Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
1.
Int J Mol Sci ; 22(2)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445793

RESUMEN

Poisoning by high concentrations of dioxin and its related compounds manifests variable toxic symptoms such as general malaise, chloracne, hyperpigmentation, sputum and cough, paresthesia or numbness of the extremities, hypertriglyceridemia, perinatal abnormalities, and elevated risks of cancer-related mortality. Such health hazards are observed in patients with Yusho (oil disease in Japanese) who had consumed rice bran oil highly contaminated with 2,3,4,7,8-pentachlorodibenzofuran, polychlorinated biphenyls, and polychlorinated quaterphenyls in 1968. The blood concentrations of these congeners in patients with Yusho remain extremely elevated 50 years after onset. Dioxins exert their toxicity via aryl hydrocarbon receptor (AHR) through the generation of reactive oxygen species (ROS). In this review article, we discuss the pathogenic implication of AHR in dioxin-induced health hazards. We also mention the potential therapeutic use of herbal drugs targeting AHR and ROS in patients with Yusho.


Asunto(s)
Dioxinas/envenenamiento , Porfirias/inducido químicamente , Porfirias/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Humanos , Especies Reactivas de Oxígeno/metabolismo , Aceite de Salvado de Arroz/efectos adversos
2.
Nat Commun ; 10(1): 5463, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784520

RESUMEN

Iron Regulatory Protein 1 (IRP1) is a bifunctional cytosolic iron sensor. When iron levels are normal, IRP1 harbours an iron-sulphur cluster (holo-IRP1), an enzyme with aconitase activity. When iron levels fall, IRP1 loses the cluster (apo-IRP1) and binds to iron-responsive elements (IREs) in messenger RNAs (mRNAs) encoding proteins involved in cellular iron uptake, distribution, and storage. Here we show that mutations in the Drosophila 1,4-Alpha-Glucan Branching Enzyme (AGBE) gene cause porphyria. AGBE was hitherto only linked to glycogen metabolism and a fatal human disorder known as glycogen storage disease type IV. AGBE binds specifically to holo-IRP1 and to mitoNEET, a protein capable of repairing IRP1 iron-sulphur clusters. This interaction ensures nuclear translocation of holo-IRP1 and downregulation of iron-dependent processes, demonstrating that holo-IRP1 functions not just as an aconitase, but throttles target gene expression in anticipation of declining iron requirements.


Asunto(s)
Enzima Ramificadora de 1,4-alfa-Glucano/genética , Proteínas de Drosophila/genética , Regulación de la Expresión Génica/genética , Proteína 1 Reguladora de Hierro/genética , Hierro/metabolismo , Porfirias/genética , Enzima Ramificadora de 1,4-alfa-Glucano/metabolismo , Transporte Activo de Núcleo Celular , Animales , Regulación hacia Abajo , Drosophila , Proteínas de Drosophila/metabolismo , Ecdisteroides/biosíntesis , Glándulas Endocrinas/metabolismo , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Hemo/metabolismo , Proteína 1 Reguladora de Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Larva/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Porfirias/metabolismo , ARN Mensajero/metabolismo
3.
Cell Mol Gastroenterol Hepatol ; 8(4): 535-548, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31233899

RESUMEN

Genetic porphyrias comprise eight diseases caused by defects in the heme biosynthetic pathway that lead to accumulation of heme precursors. Consequences of porphyria include photosensitivity, liver damage and increased risk of hepatocellular carcinoma, and neurovisceral involvement, including seizures. Fluorescent porphyrins that include protoporphyrin-IX, uroporphyrin and coproporphyrin, are photo-reactive; they absorb light energy and are excited to high-energy singlet and triplet states. Decay of the porphyrin excited to ground state releases energy and generates singlet oxygen. Porphyrin-induced oxidative stress is thought to be the major mechanism of porphyrin-mediated tissue damage. Although this explains the acute photosensitivity in most porphyrias, light-induced porphyrin-mediated oxidative stress does not account for the effect of porphyrins on internal organs. Recent findings demonstrate the unique role of fluorescent porphyrins in causing subcellular compartment-selective protein aggregation. Porphyrin-mediated protein aggregation associates with nuclear deformation, cytoplasmic vacuole formation and endoplasmic reticulum dilation. Porphyrin-triggered proteotoxicity is compounded by inhibition of the proteasome due to aggregation of some of its subunits. The ensuing disruption in proteostasis also manifests in cell cycle arrest coupled with aggregation of cell proliferation-related proteins, including PCNA, cdk4 and cyclin B1. Porphyrins bind to native proteins and, in presence of light and oxygen, oxidize several amino acids, particularly methionine. Noncovalent interaction of oxidized proteins with porphyrins leads to formation of protein aggregates. In internal organs, particularly the liver, light-independent porphyrin-mediated protein aggregation occurs after secondary triggers of oxidative stress. Thus, porphyrin-induced protein aggregation provides a novel mechanism for external and internal tissue damage in porphyrias that involve fluorescent porphyrin accumulation.


