Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Liver Int ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38813953

RESUMEN

Porphyrias are rare, mostly inherited disorders resulting from altered activity of specific enzymes in the haem synthesis pathway that lead to accumulation of pathway intermediates. Photocutaneous symptoms occur when excess amounts of photoreactive porphyrins circulate in the blood to the skin, whereas increases in potentially neurotoxic porphyrin precursors are associated with neurovisceral symptoms. Current therapies are suboptimal and their mechanisms are not well established. As described here, emerging therapies address underlying disease mechanisms by introducing a gene, RNA or other specific molecule with the potential to cure or slow progression of the disease. Recent progress in nanotechnology and nanoscience, particularly regarding particle design and formulation, is expanding disease targets. More secure and efficient drug delivery systems have extended our toolbox for transferring specific molecules, especially into hepatocytes, and led to proof-of-concept studies in animal models. Repurposing existing drugs as molecular chaperones or haem synthesis inhibitors is also promising. This review summarizes key examples of these emerging therapeutic approaches and their application for hepatic and erythropoietic porphyrias.

3.
Int J Mol Sci ; 24(1)2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36613492

RESUMEN

Porphobilinogen deaminase (PBGD) haploinsufficiency (acute intermittent porphyria, AIP) is characterized by neurovisceral attacks associated with high production, accumulation and urinary excretion of heme precursors, δ-aminolevulinic acid (ALA) and porphobilinogen (PBG). The estimated clinical penetrance for AIP is extremely low (<1%), therefore it is likely that other factors may play an important role in the predisposition to developing attacks. Fasting is a known triggering factor. Given the increased prevalence of insulin resistance in patients and the large urinary loss of succinyl-CoA to produce ALA and PBG, we explore the impact of reduced availability of energy metabolites in the severity of AIP pathophysiology. Classic studies found clinical improvement in patients affected by AIP associated with the administration of glucose and concomitant insulin secretion, or after hyperinsulinemia associated with diabetes. Molecular studies have confirmed that glucose and insulin administration induces a repressive effect on hepatic ALA Synthase, the first and regulatory step of the heme pathway. More recently, the insulin-mimicking α-lipoic acid has been shown to improve glucose metabolism and mitochondrial dysfunction in a hepatocyte cell line transfected with interfering RNA targeting PBGD. In AIP mice, preventive treatment with an experimental fusion protein of insulin and apolipoprotein A-I improved the disease by promoting fat mobilization in adipose tissue, increasing the metabolite bioavailability for the TCA cycle and inducing mitochondrial biogenesis in the liver. In this review, we analyze the possible mechanisms underlying abnormal hepatocellular carbohydrate homeostasis in AIP.


Asunto(s)
Resistencia a la Insulina , Porfiria Intermitente Aguda , Animales , Ratones , Ácido Aminolevulínico/metabolismo , Metabolismo de los Hidratos de Carbono , Glucosa/uso terapéutico , Hemo/metabolismo , Hidroximetilbilano Sintasa/genética , Insulina/metabolismo , Porfobilinógeno/orina , Porfiria Intermitente Aguda/genética , Porfiria Intermitente Aguda/terapia , Humanos
4.
Diagnostics (Basel) ; 11(11)2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34829496

RESUMEN

Inborn errors of metabolism (IEM) constitute an important group of conditions characterized by an altered metabolic pathway. There are numerous guidelines for the diagnosis and management of IEMs in the pediatric population but not for adults. Given the increasing frequency of this group of conditions in adulthood, other clinicians in addition to pediatricians should be aware of them and learn to identify their characteristic manifestations. Early recognition and implementation of an appropriate therapeutic approach would improve the clinical outcome of many of these patients. This review presents when and how to investigate a metabolic disorder with the aim of encouraging physicians not to overlook a treatable disorder.

6.
Biomedicines ; 9(3)2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807619

RESUMEN

Acute porphyria attacks are associated with the strong up-regulation of hepatic heme synthesis and over-production of neurotoxic heme precursors. First-line therapy is based on carbohydrate loading. However, altered glucose homeostasis could affect its efficacy. Our first aim was to investigate the prevalence of insulin resistance (IR) in an observational case-control study including 44 Spanish patients with acute intermittent porphyria (AIP) and 55 age-, gender- and BMI-matched control volunteers. Eight patients (18.2%) and one control (2.3%, p = 0.01) showed a high HOMA-IR index (cut-off ≥ 3.4). Patients with IR and hyperinsulinemia showed clinically stable disease. Thus, the second aim was to evaluate the effect of the co-administration of glucose and a fast-acting or new liver-targeted insulin (the fusion protein of insulin and apolipoprotein A-I, Ins-ApoAI) in AIP mice. The combination of glucose and the Ins-ApoAI promoted partial but sustained protection against hepatic heme synthesis up-regulation compared with glucose alone or co-injected with fast-acting insulin. In a prevention study, Ins-ApoAI improved symptoms associated with a phenobarbital-induced attack but maintained high porphyrin precursor excretion, probably due to the induction of hepatic mitochondrial biogenesis mediated by apolipoprotein A-I. In conclusion, a high prevalence of IR and hyperinsulinemia was observed in patients with AIP. The experimental data provide proof-of-concept for liver-targeted insulin as a way of enhancing glucose therapy for AIP.

7.
Ann Transl Med ; 8(17): 1098, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33145317

RESUMEN

Hyponatremia is a common feature during the neurovisceral acute attacks which characterize hepatic porphyrias, as well as a sign of its severity. Therapeutic options for first-line acute attacks are intravenous administration of glucose and/or exogenous heme. The former treatment can aggravate hyponatremia by dilution and cause seizures; thus, the correction of hyponatremia must be carried out with extreme caution. This review summarizes recommendations for the management of hyponatremia during acute episodes of porphyria. Hyponatremia should be corrected slowly and seizures treated with medications in order to not exacerbate motor and sensory axonal neuropathy. The syndrome of inappropriate antidiuretic hormone secretion (SIADH) is considered a frequent cause of hyponatremia in acute porphyrias and must be identified as a symptom of an acute porphyria attack. Tolvaptan produces aquaresis and is considered a safe drug in porphyria. However, its use has only been reported in isolated cases during a porphyria attack. The convenience and usefulness of this drug in acute porphyria are discussed.

8.
Genes (Basel) ; 11(8)2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32806544

RESUMEN

Acute intermittent porphyria (AIP) results from a decreased activity of hepatic hydroxymethylbilane synthase (HMBS), the third enzyme in the heme biosynthetic pathway. AIP is an autosomal dominant disorder with incomplete penetrance, characterized by acute neurovisceral attacks precipitated by several factors that induce the hepatic 5-aminolevulinic acid synthase, the first enzyme in the heme biosynthesis. Thus, a deficiency in HMBS activity results in an overproduction of porphyrin precursors and the clinical manifestation of the disease. Early diagnosis and counselling are essential to prevent attacks, and mutation analysis is the most accurate method to identify asymptomatic carriers in AIP families. In the present study, we have investigated the molecular defects in 55 unrelated Spanish patients with AIP, identifying 32 HMBS gene mutations, of which six were novel and ten were found in more than one patient. The novel mutations included a missense, an insertion, two deletions, and two splice site variants. Prokaryotic expression studies demonstrated the detrimental effect for the missense mutation, whereas reverse transcription-PCR and sequencing showed aberrant splicing caused by each splice site mutation. These results will allow for an accurate diagnosis of carriers of the disease in these families. Furthermore, they increase the knowledge about the molecular heterogeneity of AIP in Spain.


Asunto(s)
Biomarcadores , Porfiria Intermitente Aguda/etiología , Porfiria Intermitente Aguda/metabolismo , Adolescente , Adulto , Análisis Mutacional de ADN , Susceptibilidad a Enfermedades , Femenino , Genotipo , Humanos , Hidroximetilbilano Sintasa/genética , Hidroximetilbilano Sintasa/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Reacción en Cadena de la Polimerasa , Porfiria Intermitente Aguda/diagnóstico , Empalme del ARN , España , Adulto Joven
9.
Eur J Med Genet ; 62(12): 103589, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30476629

