Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Ther ; 248: 108487, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37392940

RESUMO

Protoporphyrin IX (PPIX) is an intermediate in the heme biosynthesis pathway. Abnormal accumulation of PPIX due to certain pathological conditions such as erythropoietic protoporphyria and X-linked protoporphyria causes painful phototoxic reactions of the skin, which can significantly impact daily life. Endothelial cells in the skin have been proposed as the primary target for PPIX-induced phototoxicity through light-triggered generation of reactive oxygen species. Current approaches for the management of PPIX-induced phototoxicity include opaque clothing, sunscreens, phototherapy, blood therapy, antioxidants, bone marrow transplantation, and drugs that increase skin pigmentation. In this review, we discuss the present understanding of PPIX-induced phototoxicity including PPIX production and disposition, conditions that lead to PPIX accumulation, symptoms and individual differences, mechanisms, and therapeutics.


Assuntos
Células Endoteliais , Protoporfiria Eritropoética , Humanos , Células Endoteliais/metabolismo , Protoporfirinas/farmacologia , Protoporfirinas/metabolismo , Protoporfiria Eritropoética/metabolismo , Protoporfiria Eritropoética/patologia , Protoporfiria Eritropoética/terapia , 5-Aminolevulinato Sintetase
2.
Photodermatol Photoimmunol Photomed ; 38(2): 141-149, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34420239

RESUMO

BACKGROUND: Erythropoietic protoporphyria (EPP) is a rare disorder of heme biosynthesis hallmarked by early-onset photosensitivity and mainly due to defective ferrochelatase activity leading to increased erythrocyte protoporphyrin IX (PPIX) levels. Evidence regarding the relationship between erythrocyte PPIX concentration and photosensitivity is limited. METHODS: To investigate the relationship between free erythrocyte PPIX (FEP) concentration; routine laboratory tests, particularly iron metabolism biomarkers; and ultraviolet (UV) A/visible light phototesting findings, 20 genetically confirmed EPP and one XLPP treatment-naive patients were included in our study. They underwent UVA and visible light phototesting. On the same day, blood samples were collected for measurement of FEP, serum iron, transferrin, transferrin saturation, and ferritin, 25-hydroxyvitamin D, and liver enzyme levels. RESULTS: Median FEP concentration at the time of phototesting was 57.50 (IQR: 34.58-102.70) µg/g of Hb. UVA and visible light phototesting were positive in 9 (42.9%) and 8 (38.1%) patients, respectively. Median FEP concentration was significantly higher in UVA phototest-positive patients than in those negative (64.37 [IQR: 57.45-121.82] vs 45.35 [IQR: 24.53-74.61] µg/g of Hb, respectively; P = .04486). Similarly, UVA photosensitive individuals had significantly lower median serum iron levels (61.5 [IQR: 33.5-84] µg/dL vs 109 [IQR: 63.25-154] µg/dL, respectively; P = .01862) and transferrin saturation values (15.005 [IQR: 7.0775-18.41] % vs 29.645 [IQR: 17.8225-34.3575] %; P = .0109) than those negative. CONCLUSIONS: Our study demonstrates that UVA phototest positivity is associated with higher FEP concentration and lower transferrin saturation and serum iron concentration in EPP.


Assuntos
Protoporfiria Eritropoética , Eritrócitos/metabolismo , Humanos , Protoporfiria Eritropoética/diagnóstico , Protoporfiria Eritropoética/metabolismo , Protoporfirinas/metabolismo , Transferrinas/metabolismo
3.
Cell Chem Biol ; 28(8): 1221-1234.e6, 2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-33756123

RESUMO

Erythropoietic protoporphyria (EPP) is a rare disease in which patients experience severe light sensitivity. It is caused by a deficiency of ferrochelatase (FECH), the last enzyme in heme biosynthesis (HBS). The lack of FECH causes accumulation of its photoreactive substrate protoporphyrin IX (PPIX) in patients' erythrocytes. Here, we explored an approach for the treatment of EPP by decreasing PPIX synthesis using small-molecule inhibitors directed to factors in the HBS pathway. We generated a FECH-knockout clone from K562 erythroleukemia cells, which accumulates PPIX and undergoes oxidative stress upon light exposure. We used these matched cell lines to screen a set of publicly available inhibitors of factors in the HBS pathway. Inhibitors of the glycine transporters GlyT1 and GlyT2 lowered levels of PPIX and markers of oxidative stress selectively in K56211B4 cells, and in primary erythroid cultures from an EPP patient. Our findings open the door to investigation of glycine transport inhibitors for HBS disorders.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Protoporfiria Eritropoética/tratamento farmacológico , Protoporfirinas/farmacologia , Células Cultivadas , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Humanos , Células K562 , Estrutura Molecular , Protoporfiria Eritropoética/metabolismo
4.
Front Immunol ; 12: 615620, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33664746

RESUMO

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.


Assuntos
Via Alternativa do Complemento/imunologia , Via Alternativa do Complemento/efeitos da radiação , Proteínas do Sistema Complemento/imunologia , Suscetibilidade a Doenças , Protoporfiria Eritropoética/etiologia , Luz Solar/efeitos adversos , Adulto , Biomarcadores , Complemento C5/imunologia , Complemento C5/metabolismo , Fator H do Complemento/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Properdina/imunologia , Properdina/metabolismo , Protoporfiria Eritropoética/diagnóstico , Protoporfiria Eritropoética/metabolismo , Estações do Ano
5.
J Photochem Photobiol B ; 213: 112054, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33075648

RESUMO

Erythropoietic protoporphyria (EPP) is characterised by accumulation of protoporphyrin IX (PpIX) in erythrocytes. Upon illumination PpIX is released to the skin. Activation of the photoactive substance PpIX causes painful skin symptoms. This study aimed to objectively quantify individual light exposure of EPP patients in their everyday lives through spring and summer. We further aimed to establish the associations between daily symptoms and light exposure dose to photoprimed and non-photoprimed skin, use of gloves, and erythrocyte PpIX concentration. 14 Danish EPP patients participated from April through June, the period when symptoms are most frequent. Light exposure was measured using personal electronic dosimeters with sensor sensitivity comparable to the absorption spectrum of PpIX, measuring the biological effect of the light in this disease. Concurrently participants reported symptoms and use of protective gloves in a diary. Patients had a blood sample analysed for erythrocyte PpIX. The median patient was exposed to an average daily PpIX-weighted light dose of 3.8 J/cm2 corresponding to approximately 15 min in the midday sun during summer in Denmark. The median patient reported symptoms on 29% and wore gloves on 11% of study days. There was a significant positive correlation between erythrocyte PpIX concentration and percentage of days wearing gloves (r = 0.65, p = 0.011), and a significant negative correlation between erythrocyte PpIX concentration and mean daily light dose on days not wearing gloves (r = -0.53, p = 0.049). Photosensitivity was strongly dependent on photopriming.


Assuntos
Eritrócitos/metabolismo , Protoporfiria Eritropoética/metabolismo , Protoporfirinas/metabolismo , Pele/metabolismo , Dinamarca , Eritrócitos/citologia , Feminino , Mãos/efeitos da radiação , Humanos , Luz , Masculino , Transtornos de Fotossensibilidade/etiologia , Protoporfirinas/sangue , Dosímetros de Radiação , Exposição à Radiação/efeitos adversos , Estações do Ano , Pele/citologia , Espectrofotometria , Fatores de Tempo
6.
Nucleic Acids Res ; 48(9): 4658-4671, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32313951

RESUMO

Erythropoietic protoporphyria (EPP) is a rare genetic disease in which patients experience acute phototoxic reactions after sunlight exposure. It is caused by a deficiency in ferrochelatase (FECH) in the heme biosynthesis pathway. Most patients exhibit a loss-of-function mutation in trans to an allele bearing a SNP that favors aberrant splicing of transcripts. One viable strategy for EPP is to deploy splice-switching oligonucleotides (SSOs) to increase FECH synthesis, whereby an increase of a few percent would provide therapeutic benefit. However, successful application of SSOs in bone marrow cells is not described. Here, we show that SSOs comprising methoxyethyl-chemistry increase FECH levels in cells. We conjugated one SSO to three prototypical targeting groups and administered them to a mouse model of EPP in order to study their biodistribution, their metabolic stability and their FECH splice-switching ability. The SSOs exhibited distinct distribution profiles, with increased accumulation in liver, kidney, bone marrow and lung. However, they also underwent substantial metabolism, mainly at their linker groups. An SSO bearing a cholesteryl group increased levels of correctly spliced FECH transcript by 80% in the bone marrow. The results provide a promising approach to treat EPP and other disorders originating from splicing dysregulation in the bone marrow.


Assuntos
Ferroquelatase/genética , Oligonucleotídeos/administração & dosagem , Protoporfiria Eritropoética/metabolismo , Splicing de RNA , Albuminas/metabolismo , Animais , Medula Óssea/metabolismo , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Ferroquelatase/metabolismo , Humanos , Células K562 , Camundongos , Oligonucleotídeos/sangue , Oligonucleotídeos/química , Oligonucleotídeos/farmacocinética , Polimorfismo de Nucleotídeo Único , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/terapia , Sítios de Splice de RNA , Distribuição Tecidual
7.
Photodermatol Photoimmunol Photomed ; 36(1): 29-33, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31374130

RESUMO

BACKGROUND: Erythropoietic protoporphyria (EPP) is a semi-dominantly inherited porphyria presenting with photosensitivity during early childhood. Acquired EPP has been reported; however, data regarding this rare disorder are scarce. PURPOSE: To evaluate the characteristics of acquired EPP. METHODS: A comprehensive search of PubMed, Google Scholar, ScienceDirect, and clinicaltrials.gov databases was performed by three reviewers. Studies describing patients with acquired EPP were included. Additionally, we present an index case of a 26-year-old patient who acquired clinically and biochemically typical EPP in association with myelodysplastic syndrome (MDS). RESULTS: We included 20 case reports describing 20 patients. Most (80%) patients were male of mean age 58 ± 13 years. In all patients, acquired EPP was associated with hematological disease, most commonly MDS (85%) followed by myeloproliferative disease (10%). In 86% of cases, hematological disease led to abnormality or somatic mutation in chromosome 18q (the locus of the ferrochelatase gene). The mean erythrocyte protoporphyrin IX concentration was very high (4286 µg/dL). Most (90%) patients presented with photosensitivity, 20% experienced blistering, and 25% presented with hepatic insufficiency, both uncommon in EPP. In 55% of patients, hematological disease was diagnosed after occurrence of cutaneous symptoms. Beta-carotene led to partial control of symptoms in 5 patients and resolution in another patient. Azacitidine treatment of MDS led to resolution of cutaneous symptoms in three patients. CONCLUSION: We present the distinct features of acquired EPP and highlight that any patient presenting with new-onset photosensitivity, irrespective of age should be evaluated for porphyria.


Assuntos
Azacitidina/uso terapêutico , Síndromes Mielodisplásicas , Transtornos de Fotossensibilidade , Protoporfiria Eritropoética , beta Caroteno/uso terapêutico , Adulto , Idoso , Cromossomos Humanos Par 18/genética , Cromossomos Humanos Par 18/metabolismo , Eritrócitos/metabolismo , Feminino , Ferroquelatase/genética , Ferroquelatase/metabolismo , Loci Gênicos , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Transtornos de Fotossensibilidade/induzido quimicamente , Transtornos de Fotossensibilidade/tratamento farmacológico , Transtornos de Fotossensibilidade/genética , Transtornos de Fotossensibilidade/metabolismo , Protoporfiria Eritropoética/induzido quimicamente , Protoporfiria Eritropoética/tratamento farmacológico , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/metabolismo , Protoporfirinas/genética , Protoporfirinas/metabolismo
8.
Sci Adv ; 5(9): eaaw6127, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31555729

RESUMO

Erythropoietic protoporphyria (EPP) is an inherited disease caused by loss-of-function mutations of ferrochelatase, an enzyme in the heme biosynthesis pathway that converts protoporphyrin IX (PPIX) into heme. PPIX accumulation in patients with EPP leads to phototoxicity and hepatotoxicity, and there is no cure. Here, we demonstrated that the PPIX efflux transporter ABCG2 (also called BCRP) determines EPP-associated phototoxicity and hepatotoxicity. We found that ABCG2 deficiency decreases PPIX distribution to the skin and therefore prevents EPP-associated phototoxicity. We also found that ABCG2 deficiency protects against EPP-associated hepatotoxicity by modulating PPIX distribution, metabolism, and excretion. In summary, our work has uncovered an essential role of ABCG2 in the pathophysiology of EPP, which suggests the potential for novel strategies in the development of therapy for EPP.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Protoporfiria Eritropoética , Protoporfirinas , Pele , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Ferroquelatase/genética , Ferroquelatase/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Mutantes , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/metabolismo , Protoporfiria Eritropoética/patologia , Protoporfiria Eritropoética/fisiopatologia , Protoporfirinas/genética , Protoporfirinas/metabolismo , Pele/metabolismo , Pele/patologia
9.
Biochem Pharmacol ; 154: 474-481, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29906468

RESUMO

Erythropoietic protoporphyria (EPP) is a genetic disease that results from the defective mutation in the gene encoding ferrochelatase (FECH), the enzyme that converts protoporphyrin IX (PPIX) to heme. Liver injury and even liver failure can occur in EPP patients because of PPIX accumulation in the liver. The current study profiled the liver metabolome in an EPP mouse model caused by a Fech mutation (Fech-mut). As expected, we observed the accumulation of PPIX in the liver of Fech-mut mice. In addition, our metabolomic analysis revealed the accumulation of bile acids and ceramide (Cer) in the liver of Fech-mut mice. High levels of bile acids and Cer are toxic to the liver. Furthermore, we found that the major phosphatidylcholines (PC) in the liver and the ratio of total PC to PPIX in the bile were decreased in Fech-mut mice compared to wild type mice. A decrease of the ratio of PC to PPIX in the bile can potentiate the accumulation of PPIX in the liver because PC increases PPIX solubility and excretion. These metabolomic findings suggest that the accumulation of PPIX, together with the disruption of the homeostasis of bile acids, Cer, and PC, contributes to EPP-associated liver injury.


Assuntos
Modelos Animais de Doenças , Fígado/metabolismo , Metabolômica/métodos , Protoporfiria Eritropoética/metabolismo , Animais , Masculino , Camundongos , Camundongos Transgênicos , Protoporfiria Eritropoética/genética
11.
Zhonghua Bing Li Xue Za Zhi ; 46(10): 704-707, 2017 Oct 08.
Artigo em Chinês | MEDLINE | ID: mdl-29050073

RESUMO

Objective: To investigate the clinicopathologic features of the erythropoietic protoporphyria (EPP) with liver involvement. Methods: The clinical findings and hepatic biopsy of 3 cases of EPP diagnosed between July, 2011 to August, 2014 with liver involvement were reviewed, with relevant literature review. Results: All patients presented with persistent and refractory abdominal pain, with obvious jaundice and deranged liver function. Imaging showed homogeneous hepatomegaly in all patients. Histologically, the hepatocytes were edematous, and contained numerous cytoplasmic globular brown pigments and bile pigments, which were also found in Kupffer cells, in the bile canaliculi and in some of dilated sinusoid. The pigments were of different sizes and showed uneven distribution. Some pigments showed bright red or yellow birefringence with a distinctive central maltese cross configuration on polarizing microscopy. Furthermore, some hepatocytes showed piecemeal necrosis and steatosis, the portal tracts were usually infiltrated by lymphocytes, with fibroplasia and biliary ductular reaction. There was no dilatation of intrahepatic bile ducts. Conclusion: Full understanding of the clinical and pathological features of EPP with liver involvement can help to recognize this small group of patients, and to offer proper effective treatments.


Assuntos
Protoporfiria Eritropoética/patologia , Protoporfirinas , Pigmentos Biliares/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Fígado/patologia , Fígado/fisiopatologia , Necrose , Protoporfiria Eritropoética/metabolismo
12.
J Inherit Metab Dis ; 40(3): 433-441, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28185024

RESUMO

Patients with erythropoietic protoporphyria (EPP) have reduced activity of the enzyme ferrochelatase that catalyzes the insertion of iron into protoporphyrin IX (PPIX) to form heme. As the result of ferrochelatase deficiency, PPIX accumulates and causes severe photosensitivity. Among different patients, the concentration of PPIX varies considerably. In addition to photosensitivity, patients frequently exhibit low serum iron and a microcytic hypochromic anemia. The aims of this study were to (1) search for factors related to PPIX concentration in EPP, and (2) characterize anemia in EPP, i.e., whether it is the result of an absolute iron deficiency or the anemia of chronic disease (ACD). Blood samples from 67 EPP patients (51 Italian and 16 Swiss) and 21 healthy volunteers were analyzed. EPP patients had lower ferritin (p = 0.021) and hepcidin (p = 0.031) concentrations and higher zinc-protoporphyrin (p < 0.0001) and soluble-transferrin-receptor (p = 0.0007) concentrations compared with controls. This indicated that anemia in EPP resulted from an absolute iron deficiency. Among EPP patients, PPIX concentrations correlated with both growth differentiation factor (GDF) 15 (p = 0.012) and male gender (p = 0.015). Among a subgroup of patients who were iron replete, hemoglobin levels were normal, which suggested that iron but not ferrochelatase is the limiting factor in heme synthesis of individuals with EPP.


Assuntos
Fator 15 de Diferenciação de Crescimento/metabolismo , Ferro/metabolismo , Protoporfiria Eritropoética/metabolismo , Anemia Hipocrômica/metabolismo , Estudos de Casos e Controles , Eritrócitos/metabolismo , Feminino , Ferritinas/metabolismo , Ferroquelatase/metabolismo , Hemoglobinas/metabolismo , Hepcidinas/metabolismo , Humanos , Masculino , Transtornos de Fotossensibilidade/metabolismo , Protoporfirinas/metabolismo , Índice de Gravidade de Doença
14.
Clin Genet ; 89(1): 20-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25615817

RESUMO

X-linked protoporphyria (XLP), a rare erythropoietic porphyria, results from terminal exon gain-of-function mutations in the ALAS2 gene causing increased ALAS2 activity and markedly increased erythrocyte protoporphyrin levels. Patients present with severe cutaneous photosensitivity and may develop liver dysfunction. XLP was originally reported as X-linked dominant with 100% penetrance in males and females. We characterized 11 heterozygous females from six unrelated XLP families and show markedly varying phenotypic and biochemical heterogeneity, reflecting the degree of X-chromosomal inactivation of the mutant gene. ALAS2 sequencing identified the specific mutation and confirmed heterozygosity among the females. Clinical history, plasma and erythrocyte protoporphyrin levels were determined. Methylation assays of the androgen receptor and zinc-finger MYM type 3 short tandem repeat polymorphisms estimated each heterozygotes X-chromosomal inactivation pattern. Heterozygotes with equal or increased skewing, favoring expression of the wild-type allele had no clinical symptoms and only slightly increased erythrocyte protoporphyrin concentrations and/or frequency of protoporphyrin-containing peripheral blood fluorocytes. When the wild-type allele was preferentially inactivated, heterozygous females manifested the disease phenotype and had both higher erythrocyte protoporphyrin levels and circulating fluorocytes. These findings confirm that the previous dominant classification of XLP is inappropriate and genetically misleading, as the disorder is more appropriately designated XLP.


Assuntos
Genes Ligados ao Cromossomo X , Doenças Genéticas Ligadas ao Cromossomo X/diagnóstico , Doenças Genéticas Ligadas ao Cromossomo X/genética , Fenótipo , Protoporfiria Eritropoética/diagnóstico , Protoporfiria Eritropoética/genética , Inativação do Cromossomo X , Alelos , Eritrócitos/metabolismo , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Genótipo , Humanos , Masculino , Mutação , Proteínas Nucleares/genética , Linhagem , Porfirinas/metabolismo , Protoporfiria Eritropoética/metabolismo , Protoporfirinas/metabolismo , Receptores Androgênicos/genética
15.
Eur J Clin Invest ; 45(10): 1032-41, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26199063

RESUMO

BACKGROUND: Erythropoietic protoporphyria (EPP) and X-linked protoporphyria (XLP) are genetic abnormalities of heme synthesis that result in excess production of protoporphyrin and that manifest as severe photosensitivity. These disorders are often associated with iron deficiency anaemia (IDA). Our aim was to determine whether hepcidin is increased in EPP/XLP patients, resulting in decreased enteral iron absorption and IDA. MATERIAL AND METHODS: Eight subjects with EPP, one with XLP and nine controls had baseline blood and urine samples collected, and thereafter were given oral ferrous sulphate (660 mg). Post-iron blood and urine samples were collected at 2, 4, 6 and 8 h. Blood counts, serum cytokines, ferritin and iron studies were analysed at baseline. Serum iron studies, serum and urine hepcidin, and erythropoietin (Epo) were analysed at baseline and subsequent time points. RESULTS: At baseline, EPP-XLP subjects had lower mean blood haemoglobin (13·9/15·3 g/dL) and serum ferritin (31·6/115 ng/mL) than controls. Serum iron levels increased markedly in both cohorts. Mean serum and urine hepcidin levels were significantly lower in the EPP-XLP group at 4 and 8 h post-iron (serum - 4 h, 3·79/26·6, 8 h, 5·79/34·6 nM; urine - 4 h, 0·85/2·50, 8 h, 1·44/6·63 nM/mM creatinine). Serum cytokines and Epo were normal and not different between groups. CONCLUSIONS: We conclude that serum and urine hepcidin are not inappropriately increased in EPP/XLP subjects at baseline and do not increase over time as serum iron increases after oral ferrous sulphate. Levels of serum cytokines and Epo are normal in EPP/XLP. The molecular basis for the iron-deficient phenotype in EPP/XLP remains unknown.


Assuntos
Hepcidinas/metabolismo , Ferro/metabolismo , Protoporfiria Eritropoética/metabolismo , Adulto , Idoso , Estudos de Casos e Controles , Citocinas/metabolismo , Eritropoetina/metabolismo , Ferritinas/metabolismo , Homeostase/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade
16.
Biomed Res Int ; 2015: 436319, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25945334

RESUMO

The effect of bile acids administration to an experimental mice model of Protoporphyria produced by griseofulvin (Gris) was investigated. The aim was to assess whether porphyrin excretion could be accelerated by bile acids treatment in an attempt to diminish liver damage induced by Gris. Liver damage markers, heme metabolism, and oxidative stress parameters were analyzed in mice treated with Gris and deoxycholic (DXA), dehydrocholic (DHA), chenodeoxycholic, or ursodeoxycholic (URSO). The administration of Gris alone increased the activities of glutathione reductase (GRed), superoxide dismutase (SOD), alkaline phosphatase (AP), gamma glutamyl transpeptidase (GGT), and glutathione-S-transferase (GST), as well as total porphyrins, glutathione (GSH), and cytochrome P450 (CYP) levels in liver. Among the bile acids studied, DXA and DHA increased PROTO IX excretion, DXA also abolished the action of Gris, reducing lipid peroxidation and hepatic GSH and CYP levels, and the activities of GGT, AP, SOD, and GST returned to control values. However, porphyrin accumulation was not prevented by URSO; instead this bile acid reduced ALA-S and the antioxidant defense enzymes system activities. In conclusion, we postulate that DXA acid would be more effective to prevent liver damage induced by Gris.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Protoporfiria Eritropoética/tratamento farmacológico , Animais , Catalase/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Ácido Quenodesoxicólico/administração & dosagem , Ácido Desidrocólico/administração & dosagem , Ácido Desoxicólico/administração & dosagem , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Glutationa Transferase/metabolismo , Griseofulvina/toxicidade , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Porfirinas/metabolismo , Protoporfiria Eritropoética/induzido quimicamente , Protoporfiria Eritropoética/metabolismo , Superóxido Dismutase/metabolismo , Ácido Ursodesoxicólico/administração & dosagem
18.
Br J Dermatol ; 171(2): 412-4, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24701996

RESUMO

Although erythropoietic protoporphyria (EPP) is relatively uncommon, affecting approximately 1 in 140 000 individuals in the U.K., it is an important disease not to miss owing to the risk of acute severe liver disease in 2% of cases. EPP occurs with clinical and histological changes in the skin associated with free-radical-associated dermal vascular damage. This also mediates the painful photosensitivity. Severe and disfiguring hyaline deposition is extremely rare. We demonstrate that severe EPP can cause disfiguring hyaline infiltration of the skin on the hands and face, which sheds light on the mechanism of photosensitivity in EPP; it must also be differentiated from conditions such as lipoid proteinosis.


Assuntos
Dermatoses Faciais/etiologia , Hialina/metabolismo , Transtornos de Fotossensibilidade/etiologia , Protoporfiria Eritropoética/complicações , Dermatoses Faciais/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Transtornos de Fotossensibilidade/metabolismo , Protoporfiria Eritropoética/metabolismo
19.
Blood Cells Mol Dis ; 51(3): 151-61, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23787363

RESUMO

Erythropoietic protoporphyria (EPP) results from partial deficiency of ferrochelatase (FECH). Genetically, EPP patients differ from asymptomatic mutation carriers at the unmutated FECH allele, the expression of which is modulated by single nucleotide polymorphism IVS3-48C/T. The IVS3-48C genotype, which is present among patients, leads to correct splicing of 60% of the pre-mRNA and to alternative splicing of 40%, the latter mRNA-product being destroyed by nonsense-mediated decay. An IVS3-48T genotype generates 80% correct and 20% aberrant products. Our study demonstrated that under iron deficient conditions, the aberrant splice product was increased to 56% and 50% of total FECH mRNA in erythroleukemic K562 and lymphoblastoid cell lines, respectively, both being homozygous for IVS3-48T. Concomitantly, FECH protein was decreased. Iron deficiency had less effect on the FECH splice ratio in an IVS3-48C/C lymphoblastoid cell line. Effects similar to iron deficiency were generated by siRNA knockdown of either splicing factor U2AF(65) or Fe(II)- and 2-oxoglutarate-dependent dioxygenase Jumonji domain-containing protein 6 (Jmjd6), which interacts with U2AF(65) by lysyl-hydroxylation. Based on these results, we propose that the availability of iron, a co-factor of Jmjd6, modulates U2AF(65)-lysyl-hydroxylation. This in turn, influences the relative amounts of correct and aberrant FECH mRNA splice products and thus, regulates the FECH enzyme activity.


Assuntos
Processamento Alternativo , Ferroquelatase/genética , Ferro/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Ácidos Cetoglutáricos/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Sequência de Bases , Estudos de Casos e Controles , Linhagem Celular , Cobalto/farmacologia , Desferroxamina/metabolismo , Desferroxamina/farmacologia , Ferroquelatase/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ordem dos Genes , Inativação Gênica , Genótipo , Humanos , Íntrons , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Células K562 , Dados de Sequência Molecular , Mutação , Polimorfismo de Nucleotídeo Único , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Fator de Processamento U2AF
20.
Hepatology ; 56(1): 322-31, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22334478

RESUMO

UNLABELLED: Mallory-Denk bodies (MDBs) are hepatocyte inclusions commonly seen in steatohepatitis. They are induced in mice by feeding 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) for 12 weeks, which also causes porphyrin accumulation. Erythropoietic protoporphyria (EPP) is caused by mutations in ferrochelatase (fch), and a fraction of EPP patients develop liver disease that is phenocopied in Fech(m1Pas) mutant (fch/fch) mice, which have an inactivating fch mutation. fch/fch mice develop spontaneous MDBs, but the molecular factors involved in their formation and whether they relate to DDC-induced MDBs are unknown. We tested the hypothesis that fch mutation creates a molecular milieu that mimics experimental drug-induced MDBs. In 13- and 20-week-old fch/fch mice, serum alkaline phosphatase, alanine aminotransferase, and bile acids were increased. The 13-week-old fch/fch mice did not develop histologically evident MDBs but manifested biochemical alterations required for MDB formation, including increased transglutaminase-2 and keratin overexpression, with a greater keratin 8 (K8)-to-keratin 18 (K18) ratio, which are critical for drug-induced MDB formation. In 20-week-old fch/fch mice, spontaneous MDBs were readily detected histologically and biochemically. Short-term (3-week) DDC feeding markedly induced MDB formation in 20-week-old fch/fch mice. Under basal conditions, old fch/fch mice had significant alterations in mitochondrial oxidative-stress markers, including increased protein oxidation, decreased proteasomal activity, reduced adenosine triphosphate content, and Nrf2 (redox sensitive transcription factor) up-regulation. Nrf2 knockdown in HepG2 cells down-regulated K8, but not K18. CONCLUSION: Fch/fch mice develop age-associated spontaneous MDBs, with a marked propensity for rapid MDB formation upon exposure to DDC, and therefore provide a genetic model for MDB formation. Inclusion formation in the fch/fch mice involves oxidative stress which, together with Nrf2-mediated increase in K8, promotes MDB formation.


Assuntos
Queratina-18/metabolismo , Corpos de Mallory/metabolismo , Corpos de Mallory/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/fisiologia , Protoporfiria Eritropoética/metabolismo , Protoporfiria Eritropoética/patologia , Análise de Variância , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Masculino , Corpos de Mallory/genética , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/genética , Distribuição Aleatória , Sensibilidade e Especificidade , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...