Asunto(s)
Porfirias/genética , Porfirias/metabolismo , Porfirias/fisiopatología , Animales , Carcinoma Hepatocelular/metabolismo , Dermatitis Fototóxica/metabolismo , Dermatitis Fototóxica/fisiopatología , Hemo/metabolismo , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones , Oxidación-Reducción , Estrés Oxidativo/fisiología , Trastornos por Fotosensibilidad , Porfirinas/metabolismo , Agregado de Proteínas , Protoporfirinas , Uroporfirinas , Pez Cebra
4.
Cell Mol Gastroenterol Hepatol ; 8(4): 659-682.e1, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31173894

RESUMEN

BACKGROUND & AIMS: Porphyrias are caused by porphyrin accumulation resulting from defects in the heme biosynthetic pathway that typically lead to photosensitivity and possible end-stage liver disease with an increased risk of hepatocellular carcinoma. Our aims were to study the mechanism of porphyrin-induced cell damage and protein aggregation, including liver injury, where light exposure is absent. METHODS: Porphyria was induced in vivo in mice using 3,5-diethoxycarbonyl-1,4-dihydrocollidine or in vitro by exposing human liver Huh7 cells and keratinocytes, or their lysates, to protoporphyrin-IX, other porphyrins, or to δ-aminolevulinic acid plus deferoxamine. The livers, cultured cells, or porphyrin exposed purified proteins were analyzed for protein aggregation and oxidation using immunoblotting, mass spectrometry, and electron paramagnetic resonance spectroscopy. Consequences on cell-cycle progression were assessed. RESULTS: Porphyrin-mediated protein aggregation required porphyrin-photosensitized singlet oxygen and porphyrin carboxylate side-chain deprotonation, and occurred with site-selective native protein methionine oxidation. Noncovalent interaction of protoporphyrin-IX with oxidized proteins led to protein aggregation that was reversed by incubation with acidified n-butanol or high-salt buffer. Phototoxicity and the ensuing proteotoxicity, mimicking porphyria photosensitivity conditions, were validated in cultured keratinocytes. Protoporphyrin-IX inhibited proteasome function by aggregating several proteasomal subunits, and caused cell growth arrest and aggregation of key cell proliferation proteins. Light-independent synergy of protein aggregation was observed when porphyrin was applied together with glucose oxidase as a secondary peroxide source. CONCLUSIONS: Photo-excitable porphyrins with deprotonated carboxylates mediate protein aggregation. Porphyrin-mediated proteotoxicity in the absence of light, as in the liver, requires porphyrin accumulation coupled with a second tissue oxidative injury. These findings provide a potential mechanism for internal organ damage and photosensitivity in porphyrias.


Asunto(s)
Oxígeno/metabolismo , Porfirias/metabolismo , Ácido Aminolevulínico , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular , Deferoxamina , Hemo/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos por Fotosensibilidad , Fármacos Fotosensibilizantes , Porfirias/fisiopatología , Porfirinas/metabolismo , Agregado de Proteínas , Conformación Proteica , Protoporfirinas
5.
J Coll Physicians Surg Pak ; 29(6): S23-S25, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31142411

RESUMEN

Congenital erythropoietic porphyria, also called Gunther's disease, is a very rare genetic autosomal recessive diseaseaffecting less than 1 per 1,000,000 children. Pathogenesis involves genetic mutation encoding uroporphyrinogen-III cosynthase which leads to accumulation of porphyrin in many tissues, leading to extreme skin photosensitivity, red cell lysis, splenomegaly and reduced life expectancy. Herein, we report a 12-year mentally challenged girl with multiple blisters and scars on sun exposed sites since birth. She had hepatomegaly, erythrodontia, severe anaemia with haemolytic blood picture and mildly elevated liver enzymes. Skin biopsy showed deposition of amorphous eosinophilic porphyrins in the dermis, thus confirming a diagnosis of congenital erythropoietic porphyria.


Asunto(s)
Anemia Hemolítica/diagnóstico , Discapacidad Intelectual , Trastornos por Fotosensibilidad/diagnóstico , Porfiria Eritropoyética/diagnóstico , Porfirias/congénito , Biopsia , Niño , Femenino , Hepatomegalia , Humanos , Trastornos por Fotosensibilidad/metabolismo , Trastornos por Fotosensibilidad/patología , Porfiria Eritropoyética/complicaciones , Porfiria Eritropoyética/metabolismo , Porfirias/metabolismo , Porfirias/patología
6.
Acta bioquím. clín. latinoam ; 50(4): 547-573, dic. 2016. ilus, tab
Artículo en Español | LILACS | ID: biblio-837630

RESUMEN

Las porfirias son enfermedades metabólicas consecuencia de fallas en la biosíntesis del hemo, caracterizadas por un patrón específico de acumulación y excreción de intermediarios, responsables de su patofisiología. En las porfirias agudas el exceso de ácido d-aminolevúlico (ALA) produce una sintomatología neuroabdominal asociada al daño oxidativo por formación de especies reactivas de oxígeno (ROS), originadas por autooxidaxión del ALA. En las cutáneas, la sintomatología es producto de la acumulación de porfirinas, que como el ALA, inducen la formación de ROS. Su desencadenamiento se precipita por factores endógenos (ayuno, estrés, hormonas) y/o exógenos (fármacos), en particular algunos anestésicos. Se presenta una revisión de los estudios bioquímicos y genéticos en pacientes con diferentes porfirias obtenidos en el Centro de Investigaciones de Porfirias y Porfirinas (CIPYP), durante los últimos 38 años, que permitieron ampliar el conocimiento sobre las bases moleculares sobre estas patologías. Se describen los logros resultantes del empleo de modelos experimentales de porfiria, inducida farmacológica o genéticamente, que contribuyeron a la clasificación de algunas drogas como prohibidas para pacientes con porfiria. Finalmente, las porfirinas generadoras de ROS, y por ende inductoras de muerte celular, tienen su aplicación para combatir infecciones por organismos hemo-deficientes como Trypanosoma cruzi y también para ser utilizadas como fotosensibilizadores en la terapia fotodinámica (TFD).


Porphyrias comprise a group of metabolic disorders of the heme biosynthesis pathway resulting in a specific accumulation and excretion of intermediates which are responsible for their pathophysiology. Acute porphyrias are characterized by acute neurovisceral symptoms due to the overproduction and accumulation of d-aminolevulinic acid (ALA) which leads to an oxidative damage resulting from the formation of reactive oxygen species (ROS). In cutaneous porphyrias, the symptomatology is a result of porphyrin accumulation which also induces ROS moulding. In both cases, their clinical signs are precipitated by endogenous factors (stress, hormones, low calories intake) and/or exogenous drugs, in particular some anaesthetics. A review of the biochemical and genetic results obtained from patients with different porphyrias, diagnosed at the CIPYP during the last 38 years is presented here, aimed at obtaining additional evidence about the molecular nature of these disorders. The achievements obtained from experimental porphyria models -pharmacologically or genetically induced- are also described, which contributed to the classification of some drugs as prohibited for their use in porphyric patients. Finally, as porphyrins produce ROS and therefore cellular death, they can be used to treat infections by heme-deficient organisms like Trypanosoma cruzi and also as photosensitizers in photodynamic therapy (TFD).


As Porfirias são doenças metabólicas decorrentes de falhas na biossíntese do Hemo, caracterizadas por um padrão específico de acumulação e excreção de intermediários responsáveis de sua patofisiologia. Nas Porfirias Agudas, o excesso de ácido δ-aminolevulínico (ALA) produz uma sintomatologia neuroabdominal associada ao dano oxidativo por formação de espécies reativas de oxigênio (ROS), decorrentes da auto-oxidação do ALA. Nas Cutâneas a sintomatologia é produto da acumulação de porfirinas, que como o ALA, induzem a formação de ROS. Seu desencadeamento precipita-se por fatores endógenos (jejum, estresse, hormônios) e/ou exógenos (fármacos), especialmente alguns anestésicos. Apresenta-se uma revisão dos estudos bioquímicos e genéticos em pacientes com diferentes Porfirias obtidos no Centro de Investigações de Porfirias e Porfirinas (CIPYP), durante os últimos 38 anos, que permitiram ampliar o conhecimento sobre as bases moleculares destas patologias. Descrevem-se as conquistas resultantes do uso de modelos experimentais de Porfiria, induzida farmacológica ou geneticamente, que contribuíram à classificação de algumas drogas como proibidas para pacientes com Porfiria. Afinal, as porfirinas geradoras de ROS e, por conseguinte, indutoras de morte celular têm sua aplicação para combater infecções por organismos hemo-deficientes como Trypanosoma cruzi e também ser utilizadas como fotossensibilizadores na terapia fotodinâmica (TFD).


Asunto(s)
Humanos , Anestésicos , Fotoquimioterapia , Porfirias , Porfirias/metabolismo , Porfirinas , Trypanosoma cruzi , Porfiria Eritropoyética , Protoporfiria Eritropoyética
7.
Salud colect ; 11(1): 99-114, ene.-mar. 2015. ilus, tab
Artículo en Español | LILACS | ID: lil-746687

RESUMEN

El Consejo Federal de Medicina de Brasil (CFM) -órgano normativo y fiscalizador del ejercicio ético de la medicina- prohibió, en 2008, la participación de médicos brasileños en investigaciones que utilizaran placebo para enfermedades con tratamiento eficaz y efectivo, en contraposición a la Declaración de Helsinki, que permite su uso en condiciones metodológicamente justificadas. Con el objetivo de verificar si la normativa ética del CFM modificó el uso de placebo en ensayos clínicos de fase III en Brasil, se analizaron varias características de sus registros en el ClinicalTrials.gov, en los períodos de 2003 a 2007 y de 2009 a 2013. Se concluye que: a) la normativa promulgada por el CFM en 2008 fue ineficaz y prevaleció la posición adoptada por la Declaración de Helsinki; b) el patrocinio de ensayos con placebo por parte de la industria farmacéutica multinacional fue significativo; c) predominaron las investigaciones de fármacos para enfermedades crónicas, y fueron poco significativas para las enfermedades postergadas, de importancia para Brasil.


In 2008, Brazil's Federal Council of Medicine [Conselho Federal de Medicina] (CFM) - regulatory and supervisory agency on the ethical practice of medicine - banned the participation of Brazilian doctors in studies using placebos for diseases with efficient and effective treatment. This position differs with the Helsinki Declaration, which allows the use of placebos in methodologically justified conditions. To ascertain whether the CMF's ethical regulation modified the use of placebos in phase III clinical trials in Brazil, characteristics of the records in ClinicalTrials.gov were researched in the periods from 2003 to 2007 and from 2009 to 2013. The conclusions reached were: a) the regulations issued by the CFM in 2008 were ineffective and the position adopted by the Helsinki Declaration prevails; b) there was significant sponsorship by the multinational pharmaceutical industry of trials with placebos; c) the research was predominantly on new drugs for chronic diseases, with little study done of the neglected diseases which are of great importance to Brazil.


Asunto(s)
Animales , Ratas , Apoptosis/genética , Regulación Enzimológica de la Expresión Génica/genética , Hemo/deficiencia , Degeneración Nerviosa/genética , Neuronas/metabolismo , Porfirias/complicaciones , Apoptosis/efectos de los fármacos , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Colágeno Tipo XI/efectos de los fármacos , Colágeno Tipo XI/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo/biosíntesis , Heptanoatos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Poli(ADP-Ribosa) Polimerasas , Porfirias/metabolismo , Porfirias/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas del Complejo SMN , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteínas de Transporte Vesicular/efectos de los fármacos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
8.
Chembiochem ; 15(14): 2024-35, 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25196849

RESUMEN

More than 20 years of research on heme as a temporary effector molecule of proteins have revealed its widespread impact on virtually all primary functions in the human organism. As our understanding of this influence is still growing, a comprehensive overview of compiled data will give fresh impetus for creativity and developing new strategies in heme-related research. From known data concerning heme-regulated proteins and their involvement in the development of diseases, we provide concise information of Fe(II/III) heme as a regulator and the availability of "regulatory heme". The latter is dependent on the balance between free and bound Fe(II/III) heme, here termed "hemeostasis". Imbalance of this system can lead to the development of diseases that were not always attributed to this small molecule. Diseases such as cancer or Alzheimer's disease highlight the reawakened interest in heme, whose function was previously believed to be completely understood.


Asunto(s)
Hemo/metabolismo , Hemoproteínas/metabolismo , Envejecimiento , Anemia/metabolismo , Animales , Compuestos Férricos/metabolismo , Compuestos Ferrosos/metabolismo , Hemólisis , Hemorragia/metabolismo , Humanos , MicroARNs/metabolismo , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Porfirias/metabolismo
9.
J Biol Chem ; 289(11): 7835-43, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24509859

RESUMEN

Mitochondrial iron is essential for the biosynthesis of heme and iron-sulfur ([Fe-S]) clusters in mammalian cells. In developing erythrocytes, iron is imported into the mitochondria by MFRN1 (mitoferrin-1, SLC25A37). Although loss of MFRN1 in zebrafish and mice leads to profound anemia, mutant animals showed no overt signs of porphyria, suggesting that mitochondrial iron deficiency does not result in an accumulation of protoporphyrins. Here, we developed a gene trap model to provide in vitro and in vivo evidence that iron regulatory protein-1 (IRP1) inhibits protoporphyrin accumulation. Mfrn1(+/gt);Irp1(-/-) erythroid cells exhibit a significant increase in protoporphyrin levels. IRP1 attenuates protoporphyrin biosynthesis by binding to the 5'-iron response element (IRE) of alas2 mRNA, inhibiting its translation. Ectopic expression of alas2 harboring a mutant IRE, preventing IRP1 binding, in Mfrn1(gt/gt) cells mimics Irp1 deficiency. Together, our data support a model whereby impaired mitochondrial [Fe-S] cluster biogenesis in Mfrn1(gt/gt) cells results in elevated IRP1 RNA-binding that attenuates ALAS2 mRNA translation and protoporphyrin accumulation.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Regulación de la Expresión Génica , Proteína 1 Reguladora de Hierro/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Porfirias/metabolismo , Animales , Blastocisto/citología , Diferenciación Celular , Línea Celular Tumoral , Femenino , Genotipo , Células HEK293 , Hemo/química , Humanos , Hierro/química , Proteínas Hierro-Azufre/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Protoporfirinas/metabolismo , Pez Cebra
10.
Histochem Cell Biol ; 141(5): 519-29, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24310659

RESUMEN

The Syrian hamster Harderian gland (HG) is an organ that undergoes physiological autophagy in response to oxidative stress induced by porphyrin production. Porphyrin production in the HG has marked sex differences and is closely linked to reproductive function. In the present study, we observed that the estrous cycle and associated estrogen variations may affect oxidative-stress-induced proteolytic processes. In particular, significant changes in autophagic activity were detected during the estrous cycle. Notably, increased activation of macroautophagy as well as chaperone-mediated autophagy in the estrus phase coincided with a minimal antioxidant capability and the highest protein damage levels. By contrast, autophagic machinery was found to be blocked in the diestrus phase, likely due to mammalian target of rapamycin activation, which could be corroborated by the subsequent pS6K activation. Analogous results were observed regarding proteasome activity, which also showed maximal activity in the estrus phase. Interestingly, all these mechanisms were associated with important morphological changes in the HG during the estrous cycle. We observed statistically significant increases in Type II cells, which may be related to extensive autophagy in the estrus phase. Physiologically, this would result in a significant release of porphyrins specifically when females are more receptive. These data support the role of porphyrins as pheromones, as other authors have previously suggested, thus making the HG a scent organ. In addition, these results suggest a porphyrin-based approach to the treatment of porphyria during pregnancy, a condition for which no treatment is currently known.


Asunto(s)
Autofagia , Ciclo Estral/metabolismo , Glándula de Harder/metabolismo , Porfirinas/metabolismo , Proteolisis , Animales , Estrógenos/metabolismo , Femenino , Humanos , Mesocricetus , Porfirias/metabolismo , Porfirias/patología , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/patología
11.
J Biol Chem ; 287(38): 32054-68, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22761424

RESUMEN

Liver is endowed with a mechanism to induce hepatic cytochromes P450 (CYP450s) in response to therapeutic drugs and environmental contaminants, leading to increased detoxification and elimination of the xenobiotics. Each CYP450 is composed of an apoprotein moiety and a heme prosthetic group, which is required for CYP450 activity. Thus, under conditions of CYP450 induction, there is a coordinate increase in heme biosynthesis to compensate for the increased expression of CYP450s. ABCB6, a mitochondrial ATP binding cassette transporter, which regulates coproporphyrinogen transport from the cytoplasm into the mitochondria to complete heme biosynthesis, represents a previously unrecognized rate-limiting step in heme biosynthesis. However, it is not known if exposure to drugs and environmental contaminants induces ABCB6 expression, to assure an adequate and apparently coordinated supply of heme for the generation of functional cytochrome holoprotein. In the present study, we demonstrate that polycyclic aromatic hydrocarbons (PAHs), the widely distributed environmental toxicants shown to induce porphyrin accumulation causing hepatic porphyria, up-regulate ABCB6 expression in both mice and humans. Using siRNA technology and Abcb6 knock-out mice, we demonstrate that PAH-mediated increase in hepatic porphyrins is compromised in the absence of ABCB6. Moreover, in vivo studies in aryl hydrocarbon receptor (AhR) knock-out mice demonstrate that PAH induction of ABCB6 is mediated by AhR. Promoter activation studies combined with electrophoretic mobility shift assay and chromatin immunoprecipitation assay demonstrate direct interactions between the AhR binding sites in the ABCB6 promoter and the AhR receptor, implicating drug activation mechanisms for ABCB6 similar to those found in inducible cytochrome P450s. These studies are the first to describe direct transcriptional activation of both mouse and human ABCB6 by xenobiotics.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Regulación de la Expresión Génica , Hidrocarburos Policíclicos Aromáticos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Activación Transcripcional , Adenosina Trifosfato/química , Animales , Sitios de Unión , Línea Celular , Supervivencia Celular , Células Cultivadas , Hemo/química , Hepatocitos/citología , Humanos , Hidrocarburos/química , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Modelos Biológicos , Porfirias/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Protoporfirinas/química , Xenobióticos/química
12.
Biometals ; 22(2): 345-51, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18956144

RESUMEN

Accumulating evidence, including experiments using cytochrome P450 1a2 (Cyp1a2) gene knock-out mice (Cyp1a2(-/-)), indicates that the development of chemically induced porphyria requires the expression of CYP1A2. It has also been demonstrated that iron enhances and expedites the development of experimental uroporphyria, but that iron alone without CYP1A2 expression, as in Cyp1a2(-/-) mice, does not cause uroporphyria. The role of iron in the development of porphyria has not been elucidated. We examined the in vivo effect of iron deficiency on hepatic URO accumulation in experimental porphyria. Mice were fed diets containing low (iron-deficient diet (IDD), 8.5 mg iron/kg) or normal (normal diet (ND), 213.7 mg iron/kg) levels of iron. They were treated with 3-methylcholanthrene (MC), an archetypal inducer of CYP1A, and 5-aminolevulinate (ALA), precursors of porphyrin and heme. We found that uroporphyrin (URO) levels and uroporphyrinogen oxidation (UROX) activity were markedly increased in ND mice treated with MC and ALA, while the levels were not raised in IDD mice with the same treatments. CYP1A2 levels and methoxyresorufin O-demethylase (MROD) activities, the CYP1A2-mediated reaction, were markedly induced in the livers of both ND and IDD mice treated with MC and ALA. UROX activity, supposedly a CYP1A2-dependent activity, was not enhanced in iron-deficient mice in spite of the fact of induction of CYP1A2. We showed that a sufficient level of iron is essential for the development of porphyria and UROX activity.


Asunto(s)
Ácido Aminolevulínico/farmacología , Hierro/metabolismo , Metilcolantreno/farmacología , Porfirias/inducido químicamente , Uroporfirinas/metabolismo , Animales , Citocromo P-450 CYP1A2/genética , Hierro/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/química , Porfirias/metabolismo , Factores de Tiempo , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo , Uroporfirinas/química
13.
Arch Biochem Biophys ; 474(2): 238-51, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18314007

RESUMEN

Heme is an integral part of proteins involved in multiple electron transport chains for energy recovery found in almost all forms of life. Moreover, heme is a cofactor of enzymes including catalases, peroxidases, cytochromes of the P(450) class and part of sensor molecules. Here the step-by-step biosynthesis of heme including involved enzymes, their mechanisms and detrimental health consequences caused by their failure are described. Unusual and challenging biochemistry including tRNA-dependent reactions, radical SAM enzymes and substrate derived cofactors are reported.


Asunto(s)
Hemo/biosíntesis , Tetrapirroles/biosíntesis , Ácido Aminolevulínico/metabolismo , Archaea/metabolismo , Bacterias/metabolismo , Hemo/química , Herbicidas , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fotoquimioterapia , Plantas/metabolismo , Porfirias/genética , Porfirias/metabolismo , ARN de Transferencia/metabolismo
14.
Bol. Hosp. San Juan de Dios ; 54(2): 68-94, mar.-abr. 2007. tab, graf
Artículo en Español | LILACS | ID: lil-467665

RESUMEN

Las porfirias son un conjunto de enfermedades –genéticas (monogenicas) o adquiridas debidas a disminución de la actividad de al menos una de las enzimas que participan en la vía de síntesis del grupo hem. Según sea la o las enzimas defectuosas, se genera una enfermedad con un patrón típico de alteraciones en la circulación, excreción y acumulación tisular de porfirinas o sus precursores, produciendo manifestaciones clínicas características para cada variedad. Se han diagnosticado en animales y en hombres de todas las razas y áreas geográficas. Se reconocen diversas variedades: la aguda intermitente particularmente frecuente en Suecia, la variegata común en la población blanca de Sudáfrica, la coproporfiria, la porfiria de Dõss de la que se han descrito unos pocos casos en el mundo, la eritropoyética congénita de muy baja prevalencia, la cutánea tarda que es lo mas frecuente de todas y que puede ser hereditaria o adquirida, en cuyo caso se relaciona con la hepatitis por virus C o B y VIH, por último la protoporfiria que suele expresarse muy tempranamente y ser causa de enfermedad hepática aguda grave que requiere trasplante hepático. En Chile hemos identificado la mayor parte de las variedades, existiendo tanto casos de origen europeo como de pueblos originarios. Los portadores de mutaciones genéticas pueden presentar una enfermedad clínicamente evidente o una asintomática o latente, en la que incluso puede no haber alteraciones metabólicas detectables. Los periodos sintomáticos y de latencia pueden alternarse, pero la mayoría de los enfermos tiene la enfermedad latente a lo largo de toda su vida. Las manifestaciones clínicas que se pueden observar en las porfirias son: crisis agudas, alteraciones cutáneas y diversas complicaciones tales como hepatopatias. Las primeras consisten en episodios, dramáticos por su gravedad, de alteraciones neurológicas, psíquicas, cardiovasculares, digestivas, urinarias y de compromiso del estado general que incluye hiponatremia.


Asunto(s)
Humanos , Porfirias/clasificación , Porfirias/diagnóstico , Porfirias/etiología , Porfirias/metabolismo , Porfirias/tratamiento farmacológico , Signos y Síntomas
15.
Brain Res Mol Brain Res ; 137(1-2): 23-30, 2005 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-15950757

RESUMEN

Defective heme synthesis may cause acute porphyrias, which are associated with a wide array of neurological disturbances involving both the central and peripheral nervous systems. Thus, the understanding of the roles of heme in neuronal cell function may provide insights into the molecular events underlying the pathogenesis of neuropathies associated with defective heme synthesis. In this report, we use rat pheochromocytoma (PC12) clonal cells as a model system for studying the role of heme in neuronal cell survival. We examined the effects of inhibition of heme synthesis on signaling pathways and gene expression in nerve growth factor (NGF)-induced PC12 cells. We found that succinyl acetone-induced heme deficiency selectively caused apoptosis in NGF-induced PC12 cells. Further, we found that in succinyl acetone-treated, NGF-induced cells, the pro-survival Ras-ERK1/2 signaling pathway was inactivated and the pro-apoptotic JNK signaling pathway was activated. In these cells, the activation of caspase and the cleavage of nuclear poly (ADP-ribose) polymerase (PARP) were also evident. Importantly, microarray gene expression analysis showed that more than 20 key neuronal genes that were induced by NGF were suppressed by succinyl acetone. These genes include those encoding survival motor neuron protein, synaptic vesicle protein SVOP, and neural cell adhesion molecule NCAM. These results indicate that heme is important for neuronal cell signaling and the proper functioning of neuronal cells.


Asunto(s)
Apoptosis/genética , Regulación Enzimológica de la Expresión Génica/genética , Hemo/deficiencia , Degeneración Nerviosa/genética , Neuronas/metabolismo , Porfirias/complicaciones , Animales , Apoptosis/efectos de los fármacos , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Colágeno Tipo XI/efectos de los fármacos , Colágeno Tipo XI/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo/biosíntesis , Heptanoatos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Células PC12 , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas , Porfirias/metabolismo , Porfirias/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ratas , Proteínas del Complejo SMN , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteínas de Transporte Vesicular/efectos de los fármacos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
16.
J Cutan Med Surg ; 6(4): 320-6, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12118363

RESUMEN

BACKGROUND: Pseudoporphyria is a diagnosis that is used when porphyria-like clinical lesions arise in the setting of normal porphyrin levels. This condition was first described in the 1960s and was initially related to the use of certain antibiotic drugs. In 1985, pseudoporphyria was first attributed to the use of nonsteroidal antiinflammatory drugs (NSAIDs). Subsequently, a host of NSAIDs and other drugs have been found to elicit the same clinical entity. The exact mechanism by which certain drugs create clinical lesions resembling porphyria cutanea tarda or erythropoietic protoporphyria is still unknown. A phototoxic mechanism is hypothesized. OBJECTIVE: We describe six patients diagnosed with pseudoporphyria and detail the diagnostic tests leading to the eventual diagnosis. RESULTS: The patients ranged in age from 27 to 59 years and had a female:male predominance of 2:1. The offending NSAID was DayPro (oxaprozin) for three of the patients, Relafen (nabumetone) for two of the patients, and Aleve (naproxen) for one patient. For each patient, histology and immunofluorescence was either consistent with the diagnosis of porphyria cutanea tarda or nonspecific, while serum, stool, and urine porphyrins did not support that diagnosis. Withdrawal of the offending agent provided relief from the clinical symptoms for each patient. None of our patients were rechallenged with the putative offending drug. However, prolonged avoidance has provided a sustained remission from symptoms in all six patients. CONCLUSIONS: Pseudoporphyria is a relatively rarely reported condition. Clinical suspicion with appropriate laboratory and histopathologic findings help to make this diagnosis, and exclude true porphyrias. Rechallenge with the offending drug to produce symptom relapse has been proposed to be helpful in confirming this diagnosis of exclusion. Since all 6 patients with drug-induced pseudoporphyria experienced resolution of their symptoms after discontinuing the offending agent, we propose that this clinical correlation alone is sufficient to confirm this diagnosis. Our observation of six new cases of NSAID-induced pseudoporphyria over a two-year interval suggests that this is not a rare entity.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Porfirias/inducido químicamente , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Porfirias/clasificación , Porfirias/metabolismo , Porfirias/patología , Porfirinas/metabolismo , Piel/patología
17.
Biol Chem ; 382(6): 913-8, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11501755

RESUMEN

5-Aminolevulinic acid (ALA) is a heme precursor that accumulates in lead poisoning and inborn porphyrias. It has been shown to produce reactive oxygen species upon metal-catalyzed aerobic oxidation and to cause oxidative damage to proteins, liposomes, DNA, and subcellular structures. Studies have also shown that ALA may condense to yield the cyclic product 3,6-dihydropyrazine-2,5-dipropanoic acid (DHPY). Here we propose that DHPY could be involved in DNA damage in the presence of high concentrations of ALA. Exposure of plasmid pUC19 DNA to low concentrations of DHPY (2-10 microM) in the presence of 0.1 mM Cu2+ ions causes DNA strand breaks, as demonstrated by agarose gel electrophoresis. It was also shown that in the presence of Cu2+ ions DHPY is able to increase the oxidation of monomeric 2'-deoxyguanosine to form 8-oxo-7,8-dihydro-2'-deoxyguanosine as inferred from high performance liquid chromatography measurements using electrochemical detection. Addition of a metal chelator (bathocuproine, 0.5 mM), the DNA compacting polyamines spermidine (1 mM) and spermine (1 mM) or antioxidant enzymes such as superoxide dismutase (10 microg/ml) and catalase (20 pg/ml) protect the DNA against these damages. The data presented here are discussed with respect to the increased frequency of liver cancer in patients with acute intermittent porphyria.


Asunto(s)
Ácido Aminolevulínico/metabolismo , Daño del ADN/efectos de los fármacos , Propionatos/farmacología , Pirazinas/farmacología , Ácido Aminolevulínico/farmacología , Cobre/farmacología , Ciclización , Desoxiguanosina/metabolismo , Dimerización , Oxidación-Reducción , Oxidorreductasas/farmacología , Plásmidos/metabolismo , Plásmidos/farmacología , Porfirias/metabolismo , Propionatos/síntesis química , Pirazinas/síntesis química
18.
Mutagenesis ; 15(5): 379-83, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10970442

RESUMEN

The synergistic interaction of iron overload, AHR: genotype and exposure to a mixture of polychlorinated biphenyls (PCBs) (Aroclor 1254) in mice leads to hepatic porphyria, oxidative DNA damage and cancer. In humans, hepatocellular cancer is associated with iron overload and hepatic porphyria. Neither the mechanism of hepatic carcinogenesis induced by PCBs in rodents nor hepatocellular cancer induced by iron and porphyria in humans are understood. To test the hypothesis that chronic interaction of iron and PCBs may induce mutagenesis in liver DNA, lambda /lacI transgenic C57BL/6 mice were given iron dextran (600 mg iron/kg) and then administered Aroclor 1254 in the diet (0.01%) for 7 weeks. Hepatic iron, CYP1A activity and CYP1A1/1A2 protein were elevated >20-fold as a result of iron or Aroclor treatments, respectively, but porphyria with associated histological changes only developed in the combined iron/Aroclor treatment group. lambda/lacI shuttle vectors were isolated from liver genomic DNA and the mutational frequency (MF) in the lacI gene determined. Both iron and Aroclor treatments alone caused significant small increases in MF (1.5- and 1.4-fold, respectively), however, the MF following the combined iron and Aroclor treatment (1. 6-fold) was not greater than the additive effects. In contrast, the MF was significantly elevated (4.7-fold) in liver DNA of mice 2 weeks following five daily doses of N-nitrosodimethylamine (4 mg/kg). These studies demonstrate that neither PCBs nor iron overload caused marked point mutations even in a combination regime that leads to oxidative damage and cancer. There was also no strong evidence either that porphyrins or chronic CYP1A1 expression induced by the PCBs after this period caused marked point mutagens or simple deletions. Hence, to understand the PCBs-iron synergism more complex scenarios than point mutations or simple deletions must be invoked.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Escherichia coli , Sobrecarga de Hierro/genética , Neoplasias Hepáticas Experimentales/genética , Hígado/patología , Mutación , Bifenilos Policlorados/toxicidad , Porfirias/genética , Proteínas Represoras/genética , Animales , Bacteriófago lambda/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Daño del ADN , Dimetilnitrosamina/toxicidad , Vectores Genéticos/genética , Sobrecarga de Hierro/metabolismo , Represoras Lac , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Mutagénesis , Estrés Oxidativo/efectos de los fármacos , Porfirias/metabolismo , Porfirinas/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-10982942

RESUMEN

Acute porphyrias are classified into 3 distinct groups of rare genetic disorders of metabolic enzyme biosynthesis. Acute porphyrias can significantly impact multiple organ systems, which often provides a challenge to the dentist presented with such a patient. A case of hereditary coproporphyria is reported in a patient with many of the classical signs and symptoms. The patient also had complex dental needs that required special medical and pharmacotherapeutic modifications. The acute porphyrias are reviewed by the authors with presentation of this challenging case. Recommendations for other dental health care professionals encountering these patients are then presented.


Asunto(s)
Atención Dental para Enfermos Crónicos , Enfermedades de la Boca/etiología , Porfirias Hepáticas/complicaciones , Porfirias , Enfermedad Aguda , Adulto , Caries Dental/etiología , Femenino , Humanos , Periodontitis/etiología , Porfirias/clasificación , Porfirias/complicaciones , Porfirias/metabolismo , Porfirias/terapia , Trastornos de la Articulación Temporomandibular/etiología
20.
Arch Biochem Biophys ; 375(2): 240-50, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10700380

RESUMEN

To confirm and extend our previous microspectrophotometric observations of 30-week-old male Long-Evans Cinnamon (LEC) rats, an animal model of human Wilson's disease, we analyzed the porphyrin patterns of the organs, urine, and plasma of LEC rats. Abnormal accumulation of porphyrins, especially highly carboxylated porphyrins (uro- and heptaporphyrin), in the kidneys and liver was seen in male and female LEC rats aged 30 weeks and also in 10-week-old rats, before the onset of spontaneous hepatic dysfunction. Accumulation of copper and iron in the kidneys was not observed in the 10-week-old rats. Massive accumulation of porphyrins was observed only in the kidneys of the 30-week-old male LEC rat, indicating that this symptom is related to sex and age. Renal accumulation of porphyrins was reflected in the rate of urinary porphyrin excretion. Hepatic accumulation of porphyrins appeared to be independent of sex and age. These results indicate that neither renal nor hepatic porphyrin accumulation is the result of renal deposition of metals or of spontaneous hepatic dysfunction and that porphyrinuria in the LEC rat is closely related to the renal accumulation of porphyrins. In contrast to these organs, a reduction in the porphyrin levels was observed in the brain of the LEC rat. This was independent of sex and age. The present work stresses the existence of an abnormal heme metabolism in the LEC rat, and thus, the necessity to study the heme metabolism in human Wilson's disease is strongly suggested.


Asunto(s)
Modelos Animales de Enfermedad , Degeneración Hepatolenticular/metabolismo , Porfirias/metabolismo , Porfirinas/análisis , Envejecimiento/fisiología , Animales , Médula Ósea/química , Química Encefálica , Cromatografía Líquida de Alta Presión , Femenino , Hemo/metabolismo , Degeneración Hepatolenticular/sangre , Degeneración Hepatolenticular/fisiopatología , Degeneración Hepatolenticular/orina , Humanos , Riñón/química , Hígado/química , Masculino , Microscopía Fluorescente , Porfirias/sangre , Porfirias/fisiopatología , Porfirias/orina , Porfirinas/sangre , Porfirinas/química , Porfirinas/orina , Ratas , Ratas Endogámicas LEC , Caracteres Sexuales , Bazo/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA