Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Liver Int ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39011756

RESUMO

The erythropoietic protoporphyrias consist of three ultra-rare genetic disorders of the erythroid heme biosynthesis, including erythropoietic protoporphyria (EPP1), X-linked protoporphyria (XLEPP) and CLPX-protoporphyria (EPP2), which all lead to the accumulation of protoporphyrin IX (PPIX) in erythrocytes. Affected patients usually present from early childhood with episodes of severe phototoxic pain in the skin exposed to visible light. The quantification of PPIX in erythrocytes with a metal-free PPIX ≥3 times the upper limit of normal confirms the diagnosis. Protoporphyria-related complications include liver failure, gallstones, mild anaemia and vitamin D deficiency with reduced bone mineral density. The management is focused on preventing phototoxic reactions and treating the complications. Vitamin D should be supplemented, and DEXA scans in adults should be considered. In EPP1, even in cases of biochemically determined iron deficiency, supplementation of iron may stimulate PPIX production, resulting in an increase in photosensitivity and the risk of cholestatic liver disease. However, for patients with XLEPP, iron supplementation can reduce PPIX levels, phototoxicity and liver damage. Because of its rarity, there is little data on the management of EPP-related liver disease. As a first measure, any hepatotoxins should be eliminated. Depending on the severity of the liver disease, phlebotomies, exchange transfusions and ultimately liver transplantation with subsequent haematopoietic stem cell transplantation (HSCT) are therapeutic options, whereby multidisciplinary management including porphyria experts is mandatory. Afamelanotide, an alpha-melanocyte-stimulating hormone analogue, is currently the only approved specific treatment that increases pain-free sunlight exposure and quality of life.

3.
Front Genet ; 13: 993453, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36386813

RESUMO

Background and aims: Porphyrias constitute a group of rare genetic diseases due to various, mostly autosomal dominant mutations, causing enzymatic deficiency in heme biosynthesis. As a result, neurotoxic porphyrin precursors and light-sensitive porphyrins accumulate, while dysfunction in their targets determines the disease symptoms. Variegate porphyria (VP), one of the acute hepatic porphyrias, is caused by a protoporphyrinogen oxidase (PPOX) mutation. During acute attacks, among other factors, triggered by drugs, stressors, or fasting, an increase in urinary and fecal porphobilinogen (PBG), aminolevulinic acid (ALA), and porphyrins occurs, damaging the autonomous, peripheral, or central nervous system. The disease remains often latent or displays minimal symptoms usually overlooked, exposing undiagnosed patients to potentially serious complications in the presence of the aforementioned triggers. Case report: This 46-year-old woman presented, some days after a bariatric surgery, with severe flaccid tetraparesis and neuropathic pain, initially misdiagnosed as a functional neurological disorder. The severe axonal sensorimotor polyneuropathy led to further investigations, disclosing high urinary porphobilinogen, ALA, and porphyrin levels due to a new PPOX mutation. Retrospectively, it appeared that the patient had had typical VP symptoms (abdominal pain, fragile skin, and dark urine episodes) for years prior to the surgery. Treated with carbohydrate load, neurorehabilitation, and analgesics, she slowly recovered to full mobility, with partial autonomy in her daily life activities, although fatigue and severe pain persisted, preventing her from returning to work. Conclusion: This case documents gastric bypass surgery as a trigger of severe VP invalidating neurological symptoms and illustrates how the delayed diagnosis and post-interventional complications could have been prevented by screening for porphyria cardinal symptoms prior to the intervention. Likewise, this cost-effective screening should be performed before any treatment influencing the diet, which would dramatically improve the porphyria diagnosis rate and outcome.

4.
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
5.
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
6.
Mol Genet Metab ; 128(3): 304-308, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31076252

RESUMO

Deficiency in ferrochelatase (FECH), the last enzyme in the heme biosynthetic pathway, leads to an accumulation of protoporphyrin IX (PPIX) that causes a severely painful phototoxic reaction of the skin in patients with erythropoietic protoporphyria (EPP). Besides phototoxicity of the skin, EPP patients often present with symptoms of iron deficiency in form of a microcytic and hypochromic anemia with low serum iron and ferritin. In addition, elevated aminolevulinic acid synthase 2 (ALAS2) both at the mRNA and protein levels have been observed among EPP patients. ALAS is the first enzyme in the pathway and exists in two isoforms, whereby the isoform 2 (ALAS2) is expressed exclusively in erythropoiesis. The mRNA of ALAS2 contains an iron response element (IRE) at its 5'UTR. When iron is limited, iron response element binding protein 2 (IRP2) binds to the IRE of ALAS2 mRNA and suppresses its translation. In this study, we demonstrated that iron deprivation increased the amount of ALAS2 mRNA as well as the ratio of ALAS2 to FECH mRNAs in cultured erythroleukemic K562 cells. At the protein level, however, iron deprivation in the cell line caused reductions in both enzymes as shown by the Western blot analysis. A comparable increase in the ratio of ALAS2 to FECH mRNAs was also found in EPP patients indicating an imbalance in heme biosynthesis. As iron cannot be completely missing from an organism, we assume that in EPP patients, a certain amount of ALAS2 mRNA is translated despite a partial deficiency of FECH. The increase in ALAS2 enzyme contributes to the accumulation in PPIX in the patients. Targeted inhibition of ALAS2 could therefore be a treatment option for EPP.


Assuntos
5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Ferro/metabolismo , Protoporfiria Eritropoética/enzimologia , Vias Biossintéticas , Ferroquelatase/genética , Humanos , Ferro/sangue , Proteína 2 Reguladora do Ferro/metabolismo , Proteínas Reguladoras de Ferro/metabolismo , Células K562 , Protoporfiria Eritropoética/terapia , Protoporfirinas/metabolismo
7.
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
9.
Blood Cells Mol Dis ; 54(1): 71-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25179834

RESUMO

The activity of the erythroid-specific isoenzyme of 5-aminolevulinic acid synthase (ALAS2), the first and rate-limiting enzyme in heme biosynthesis, is down-regulated during iron-deficiency. Ferrochelatase (FECH), the last enzyme of this pathway, inserts iron into protoporphyrin IX (PPIX) to form heme. Patients with erythropoietic protoporphyria (EPP), an inherited deficiency in FECH, often show signs of iron deficiency in addition to phototoxicity which is caused by PPIX accumulation. However, iron supplementation often leads to exacerbation of phototoxicity. We report three EPP patients who had reduced erythrocytic PPIX concentrations when they were iron-deficient and their microcytic and hypochromic anemia deteriorated. Additionally, we found a significant increase in the amount of ALAS2 mRNA and protein among EPP patients. To verify the connection between FECH deficiency and ALAS2 over-expression, we inhibited FECH in cultured cells and found a subsequent increase in ALAS2 mRNA. We conclude that the primary deficiency in ferrochelatase leads to a secondary increase in ALAS2 expression. The combined action of these two enzymes within the heme biosynthetic pathway contributes to the accumulation of PPIX. Furthermore, we hypothesize that EPP patients may benefit from a mild iron deficiency since it would limit PPIX production by restricting ALAS2 over-expression.


Assuntos
5-Aminolevulinato Sintetase/biossíntese , Eritrócitos/enzimologia , Regulação Enzimológica da Expressão Gênica , Ferro/metabolismo , Protoporfiria Eritropoética/enzimologia , Protoporfirinas/metabolismo , Adolescente , Adulto , Eritrócitos/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Protoporfiria Eritropoética/patologia , RNA Mensageiro/biossíntese
10.
J Hepatol ; 62(3): 734-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25445397

RESUMO

Variegate porphyria (VP) and acute intermittent porphyria (AIP), the two most common types of acute porphyrias (AHPs), result from a partial deficiency of protoporphyrinogen oxidase (PPOX) and hydroxymethylbilane synthase (HMBS), respectively. A rare but serious complication in the AHPs is hepatocellular carcinoma (HCC). However, the underlying pathomechanisms are yet unknown. We performed DNA sequence analysis in cancerous and non-cancerous liver tissue of a VP and an AIP patient, both with HCC. In samples of both cancerous and non-cancerous liver tissues from the patients, we identified the underlying PPOX and HMBS germline mutations, c.1082dupC and p.G111R, respectively. Additionally, we detected a second somatic mutation, only in the cancer tissue i.e., p.L416X in the PPOX gene of the VP patient and p.L220X in the HMBS gene of the AIP patient, both located in trans to the respective germline mutations. Both somatic mutations were not detected in 10 non-porphyria-associated HCCs. Our data demonstrate that in the hepatic cancer tissue of AHP patients, somatic second-hit mutations result in nearly complete inactivation of the enzymes catalyzing major steps in the heme biosynthetic pathway. Both PPOX and HMBS, which might act as tumor suppressors, play a crucial role in the development of HCC in these individuals.


Assuntos
Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/genética , Flavoproteínas/genética , Hidroximetilbilano Sintase/genética , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/genética , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Mutação , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/genética , Porfiria Variegada/complicações , Porfiria Variegada/genética , Protoporfirinogênio Oxidase/genética , Idoso , Idoso de 80 Anos ou mais , Carcinoma Hepatocelular/enzimologia , Feminino , Mutação em Linhagem Germinativa , Humanos , Neoplasias Hepáticas/enzimologia , Porfiria Aguda Intermitente/enzimologia , Porfiria Variegada/enzimologia , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
11.
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
12.
J Pharm Biomed Anal ; 75: 192-8, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23277150

RESUMO

The tridecapeptide afamelanotide (Scenesse®) is a congener of α-melanocyte stimulating hormone (α-MSH). Upon binding to the melanocortin 1 receptor (MC1R) on the surface of pigment cells of the skin, the melanocytes, α-MSH or afamelanotide trigger the synthesis of cAMP, which stimulates the synthesis of melanin and therefore induces skin tanning. In a recent trial, afamelanotide administered as controlled release implants protected erythropoietic protoporphyria (EPP) patients from sunlight induced phototoxic skin reactions. Administration of biological therapeutic peptides may elicit unwanted immunogenic responses in recipients of these products. Although in a previous study using ELISA technique we excluded any newly developed immunogenicity during prolonged exposure to afamelanotide, we confirmed the previously published existence of low titers of antibodies against α-MSH in drug-naïve individuals that cross-reacted with afamelanotide. In order to investigate whether such antibodies are neutralizing, i.e. could block the biological effect of afamelanotide, we developed a cell culture-based bioassay. The basis of our assay was the measurement of afamelanotide-induced cAMP formation in a strain of the B16 mouse melanoma cell line, G4F-7, expressing the transfected human MC1R. Average half-effective concentrations of the natural hormone α-MSH and its congener afamelanotide were 38.8 ± 10.6 and 10.9 ± 7.17 nM (n=5), respectively. Neutralizing antibodies would reduce the cAMP formation. Two neutralizing anti-α-MSH antibodies served as positive controls. cAMP formation in the G4F-7 cells after addition of sera of drug-naïve (n=6) and of drug-exposed EPP patients (n=17) was significantly lower than after that from healthy volunteers (n=13). There was no difference between drug-naïve and drug-exposed patients. Using forskolin as a hormone-independent stimulator of cAMP formation, we excluded an unspecific interference of EPP sera with cAMP formation. We conclude that afamelanotide even after prolonged application to EPP patients did not elicit neutralizing antibodies. Further, the low titer immunoreactivity observed in sera of some drug-naïve individuals had no effect on the biological activity of afamelanotide.


Assuntos
Anticorpos Neutralizantes/análise , Fármacos Dermatológicos/antagonistas & inibidores , Porfiria Eritropoética/imunologia , alfa-MSH/análogos & derivados , Animais , Linhagem Celular Tumoral , Reações Cruzadas , AMP Cíclico/metabolismo , Fármacos Dermatológicos/farmacologia , Fármacos Dermatológicos/uso terapêutico , Humanos , Hipopigmentação/etiologia , Hipopigmentação/prevenção & controle , Melanócitos/efeitos dos fármacos , Melanócitos/imunologia , Melanócitos/metabolismo , Camundongos , Monitorização Imunológica , Concentração Osmolar , Porfiria Eritropoética/sangue , Porfiria Eritropoética/tratamento farmacológico , Porfiria Eritropoética/fisiopatologia , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/metabolismo , Proteínas Recombinantes/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , alfa-MSH/antagonistas & inibidores , alfa-MSH/metabolismo , alfa-MSH/farmacologia , alfa-MSH/uso terapêutico
13.
J Inherit Metab Dis ; 33 Suppl 3: S465-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21069463

RESUMO

Erythropoietic protoporphyria (EPP) is a hereditary disorder caused by deficiency of ferrochelatase, the last enzyme in the heme biosynthetic pathway. The majority of EPP patients present with a clinical symptom of painful phototoxicity. Liver damage, the most serious complication of EPP, occurs in <5% of the patients. This report describes a case of an EPP patient who complained of worsening cutaneous symptoms, nervousness, and insomnia. Laboratory tests showed highly increased protoporphyrin concentration in erythrocytes and elevated serum transaminases that are indicative of EPP-related liver damage. The subsequent finding of decreased serum thyroid-stimulating hormone (TSH) and increased free triiodothyronine (FT3) and free thyroxine (FT4) concentrations, as well antibodies against both thyroid peroxidase (TPO) and TSH receptors, led to the diagnosis of Graves' disease. The patient received 500 MBq of radioiodine (I(131)). Three months after the radioactive iodine therapy, the thyroid volume was reduced to 30% of pretherapeutic volume. Although the patient was slightly hypothyroidic, his liver enzymes returned to normal, his erythrocytic protoporphyrin concentration dropped fivefold, and his skin symptoms improved dramatically. The coexistence of Graves' disease and EPP is a statistically rare event as, besides our patient, there was one additional case reported in the literature. Although the exact mechanism whereby Graves' disease interacts with EPP is yet to be explored, we recommend testing thyroid function in EPP patients with liver complication to exclude hyperthyroidism as a potential cause.


Assuntos
Doença de Graves/complicações , Hepatopatias/etiologia , Porfiria Eritropoética/complicações , Adulto , Autoantígenos/imunologia , Biomarcadores/sangue , Progressão da Doença , Eritrócitos/metabolismo , Doença de Graves/sangue , Doença de Graves/diagnóstico , Doença de Graves/imunologia , Doença de Graves/radioterapia , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/sangue , Iodeto Peroxidase/imunologia , Radioisótopos do Iodo/uso terapêutico , Proteínas de Ligação ao Ferro/imunologia , Hepatopatias/sangue , Hepatopatias/diagnóstico , Masculino , Porfiria Eritropoética/sangue , Porfiria Eritropoética/diagnóstico , Protoporfirinas/sangue , Hormônios Tireóideos/sangue , Fatores de Tempo , Transaminases/sangue , Resultado do Tratamento
16.
Photodiagnosis Photodyn Ther ; 6(1): 12-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19447367

RESUMO

BACKGROUND: The in vitro experiments described in this study were aimed at exploring a synergistic effect between 5-aminolevulinic acid (5-ALA) induced protoporphyrin IX (PpIX) and hypericin. In a previous study, enhanced phototoxicity was observed in a patient during a clinical study on 5-ALA-based photodynamic tumor localization of breast cancer. This patient ingested a hypericin containing plant extract in parallel to orally applied 5-ALA. METHODS: Human endometrial cancer cells (HEC-1A) were treated with 0.5mM of 5-ALA and 60 nM of hypericin, either separately or combined. Colony formation was assessed after illumination of the cells with both red (635 nm) and white light (400-800 nm) at a dose of 2.5 J/cm(2). Porphyrin metabolites were quantified by HPLC in cells treated with photosensitizers without subsequent illumination. RESULTS: After white light illumination, cells treated with a combination of 5-ALA and hypericin had a significant reduction in colony formation compared with cells treated with 5-ALA only. No significantly enhanced toxicity was found with red light and the 5-ALA plus hypericin combination. In addition, cells treated with both 5-ALA and hypericin tended to produce more PpIX than cells treated with 5-ALA only. CONCLUSIONS: This study demonstrated that treatment of endometrial cancer cells with both 5-ALA and hypericin followed by illumination with white light induced a significantly higher phototoxicity as revealed by colony formation. This setting which generated an in vitro effect similar to the patient's situation, might be applied in the future as an affordable and effective photodynamic therapy (PDT) modality.


Assuntos
Ácido Aminolevulínico/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Neoplasias do Endométrio/fisiopatologia , Perileno/análogos & derivados , Protoporfirinas/administração & dosagem , Antracenos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/patologia , Feminino , Humanos , Luz , Perileno/administração & dosagem , Fármacos Fotossensibilizantes/administração & dosagem
17.
Swiss Med Wkly ; 139(13-14): 198-206, 2009 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-19350426

RESUMO

BACKGROUND: The porphyrias, a group of seven metabolic disorders in the haem biosynthesis, can be classified into acute and non-acute porphyrias. A common symptom of acute porphyrias is severe acute abdominal pain, whereas cutaneous photosensitivity can occur in both acute and non-acute porphyrias. All porphyrias, except for sporadic porphyria cutanea tarda (sPCT), are hereditary disorders caused by mutations in the respective genes. We present porphyria cases documented in our porphyria centre during the past 15 years. METHODS: Diagnosis was based on clinical symptoms and biochemical analyses. Mutation analysis was performed in patients/families with a confirmed hereditary porphyria. RESULTS AND CONCLUSIONS: As the porphyria specialist centre of Switzerland, we perform the specialized analyses required for the diagnosis of all types of porphyrias, and give advice to patients, physicians and other laboratories. We therefore estimated that our data cover 80-90% of all diagnosed Swiss cases. A total of 217 patients from 170 families were diagnosed including, 111 acute intermittent porphyria, 45 erythropoietic protoporphyria, 30 variegate porphyria, 21 sPCT, five congenital erythropoietic porphyria, four hereditary coproporphyria and one hepatoerythropoietic porphyria patient. Systematic monitoring of the patients would allow early detection of the potential life-threatening complications such as hepatocellular carcinoma and renal insufficiency in acute porphyrias, and liver failure in EPP. Seventy-five percent of all families underwent genetic testing. Identification of pre-symptomatic mutation carriers so that these individuals and their physicians can be consulted with safety on drug use and other preventive measures, is important in managing acute porphyrias. The unique phenomenon of founder mutations in the Swiss population is also discussed.


Assuntos
Porfiria Eritropoética/epidemiologia , Porfiria Eritropoética/genética , Carcinoma Hepatocelular/epidemiologia , Carcinoma Hepatocelular/genética , Feminino , Ferroquelatase/genética , Flavoproteínas/genética , Efeito Fundador , Heterozigoto , Humanos , Hidroximetilbilano Sintase/genética , Masculino , Proteínas Mitocondriais/genética , Porfiria Eritropoética/complicações , Prevalência , Protoporfirinogênio Oxidase/genética , Suíça/epidemiologia , Uroporfirinogênio III Sintetase/genética
18.
Eur J Haematol ; 76(1): 33-41, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16343269

RESUMO

Mutations of the erythroid-specific 5-aminolaevulinate synthase (ALAS2) gene are known to be responsible for X-linked sideroblastic anaemia (XLSA). An amino acid (AA) substitution for arginine at the 452 AA position of the ALAS2 protein is the most frequent mutation, which has been found in approximately one-quarter of patients with XLSA. Despite its high frequency, there has been no report on the enzymatic activity of Arg452 mutant proteins. In this study, we examined enzymatic activity in vitro of two Arg452 mutants, Arg452Cys and Arg452His, which were found in two new pedigrees of XLSA. While these mutations must be responsible for the clinical phenotype of XLSA in patients, the enzymatic activity and stability of these mutant proteins studied in vitro are indistinguishable from those of the wild type protein. These findings suggest that the Arg452 mutation of the ALAS2 gene by itself does not decrease the enzymatic activity or the stability in vitro, and that there may be an additional factor(s) in the bone marrow, which ensures the full ALAS2 activity in vivo.


Assuntos
5-Aminolevulinato Sintetase/genética , Substituição de Aminoácidos , Anemia Sideroblástica/enzimologia , Eritrócitos/enzimologia , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Mutação Puntual , 5-Aminolevulinato Sintetase/metabolismo , Anemia Sideroblástica/genética , Arginina/genética , Medula Óssea/enzimologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Ligação Genética , Especificidade de Órgãos/genética , Linhagem , Fenótipo
19.
J Am Coll Cardiol ; 41(11): 2004-9, 2003 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-12798573

RESUMO

OBJECTIVES: We sought to assess the mechanism and prognostic value of elevated troponins in patients without acute coronary syndromes (ACS). BACKGROUND: Cardiac troponins are used as specific markers for the diagnosis of ACS. Recent studies reported a considerable number of critically ill patients without ACS as being troponin-positive, especially patients with sepsis, pulmonary embolism, renal failure, and stroke. METHODS: We analyzed 58 consecutive, critically ill patients admitted for reasons other than ACS, according to their troponin status. Thirty-day mortality, left ventricular ejection fraction (LVEF), and a panel of inflammatory cytokines were compared between troponin-positive and troponin-negative patients. Relevant coronary artery disease was excluded either by stress echocardiography or autopsy. RESULTS: Of the 58 critically ill patients, 32 (55%) without evidence of ACS were troponin-positive. Positive troponin levels were associated with higher mortality (22.4% vs. 5.2%, p < 0.018) and a lower LVEF (p = 0.0006). Troponin-positive patients had significantly higher median levels of tumor necrosis factor (TNF)-alpha, its soluble receptor, and interleukin (IL)-6. A subgroup of 10 aplastic patients was troponin-negative at study entry. Three became troponin-positive during leukocyte recovery and subsequently died, whereas all the others stayed troponin-negative and survived. Flow-limiting coronary artery disease was not demonstrable at autopsy or stress echocardiography in 72% of troponin-positive patients. CONCLUSIONS: Elevated troponin is a mortality risk factor for medical intensive care patients admitted for reasons other than ACS. It is associated with decreased left ventricular function and higher levels of TNF-alpha and IL-6.


Assuntos
Doença das Coronárias/sangue , Doença das Coronárias/epidemiologia , Troponina/sangue , Doença Aguda , Adulto , Idoso , Biomarcadores/sangue , Angiografia Coronária , Estado Terminal/mortalidade , Citocinas/metabolismo , Ecocardiografia sob Estresse , Feminino , Infecções por Bactérias Gram-Negativas/sangue , Infecções por Bactérias Gram-Negativas/mortalidade , Infecções por Bactérias Gram-Positivas/sangue , Infecções por Bactérias Gram-Positivas/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Neutrófilos/fisiologia , Fatores de Risco , Choque Séptico/sangue , Choque Séptico/mortalidade , Volume Sistólico/fisiologia , Análise de Sobrevida , Síndrome , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA