical binding sites for transcriptional factors, which confirms that these regions represent an important molecular target for pathogenesis of non-erythroid form of acute intermittent porphyria.


Asunto(s)
Regiones no Traducidas 5' , Hidroximetilbilano Sintasa/genética , Porfiria Intermitente Aguda/genética , Secuencia de Bases , Sitios de Unión , Genes Dominantes , Genes Reporteros , Humanos , Isoenzimas/genética , Células K562 , Luciferasas/genética , Datos de Secuencia Molecular , Mutación , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Porfiria Intermitente Aguda/clasificación , Porfiria Intermitente Aguda/diagnóstico , Regiones Promotoras Genéticas , Unión Proteica , Eliminación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...