RESUMEN

Porphyrias are rare diseases caused by alterations in the heme biosynthetic pathway. Depending on the afected enzyme, porphyrin precursors or porphyrins are overproduced, causing acute neurovisceral attacks or dermal photosensitivity, respectively. Hereditary Coproporphyria (HCP) and Variegate Porphyria (VP) are mixed porphyrias since they can present acute and/or cutaneous symptoms. These diseases are caused by a deficiency of coproporphyrinogen oxidase (CPOX) in HCP, and protoporphyrinogen oxidase (PPOX) in VP. Herein, we studied nineteen unrelated Spanish patients with mixed porphyrias. The diagnosis of either, HCP or VP was made on the basis of clinical symptoms, biochemical findings and the identification of the mutation responsible in the CPOX or PPOX genes. Two patients presented both acute and cutaneous symptoms. In most patients, the biochemical data allowed the diagnosis. Among eleven patients with HCP, ten CPOX mutations were identified, including six novel ones: two frameshift (c.32delG and c.1102delC), two nonsense (p.Cys239Ter and p.Tyr365Ter), one missense (p.Trp275Arg) and one amino acid deletion (p.Gly336del). Moreover, seven previously described PPOX mutations were identified in eight patients with VP. The impacts of CPOX mutations p.Trp275Arg and p.Gly336del, were evaluated using prediction softwares and their functional consequences were studied in a prokaryotic expression system. Both alterations were predicted as deleterious by in silico analysis. Aditionally, when these alleles were expressed in E. coli, only p.Trp275Arg retained some residual activity. These results emphasize the usefulness of integrated the biochemical tests and molecular studies in the diagnosis. Furthermore, they extend knowledge on the molecular heterogeneity of mixed porphyrias in Spain.


Asunto(s)
Porfirias/genética , Adulto , Anciano , Coproporfirinógeno Oxidasa/genética , Coproporfirinógeno Oxidasa/metabolismo , Femenino , Flavoproteínas/genética , Flavoproteínas/metabolismo , Pruebas Genéticas/estadística & datos numéricos , Humanos , Mutación con Pérdida de Función , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mutación Missense , Porfirias/epidemiología , Protoporfirinógeno-Oxidasa/genética , Protoporfirinógeno-Oxidasa/metabolismo , España
10.
Hum Gene Ther ; 29(4): 480-491, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28990424

RESUMEN

Porphobilinogen deaminase (PBGD) gene therapy represents a promising therapeutic option for acute intermittent porphyria (AIP) patients suffering recurrent acute attacks. A first-in-human Phase I clinical trial confirmed the safety and tolerability of adeno-associated virus (AAV)-AAT-PBGD gene therapy, but higher doses and/or more efficient vectors are needed to achieve therapeutic expression of the transgene. This study assayed the insertion into the promoter of a short enhancer element able to induce transgene expression during exposure to endogenous and exogenous stimuli related to the pathology of the disease. The inclusion in tandem of two elements of the minimal functional sequence of human δ-aminolevulinic acid synthase drug-responsive enhancing sequence (ADRES) positioned upstream of the promoter strongly induced transgene expression in the presence of estrogens, starvation, and certain drugs known to trigger attacks in porphyria patients. The inclusion of two ADRES motives in an AAV vector improved therapeutic efficacy, reducing 10-fold the effective dose in AIP mice. In conclusion, the inclusion of specific enhancer elements in the promoter of gene therapy vectors for AIP was able to overexpress the therapeutic transgene when it is most needed, at the time when porphyrinogenic factors increase the demand for hepatic heme and precipitate acute porphyria attacks.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Porfiria Intermitente Aguda/terapia , 5-Aminolevulinato Sintetasa/genética , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Vectores Genéticos/genética , Humanos , Hidroximetilbilano Sintasa/genética , Hígado/metabolismo , Hígado/patología , Ratones , Porfiria Intermitente Aguda/genética , Porfiria Intermitente Aguda/patología , Regiones Promotoras Genéticas
11.
J Gynecol Oncol ; 28(5): e58, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28657220

RESUMEN

OBJECTIVE: To externally validate the prognostic models for predicting the time-dependent outcome in patients with locally advanced cervical cancer (LACC) who were treated with concurrent chemoradiotherapy in an independent cohort. METHODS: A historical cohort of 297 women with LACC who were treated with radical concurrent chemoradiotherapy from 1999 to 2014 at the 12 de Octubre University Hospital (H12O), Madrid, Spain. The external validity of prognostic models was quantified regarding discrimination, calibration, measures of overall performance, and decision curve analyses. RESULTS: The review identified 8 studies containing 13 prognostic models. Different (International Federation of Gynecology and Obstetrics [FIGO] stages, parametrium involvement, hydronephrosis, location of positive nodes, and race) but related cohorts with validation cohort (5-year overall survival [OS]=70%; 5-year disease-free survival [DFS]=64%; average age of 50; and over 79% squamous cell) were evaluated. The following models exhibited good external validity in terms of discrimination and calibration but limited clinical utility: the OS model at 3 year from Kidd et al.'s study (area under the receiver operating characteristic curve [AUROC]=0.69; threshold of clinical utility [TCU] between 36% and 50%), the models of DFS at 1 year from Kidd et al.'s study (AUROC=0.64; TCU between 24% and 32%) and 2 years from Rose et al.'s study (AUROC=0.70; TCU between 19% and 58%) and the distant recurrence model at 5 years from Kang et al.'s study (AUROC=0.67; TCU between 12% and 36%). CONCLUSION: The external validation revealed the statistical and clinical usefulness of 4 prognostic models published in the literature.


Asunto(s)
Neoplasias del Cuello Uterino/diagnóstico , Neoplasias del Cuello Uterino/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Quimioradioterapia , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/epidemiología , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Riesgo , España , Resultado del Tratamiento , Neoplasias del Cuello Uterino/terapia
12.
Hum Mol Genet ; 25(7): 1318-27, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908609

RESUMEN

Porphobilinogen deaminase (PBGD) haploinsufficiency (acute intermittent porphyria, AIP) is characterized by neurovisceral attacks when hepatic heme synthesis is activated by endogenous or environmental factors including fasting. While the molecular mechanisms underlying the nutritional regulation of hepatic heme synthesis have been described, glucose homeostasis during fasting is poorly understood in porphyria. Our study aimed to analyse glucose homeostasis and hepatic carbohydrate metabolism during fasting in PBGD-deficient mice. To determine the contribution of hepatic PBGD deficiency to carbohydrate metabolism, AIP mice injected with a PBGD-liver gene delivery vector were included. After a 14 h fasting period, serum and liver metabolomics analyses showed that wild-type mice stimulated hepatic glycogen degradation to maintain glucose homeostasis while AIP livers activated gluconeogenesis and ketogenesis due to their inability to use stored glycogen. The serum of fasted AIP mice showed increased concentrations of insulin and reduced glucagon levels. Specific over-expression of the PBGD protein in the liver tended to normalize circulating insulin and glucagon levels, stimulated hepatic glycogen catabolism and blocked ketone body production. Reduced glucose uptake was observed in the primary somatosensorial brain cortex of fasted AIP mice, which could be reversed by PBGD-liver gene delivery. In conclusion, AIP mice showed a different response to fasting as measured by altered carbohydrate metabolism in the liver and modified glucose consumption in the brain cortex. Glucose homeostasis in fasted AIP mice was efficiently normalized after restoration of PBGD gene expression in the liver.


Asunto(s)
Modelos Animales de Enfermedad , Ayuno/metabolismo , Glucosa/metabolismo , Hidroximetilbilano Sintasa/genética , Hígado/metabolismo , Porfiria Intermitente Aguda/metabolismo , Animales , Corteza Cerebral/metabolismo , Ayuno/sangre , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Glucagón/sangre , Homeostasis , Insulina/sangre , Masculino , Ratones , Ratones Noqueados , Porfiria Intermitente Aguda/sangre , Porfiria Intermitente Aguda/terapia
13.
J Proteomics ; 127(Pt B): 377-85, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25979770

RESUMEN

UNLABELLED: Acute intermittent porphyria (AIP) is an autosomal dominant metabolic disorder caused by a deficiency of hepatic porphobilinogen deaminase (PBGD). The disease is characterized by life threatening acute neurovisceral attacks. The aim of this study was to identify metabolites secreted by the hepatocytes that reflect differential metabolic status in the liver and that may predict response to the acute attack treatment. Plasma vitamin D binding protein (VDBP) from a mouse model of AIP displayed an abnormal migration in 2D-electrophoresis that is efficiently recovered upon gene therapy leading to liver specific over-expression of the PBGD protein. The change in VDBP mobility results from a differential isoelectric point suggesting a post-translational modification that takes place preferably in the liver. Liquid chromatography-mass spectrometry (LC-MS) analysis of human samples before and after glycosidase treatment revealed glycosylated plasma VDBP specifically in patients with recurrent attacks of AIP. Glycosylated VDBP recovered normal values in three severely afflicted AIP patients submitted to therapeutic liver transplantation. Our findings suggest that post-translational modification of VDBP might be considered as a promising biomarker to study and monitor the liver metabolic status in patients with AIP. SIGNIFICANCE: We describe an increased glycosylation of VDBP in porphyric livers. Normal glycosylation was recovered upon liver gene therapy in a mouse model of porphyria or after liver transplantation in severely afflicted patients with AIP. Moreover, quantification of glycosylated VDBP by our ELISA immunoassay or LC-MS protocol in patients undergoing PBGD-gene therapy (www.aipgene.org) may be used as a marker indicating improvement or normalization of the patient's hepatic metabolism. This article is part of a Special Issue entitled: HUPO 2014.


Asunto(s)
Hígado/metabolismo , Porfiria Intermitente Aguda/metabolismo , Proteína de Unión a Vitamina D/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Cromatografía Liquida , Femenino , Glicosilación , Humanos , Hígado/patología , Masculino , Espectrometría de Masas , Ratones , Porfiria Intermitente Aguda/genética , Porfiria Intermitente Aguda/patología
14.
Am J Hematol ; 89(7): 689-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24644245

RESUMEN

Ferroportin disease is an inherited disorder of iron metabolism and is caused by mutations in the ferroportin gene (SLC40A1). We present a patient with hyperferritinemia, iron overload in the liver with reticuloendothelial distribution and also in the spleen, and under treatment with erythropheresis. A molecular study of the genes involved in iron metabolism (HFE, HJV, HAMP, TFR2, SLC40A1) was undertaken. In vitro functional studies of the novel mutation found in the SLC40A1 gene was performed. The patient was heterozygous for a novel mutation, c.386T>C (p.L129P) in the SLC40A1 gene; some of his relatives were also heterozygous for this mutation. In vitro functional studies of the L129P mutation on ferroportin showed it impairs its capacity to export iron from cells but does not alter its sensitivity to hepcidin. These findings and the iron overload phenotype of the patient suggest that the novel mutation c.386T>C (p.L129P) in the SLC40A1 gene has incomplete penetrance and causes the classical form of ferroportin disease.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Sobrecarga de Hierro/genética , Hierro/metabolismo , Mutación , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Preescolar , Femenino , Ferritinas/sangre , Genotipo , Células HEK293 , Hepcidinas/administración & dosificación , Humanos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/metabolismo , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Fenotipo
15.
Hum Mol Genet ; 22(14): 2929-40, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23562909

RESUMEN

Acute intermittent porphyria (AIP) is a hepatic metabolic disease that results from haplo-insufficient activity of porphobilinogen deaminase (PBGD). The dominant clinical feature is acute intermittent attacks when hepatic heme synthesis is activated by endocrine or exogenous factors. Gene therapy vectors over-expressing PBGD protein in the liver offers potential as a cure for AIP. Here, we developed a helper-dependent adenovirus (HDA) encoding human PBGD (hPBGD) and assessed its therapeutic efficacy in a murine model of AIP. Intravenous or intrahepatic administration of HDA-hPBGD to AIP mice resulted in a sustained hepatic hPBGD expression in a dose-dependent manner. Intrahepatic administration conveyed full protection against induced porphyria attacks at a significantly lower viral dose than intravenous injection. Transgenic hPBGD accumulated only in the cytosol of hepatocytes as the endogenous protein. Characterization of PBGD-deficient mouse strains revealed that a strong PBGD deficiency causes the chronic disturbance of cytosolic and endoplasmic reticulum folding machineries. This disturbance was completely restored over time by the over-expression of hPBGD. HDA-hPBGD is a promising vector that protects against porphyria attacks and resolves the chronic folding stress associated with low levels of PBGD activity.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Hidroximetilbilano Sintasa/genética , Porfiria Intermitente Aguda/genética , Porfiria Intermitente Aguda/terapia , Adenoviridae/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Hepatocitos/enzimología , Hepatocitos/virología , Humanos , Hidroximetilbilano Sintasa/metabolismo , Hígado/enzimología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Porfiria Intermitente Aguda/enzimología , Porfiria Intermitente Aguda/prevención & control , Pliegue de Proteína
16.
Gene ; 522(1): 89-95, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23545314

RESUMEN

Porphyria cutanea tarda (PCT) results from decreased activity of uroporphyrinogen decarboxylase (UROD) in the liver. Deficiency in this enzyme results in accumulation of highly carboxylated porphyrins responsible for the disease. PCT usually occurs in adulthood and is characterized by cutaneous photosensitivity, hyperpigmentation, skin fragility and hypertrichosis. Familial PCT (F-PCT) occurs in 20-30% of patients in whom UROD gene mutations in heterozygosity decrease the enzymatic activity to about 50% in all tissues. The rare homozygous form of F-PCT (hepatoerythropoietic porphyria) has more severe clinical features and onset in childhood. In Spain, F-PCT is molecularly heterogeneous and the most frequent UROD mutation is p.G281E. In the present study, we searched for the molecular defect causing F-PCT in a group of Spanish patients and investigated whether the p.G281E mutation in the Spanish population came from a single or various origins. Among seventeen F-PCT patients, sixteen UROD mutations were identified, including eight novel ones: six missense (p.A23V, p.L78P, p.W180G, p.T196I, p.E278G and p.V279M), one frameshift (c.233delT) and one splice site mutation (c.774G>C). Prokaryotic expression studies showed the detrimental effect for each missense mutation, whereas reverse transcription-PCR and sequencing demonstrated that the novel splice site mutation caused exon 7 skipping. Moreover, haplotype analysis performed in Spanish families with the p.G281E mutation indicated that this lesion is associated with at least five haplotype backgrounds. These results extend knowledge on the molecular heterogeneity of F-PCT and suggest multiple origins of the p.G281E mutation.


Asunto(s)
Mutación , Porfiria Cutánea Tardía/genética , Uroporfirinógeno Descarboxilasa/genética , Adulto , Alelos , Exones/genética , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Porfiria Cutánea Tardía/enzimología , España
17.
Gene ; 508(1): 15-20, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22890139

RESUMEN

Hereditary hemochromatosis causes iron overload and is associated with a variety of genetic and phenotypic conditions. Early diagnosis is important so that effective treatment can be administered and the risk of tissue damage avoided. Most patients are homozygous for the c.845G>A (p.C282Y) mutation in the HFE gene; however, rare forms of genetic iron overload must be diagnosed using a specific genetic analysis. We studied the genotype of 5 patients who had hyperferritinemia and an iron overload phenotype, but not classic mutations in the HFE gene. Two patients were undergoing phlebotomy and had no iron overload, 1 with metabolic syndrome and no phlebotomy had mild iron overload, and 2 patients had severe iron overload despite phlebotomy. The patients' first-degree relatives also underwent the analysis. We found 5 not previously published mutations: c.-408_-406delCAA in HFE, c.1118G>A (p.G373D), c.1473G>A (p.E491E) and c.2085G>C (p.S695S) in TFR2; and c.-428_-427GG>TT in SLC40A1. Moreover, we found 3 previously published mutations: c.221C>T (p.R71X) in HFE; c.1127C>A (p.A376D) in TFR2; and c.539T>C (p.I180T) in SLC40A1. Four patients were double heterozygous or compound heterozygous for the mutations mentioned above, and the patient with metabolic syndrome was heterozygous for a mutation in the TFR2 gene. Our findings show that hereditary hemochromatosis is clinically and genetically heterogeneous and that acquired factors may modify or determine the phenotype.


Asunto(s)
Proteínas de Transporte de Catión/genética , Hemocromatosis/genética , Antígenos de Histocompatibilidad Clase I/genética , Sobrecarga de Hierro/etiología , Proteínas de la Membrana/genética , Mutación/genética , Receptores de Transferrina/genética , Adulto , ADN/genética , Femenino , Genotipo , Hemocromatosis/complicaciones , Hemocromatosis/diagnóstico , Proteína de la Hemocromatosis , Heterocigoto , Homocigoto , Humanos , Sobrecarga de Hierro/diagnóstico , Masculino , Persona de Mediana Edad , Fenotipo , Reacción en Cadena de la Polimerasa
18.
PLoS One ; 7(3): e32978, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22412963

RESUMEN

Chronic kidney disease is a long-term complication in acute intermittent porphyria (AIP). The pathophysiological significance of hepatic overproduction of the porphyrin precursors aminolevulinate acid (ALA) and porphobilinogen (PBG) in chronic kidney disease is unclear. We have investigated the effect of repetitive acute attacks on renal function and the effect of total or five-sixth nephrectomy causing renal insufficiency on hepatic heme synthesis in the porphobilinogen deaminase (PBGD)-deficient (AIP) mouse. Phenobarbital challenge in the AIP-mice increased urinary porphyrin precursor excretion. Successive attacks throughout 14 weeks led to minor renal lesions with no impact on renal function. In the liver of wild type and AIP mice, 5/6 nephrectomy enhanced transcription of the first and rate-limiting ALA synthase. As a consequence, urinary PBG excretion increased in AIP mice. The PBG/ALA ratio increased from 1 in sham operated AIP animals to over 5 (males) and over 13 (females) in the 5/6 nephrectomized mice. Total nephrectomy caused a rapid decrease in PBGD activity without changes in enzyme protein level in the AIP mice but not in the wild type animals. In conclusion, high concentration of porphyrin precursors had little impact on renal function. However, progressive renal insufficiency aggravates porphyria attacks and increases the PBG/ALA ratio, which should be considered a warning sign for potentially life-threatening impairment in AIP patients with signs of renal failure.


Asunto(s)
Hemo/biosíntesis , Hidroximetilbilano Sintasa/metabolismo , Hígado/enzimología , Porfiria Intermitente Aguda/complicaciones , Porfiria Intermitente Aguda/enzimología , Insuficiencia Renal/enzimología , Insuficiencia Renal/etiología , Ácido Aminolevulínico/orina , Animales , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Pruebas de Función Renal , Masculino , Ratones , Ratones Endogámicos C57BL , Nefrectomía/efectos adversos , Porfirinógenos/orina , Porfirinas/orina , Factores Sexuales
19.
Mol Genet Metab ; 105(4): 629-33, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22382040

RESUMEN

Porphyria cutanea tarda (PCT) is caused by decreased activity of uroporphyrinogen decarboxylase (UROD) in the liver. The disease usually occurs in adulthood and is characterized by cutaneous photosensitivity, hyperpigmentation, skin fragility and hypertrichosis, due to the accumulation of porphyrins produced by oxidation of uroporphyrinogen and other highly carboxylated porphyrinogens overproduced as a result of the enzyme deficiency. PCT is generally sporadic, but about 20-30% of patients have familial-PCT (F-PCT) which is associated with heterozygosity of mutations in the UROD gene. In the present study we have found the molecular defect in seventeen unrelated Argentinean patients with F-PCT, identifying a total of eleven UROD gene mutations: four novel and seven previously described. The novel mutations were: a guanine insertion at the 5' splice junction of intron 2, a three nucleotide deletion causing the lost of valine 90, a deletion of 22 bp in exon 6 and a deletion of part of the polyadenylation signal. Prokaryotic expression studies showed that the novel amino acid deletion resulted in an inactive protein. Mutations c.10insA and p.M165R, previously found in Argentinean patients, were recurrent in this study; they are the most frequent in Argentina accounting for 40% of the mutant alleles characterized to date.


Asunto(s)
Predisposición Genética a la Enfermedad , Mutación/genética , Porfiria Cutánea Tardía/enzimología , Porfiria Cutánea Tardía/genética , Uroporfirinógeno Descarboxilasa/genética , Adolescente , Adulto , Argentina , Niño , Preescolar , ADN/genética , Exones/genética , Femenino , Humanos , Intrones/genética , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia , Adulto Joven
20.
Rev Esp Enferm Dig ; 103(7): 379-82, 2011 Jul.
Artículo en Inglés, Español | MEDLINE | ID: mdl-21770687

RESUMEN

Iron overload disease has a wide variety of genotypes. The genetic study of this disease confirms its hereditary nature and enables us to provide genetic counseling for first-degree relatives. We performed magnetic resonance imaging and liver biopsy in an asymptomatic patient with more than 1,000 µg/L of serum ferritin and studied the genes involved in this condition. The phenotype of iron overload is confirmed by a predominantly periportal pattern of iron deposits in the liver suggestive of genetic disease. In the case we present the molecular study revealed a double heterozygosity for the mutations c.187C>G (p.H63D) and c.840C>G (p.F280L) in the HFE and transferrin receptor 2 (TFR2) genes, respectively.


Asunto(s)
Hemocromatosis/genética , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Receptores de Transferrina/genética , Biopsia , ADN/genética , Ferritinas/sangre , Hemocromatosis/patología , Proteína de la Hemocromatosis , Humanos , Hierro/sangre , Hierro/metabolismo , Hígado/metabolismo , Hígado/patología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Receptores de Transferrina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transferrina/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA