Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Drug Metab Dispos ; 50(6): 781-797, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34154993

RESUMEN

Conjugation of oligonucleotide therapeutics, including small interfering RNAs (siRNAs) or antisense oligonucleotides, to N-acetylgalactosamine (GalNAc) ligands has become the primary strategy for hepatocyte-targeted delivery, and with the recent approvals of GIVLAARI (givosiran) for the treatment of acute hepatic porphyria, OXLUMO (lumasiran) for the treatment of primary hyperoxaluria, and Leqvio (inclisiran) for the treatment of hypercholesterolemia, the technology has been well validated clinically. Although much knowledge has been gained over decades of development, there is a paucity of published literature on the drug metabolism and pharmacokinetic properties of GalNAc-siRNA. With this in mind, the goals of this minireview are to provide an aggregate analysis of these nonclinical absorption, distribution, metabolism, and excretion (ADME) data to build confidence on the translation of these properties to human. Upon subcutaneous administration, GalNAc-conjugated siRNAs are quickly distributed to the liver, resulting in plasma pharmacokinetic (PK) properties that reflect rapid elimination through asialoglycoprotein receptor-mediated uptake from circulation into hepatocytes. These studies confirm that liver PK, including half-life and, most importantly, siRNA levels in RNA-induced silencing complex in hepatocytes, are better predictors of pharmacodynamics (PD) than plasma PK. Several in vitro and in vivo nonclinical studies were conducted to characterize the ADME properties of GalNAc-conjugated siRNAs. These studies demonstrate that the PK/PD and ADME properties of GalNAc-conjugated siRNAs are highly conserved across species, are largely predictable, and can be accurately scaled to human, allowing us to identify efficacious and safe clinical dosing regimens in the absence of human liver PK profiles. SIGNIFICANCE STATEMENT: Several nonclinical ADME studies have been conducted in order to provide a comprehensive overview of the disposition and elimination of GalNAc-conjugated siRNAs and the pharmacokinetic/pharmacodynamic translation between species. These studies demonstrate that the ADME properties of GalNAc-conjugated siRNAs are well correlated and predictable across species, building confidence in the ability to extrapolate to human.


Asunto(s)
Acetilgalactosamina , Porfirias Hepáticas , Acetilgalactosamina/farmacocinética , Receptor de Asialoglicoproteína/metabolismo , Hepatocitos/metabolismo , Humanos , Porfirias Hepáticas/metabolismo , ARN Interferente Pequeño/genética
2.
Hepatology ; 66(4): 1314-1322, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28605040

RESUMEN

The acute hepatic porphyrias are a group of four inherited disorders, each resulting from a deficiency in the activity of a specific enzyme in the heme biosynthetic pathway. These disorders present clinically with acute neurovisceral symptoms which may be sporadic or recurrent and, when severe, can be life-threatening. The diagnosis is often missed or delayed as the clinical features resemble other more common medical conditions. There are four major subgroups: symptomatic patients with sporadic attacks (<4 attacks/year) or recurrent acute attacks (≥4 attacks/year), asymptomatic high porphyrin precursor excretors, and asymptomatic latent patients without symptoms or porphyrin precursor elevations. Given their clinical heterogeneity and potential for significant morbidity with suboptimal management, comprehensive clinical guidelines for initial evaluation, follow-up, and long-term management are needed, particularly because no guidelines exist for monitoring disease progression or response to treatment. The Porphyrias Consortium of the National Institutes of Health's Rare Diseases Clinical Research Network, which consists of expert centers in the clinical management of these disorders, has formulated these recommendations. These recommendations are based on the literature, ongoing natural history studies, and extensive clinical experience. Initial assessments should include diagnostic confirmation by biochemical testing, subsequent genetic testing to determine the specific acute hepatic porphyria, and a complete medical history and physical examination. Newly diagnosed patients should be counseled about avoiding known precipitating factors. The frequency of follow-up depends on the clinical subgroup, with close monitoring of patients with recurrent attacks who may require treatment modifications as well as those with clinical complications. Comprehensive care should include subspecialist referrals when needed. Annual assessments include biochemical testing and monitoring for long-term complications. These guidelines provide a framework for monitoring patients with acute hepatic porphyrias to ensure optimal outcomes. (Hepatology 2017;66:1314-1322).


Asunto(s)
Porfirias Hepáticas/terapia , Manejo de la Enfermedad , Humanos , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/metabolismo
3.
Curr Opin Hematol ; 24(3): 198-207, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28118224

RESUMEN

PURPOSE OF REVIEW: Many studies over the past decade have together identified new genes including modifier genes and new regulation and pathophysiological mechanisms in inherited inborn diseases of the heme biosynthetic pathway. A new porphyria has been characterized: X-linked protoporphyria and the perspective to have innovative treatment at very short-term became a reality. We will summarize how recent data on both ALAS1 and ALAS2 have informed our understanding of disease pathogenesis with an emphasis on how this information may contribute to new therapeutic strategies. RECENT FINDINGS: The development of clinical and biological porphyria networks improved the long-term follow up of cohorts. The ageing of patients have allowed for the identification of novel recurrently mutated genes, and highlighted long-term complications in acute hepatic porphyrias. The treatment of hepatic porphyrias by an RNAi-targeting hepatic ALAS1 is actually tested and may lead to improve the management of acute attacks.In erythropoietic porphyrias, the key role of ALAS2 as a gate keeper of the heme and subsequently hemoglobin synthesis has been demonstrated. Its implication as a modifier gene in over erythroid disorders has also been documented. SUMMARY: The knowledge of both the genetic abnormalities and the regulation of heme biosynthesis has increased over the last 5 years and open new avenues in the management of erythropoietic and acute hepatic porphyrias.


Asunto(s)
Porfobilinógeno Sintasa/deficiencia , Porfiria Eritropoyética/etiología , Porfiria Eritropoyética/metabolismo , Porfirias Hepáticas/etiología , Porfirias Hepáticas/metabolismo , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , 5-Aminolevulinato Sintetasa/uso terapéutico , Factores de Edad , Animales , Biomarcadores , Dolor Crónico/etiología , Activación Enzimática , Eritrocitos/metabolismo , Regulación de la Expresión Génica , Genes Ligados a X , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Hemo/biosíntesis , Humanos , Mutación , Fenotipo , Porfobilinógeno Sintasa/metabolismo , Porfiria Eritropoyética/diagnóstico , Porfiria Eritropoyética/terapia , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/terapia , ARN Interferente Pequeño/genética
4.
Mol Genet Metab ; 119(3): 278-283, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27595545

RESUMEN

BACKGROUND: The acute hepatic porphyrias (AHPs) are rare inborn errors of heme biosynthesis, characterized clinically by life-threatening acute neurovisceral attacks. Patients with recurrent attacks have a decreased quality of life (QoL); however, no interactive assessment of these patients' views has been reported. We conducted guided discussions regarding specific topics, to explore patients' disease experience and its impact on their lives. METHODS: Sixteen AHP patients experiencing acute attacks were recruited to moderator-led online focus groups. Five groups (3-4 patients each) were conducted and thematic analyses to identify, examine, and categorize patterns in the data was performed. RESULTS: All patients identified prodromal symptoms that began days prior to acute severe pain; the most common included confusion ("brain fog"), irritability, and fatigue. Patients avoided hospitalization due to prior poor experiences with physician knowledge of AHPs or their treatment. All patients used complementary and alternative medicine treatments to avoid hospitalization or manage chronic pain and 81% reported varying degrees of effectiveness. All patients indicated their disease impacted personal relationships due to feelings of isolation and difficulty adjusting to the disease's limitations. CONCLUSION: Patients with recurrent attacks recognize prodromal warning symptoms, attempt to avoid hospitalization, turn to alternative treatments, and have markedly impaired QoL. Counseling and individualized support is crucial for AHP patients with recurrent attacks.


Asunto(s)
Pacientes/psicología , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/fisiopatología , Porfirias Hepáticas/psicología , Adulto , Anciano , Femenino , Hemo/biosíntesis , Hemo/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Relaciones Médico-Paciente , Porfobilinógeno Sintasa/metabolismo , Porfirias Hepáticas/epidemiología , Porfirias Hepáticas/metabolismo , Calidad de Vida
5.
J Cell Sci ; 126(Pt 14): 3105-12, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23641075

RESUMEN

Oxidative liver injury during steatohepatitis results in aggregation and transglutaminase-2 (TG2)-mediated crosslinking of the keratin cytoplasmic intermediate filament proteins (IFs) to form Mallory-Denk body (MDB) inclusions. The effect of liver injury on lamin nuclear IFs is unknown, though lamin mutations in several human diseases result in lamin disorganization and nuclear shape changes. We tested the hypothesis that lamins undergo aggregation during oxidative liver injury using two MDB mouse models: (i) mice fed the porphyrinogenic drug 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) and (ii) mice that harbor a mutation in ferrochelatase (fch), which converts protoporphyrin IX to heme. Dramatic aggregation of lamin A/C and B1 was noted in the livers of both models in association with changes in lamin organization and nuclear shape, as determined by immunostaining and electron microscopy. The lamin aggregates sequester other nuclear proteins including transcription factors and ribosomal and nuclear pore components into high molecular weight complexes, as determined by mass-spectrometry and confirmed biochemically. Lamin aggregate formation is rapid and precedes keratin aggregation in fch livers, and is seen in liver explants of patients with alcoholic cirrhosis. Exposure of cultured cells to DDC, protoporphyrin IX or N-methyl-protoporphyrin, or incubation of purified lamins with protoporphyrin IX, also results in lamin aggregation. In contrast, lamin aggregation is ameliorated by TG2 inhibition. Therefore, lamin aggregation is an early sensor of porphyria-associated liver injury and might serve to buffer oxidative stress. The nuclear shape and lamin defects associated with porphyria phenocopy the changes seen in laminopathies and could result in transcriptional alterations due to sequestration of nuclear proteins.


Asunto(s)
Hígado Graso/metabolismo , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Porfirias Hepáticas/metabolismo , Animales , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/genética , Ferroquelatasa/genética , Proteínas de Unión al GTP/antagonistas & inhibidores , Células Hep G2 , Humanos , Cuerpos de Mallory/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación/genética , Estrés Oxidativo , Porfirias Hepáticas/complicaciones , Porfirias Hepáticas/genética , Proteína Glutamina Gamma Glutamiltransferasa 2 , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Protoporfirinas/farmacología , Piridinas/toxicidad , Transglutaminasas/antagonistas & inhibidores
6.
Eksp Klin Gastroenterol ; (7): 69-77, 2015.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-26817125

RESUMEN

Porphyrias are a group of rare disorders caused by enzyme defects in haem biosynthesis pathway. Acute intermittent porphyria is the most common hepatic porphyria. The disorder presents with severe neuropathic abdominal pain that can be accompanied by a wide range of gastrointestinal, psychiatric and neurological symptoms, making the diagnosis clarification very challenging. We report a case of a 27-year-old female patient who presented with acute abdominal pain, vomiting and marked hyponatremia, developed seizures and disorientation, and eventually required intensive care unit treatment to maintain breathing. Her symptoms were initially misinterpreted as a functional gastrointestinal disorder, thus delaying the needed specific treatment. She was diagnosed a week after the initial hospital admission, and her condition improved after receiving treatment with intravenous glucose and haemin. For patients with acute neurovisceral attacks, early clinical recognition is essential. Severe hyponatremia, urine that develops orange colour on exposure to light and gastrointestinal symptom combination with neurologic symptoms are three valuable clues that may lead to the right diagnosis faster. Pathophysiology of hyponatremia in case of acute intermittent porphyria in only partly understood and can be associated with syndrome of inappropriate antidiuretic hormone secretion, gastrointestinal or renal sodium loss.


Asunto(s)
Dolor Abdominal , Neuralgia , Porfirias Hepáticas , Dolor Abdominal/diagnóstico , Dolor Abdominal/metabolismo , Dolor Abdominal/patología , Dolor Abdominal/fisiopatología , Adulto , Femenino , Humanos , Neuralgia/diagnóstico , Neuralgia/metabolismo , Neuralgia/patología , Neuralgia/fisiopatología , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/metabolismo , Porfirias Hepáticas/patología , Porfirias Hepáticas/fisiopatología
7.
Dtsch Med Wochenschr ; 146(15): 955-958, 2021 08.
Artículo en Alemán | MEDLINE | ID: mdl-34344029

RESUMEN

Givosiran is a small synthetic double-stranded siRNA (small interfering RNA) conjugated with N-acetyl-galactosamine (GalNAc) for specific hepatocyte targeting via the asialoglycoprotein receptor. A prospective randomized multicenter study (Envision) demonstrated the clinical efficacy of monthly subcutaneous injection of Givosiran for the prevention of attacks of acute hepatic porphyria (AHP). This leads to highly selective transcriptional inhibition of the key hepatic enzyme, aminolaevulinate synthase 1, that is overexpressed in AHP. The success of the Envision study has led to the approval of Givosiran in the US and Europe for the treatment of severe AHP. This innovative guided siRNA therapy has opened up the possibility to selectively inhibit the expression of any hepatocyte gene whose overexpression that causes pathology, which can be considered a milestone development in hepatology. However, currently this treatment with givosiran is very costly. Moreover, since some patients experience worsening of kidney function and elevated aminotransferases, monthly monitoring of these parameters is necessary in the first half year of treatment.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/tratamiento farmacológico , Pirrolidinas , ARN Interferente Pequeño , Tratamiento con ARN de Interferencia , 5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/uso terapéutico , Humanos , Porfobilinógeno Sintasa/metabolismo , Porfirias Hepáticas/metabolismo , Porfirias Hepáticas/fisiopatología , Porfirias Hepáticas/prevención & control , Pirrolidinas/administración & dosificación , Pirrolidinas/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/uso terapéutico , ARN Interferente Pequeño/ultraestructura , Ensayos Clínicos Controlados Aleatorios como Asunto
8.
Methods Mol Biol ; 2282: 57-75, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33928570

RESUMEN

Small interfering RNA (siRNA)-based therapeutics holds the promise to treat a wide range of human diseases that are currently incurable using conventional therapies. Most siRNA therapeutic efforts to date have focused on the treatment of liver diseases due to major breakthroughs in the development of efficient strategies for delivering siRNA drugs to the liver. Indeed, the development of lipid nanoparticle-formulated and GalNAc-conjugated siRNA therapeutics has resulted in recent FDA approvals of the first siRNA-based drugs, patisiran for the treatment of hereditary transthyretin amyloidosis and givosiran for the treatment of acute hepatic porphyria, respectively. Here, we describe the current strategies for delivering siRNA drugs to the liver and summarize recent advances in clinical development of siRNA therapeutics for the treatment of liver diseases.


Asunto(s)
Hepatopatías/terapia , Interferencia de ARN , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia , Acetilgalactosamina/análogos & derivados , Acetilgalactosamina/uso terapéutico , Neuropatías Amiloides Familiares/genética , Neuropatías Amiloides Familiares/metabolismo , Neuropatías Amiloides Familiares/terapia , Animales , Técnicas de Transferencia de Gen , Humanos , Hepatopatías/genética , Hepatopatías/metabolismo , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/metabolismo , Porfirias Hepáticas/terapia , Pirrolidinas/uso terapéutico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
9.
Clin Pharmacol Ther ; 110(5): 1250-1260, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34510420

RESUMEN

Givosiran (trade name GIVLAARI) is a small interfering ribonucleic acid that targets hepatic delta-aminolevulinic acid synthase 1 (ALAS1) messenger RNA for degradation through RNA interference (RNAi) that has been approved for the treatment of acute hepatic porphyria (AHP). RNAi therapeutics, such as givosiran, have a low liability for drug-drug interactions (DDIs) because they are not metabolized by cytochrome 450 (CYP) enzymes, and do not directly inhibit or induce CYP enzymes in the liver. The pharmacodynamic effect of givosiran (lowering of hepatic ALAS1, the first and rate limiting enzyme in the heme biosynthesis pathway) presents a unique scenario where givosiran could potentially impact heme-dependent activities in the liver, such as CYP enzyme activity. This study assessed the impact of givosiran on the pharmacokinetics of substrates of 5 major CYP450 enzymes in subjects with acute intermittent porphyria (AIP), the most common type of AHP, by using the validated "Inje cocktail," comprised of caffeine (CYP1A2), losartan (CYP2C9), omeprazole (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A4). We show that givosiran treatment had a differential inhibitory effect on CYP450 enzymes in the liver, resulting in a moderate reduction in activity of CYP1A2 and CYP2D6, a minor effect on CYP3A4 and CYP2C19, and a similar weak effect on CYP2C9. To date, this is the first study evaluating the DDI for an oligonucleotide therapeutic and highlights an atypical drug interaction due to the pharmacological effect of givosiran. The results of this study suggest that givosiran does not have a large effect on heme-dependent CYP enzyme activity in the liver.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas/fisiología , Activación Enzimática/fisiología , Hígado/metabolismo , Pirrolidinas/metabolismo , ARN Interferente Pequeño/metabolismo , 5-Aminolevulinato Sintetasa/metabolismo , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/metabolismo , Adulto , Cafeína/administración & dosificación , Cafeína/metabolismo , Estudios Cruzados , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Hígado/efectos de los fármacos , Masculino , Tasa de Depuración Metabólica/efectos de los fármacos , Tasa de Depuración Metabólica/fisiología , Midazolam/administración & dosificación , Midazolam/metabolismo , Persona de Mediana Edad , Omeprazol/administración & dosificación , Omeprazol/metabolismo , Porfirias Hepáticas/tratamiento farmacológico , Porfirias Hepáticas/metabolismo , Pirrolidinas/administración & dosificación
10.
Drugs ; 80(3): 335-339, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32034693

RESUMEN

Givosiran (Givlaari™) is an aminolevulinate synthase 1 (ALAS1)-directed small interfering RNA (siRNA) covalently linked to a ligand to enable specific delivery of the siRNA to hepatocytes. This results in downregulation of ALAS1 mRNA and prevents accumulation of neurotoxic δ-aminolevulinic acid and porphobilinogen levels that are associated with acute porphyria attacks. Givosiran is being developed by Alnylam Pharmaceuticals for the treatment of acute hepatic porphyria (AHP). In November 2019, givosiran was approved in the USA for the treatment of adults with AHP based on the positive results from the multinational, phase III ENVISION trial. In the EU, givosiran received a positive opinion in January 2020 for the treatment of AHP in adults and adolescents aged 12 years and older. This article summarizes the milestones in the development of givosiran leading to this first approval for the treatment of adults with AHP.


Asunto(s)
5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Acetilgalactosamina/análogos & derivados , Aprobación de Drogas , Inhibidores Enzimáticos/farmacología , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/tratamiento farmacológico , Pirrolidinas/farmacología , 5-Aminolevulinato Sintetasa/metabolismo , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacología , Inhibidores Enzimáticos/administración & dosificación , Humanos , Porfobilinógeno Sintasa/metabolismo , Porfirias Hepáticas/metabolismo , Pirrolidinas/administración & dosificación , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo
11.
Med Hypotheses ; 131: 109314, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31443750

RESUMEN

Metabolic targeting of liver 5-aminolevulinate synthase (5-ALAS) by inhibition of heme utilisation by tryptophan (Trp) 2,3-dioxygenase (TDO) or the use of tryptophan is proposed as a therapy of acute hepatic porphyrias. 5-ALAS, the rate-limiting enzyme of heme biosynthesis, is under negative feedback control by a small regulatory heme pool in the hepatic cytosol. Acute porphyric attacks, precipitated by fasting, certain hormones and some drugs, involve induction of 5-ALAS secondarily to depletion of the above pool, and the resultant elevation of 5-ALA levels initiates the abdominal and neurological symptoms of attacks. By utilising the regulatory heme, cytosolic TDO undermines the feedback control, thus allowing 5-ALAS induction to occur, e.g. upon glucocorticoid induction of TDO during fasting (starvation) and exogenous glucocorticoid administration. Currently, glucose therapy is the preferred strategy for reversing moderate attacks induced by fasting (calorie restriction), with more severe attacks being treated by intravenous heme preparations. Reversal of fasting-induced attacks by glucose is explained by the previously demonstrated reversal of increased heme utilisation by TDO. Inhibitors of this utilisation are therefore potential therapeutic targets in acute attacks and also for maintenance of a symptomless state. Existing TDO inhibitors other than glucose include allopurinol, nicotinamide and recently developed potent inhibitors such as LM10 used in cancer therapy. Based on studies in rats, the hypothesis predicts that the safety or otherwise of drugs in the hepatic porphyrias is determined by their ability to inhibit TDO utilisation of heme under basal conditions or after glucocorticoid induction or heme activation of TDO, in parallel with reciprocal changes in 5-ALAS induction. Tryptophan is also proposed as a potential therapy of acute attacks either alone or as an adjunct to the recently proposed 5-ALAS1 gene silencing. Trp increases heme biosynthesis by enhancing 5-ALA dehydratase activity and, based on a Trp-5-ALA model presented herein, Trp offers several advantages over heme therapy, namely rapid conversion of 5-ALA into heme, a greatly enhanced heme availability, a near complete inhibition of 5-ALAS induction, assumed rapid clearance of 5-ALA and hence accelerated resolution of symptoms of attacks, and finally provision of the neuroprotective metabolite kynurenic acid to neutralise the neurological symptoms. The hypothesis also addresses heme regulation in species lacking the TDO free apoenzyme and its glucocorticoid induction mechanism and proposes detailed assessment of heme biosynthesis in these species. Detailed proposals for testing the hypothesis are presented.


Asunto(s)
5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Hemo/metabolismo , Terapia Molecular Dirigida , Porfirias Hepáticas/tratamiento farmacológico , Triptófano Oxigenasa/antagonistas & inhibidores , Triptófano/uso terapéutico , 5-Aminolevulinato Sintetasa/genética , Alopurinol/farmacología , Alopurinol/uso terapéutico , Animales , Inducción Enzimática/efectos de los fármacos , Ayuno/metabolismo , Retroalimentación Fisiológica , Silenciador del Gen , Glucosa/metabolismo , Glucosa/uso terapéutico , Cobayas , Hemo/uso terapéutico , Humanos , Quinurenina/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Modelos Biológicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/fisiología , Porfirias Hepáticas/inducido químicamente , Porfirias Hepáticas/genética , Porfirias Hepáticas/metabolismo , Roedores , Especificidad de la Especie , Triptófano/efectos adversos , Triptófano/farmacología
12.
Clin Nucl Med ; 43(8): 617-618, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29894336

RESUMEN

An F-FDG PET/CT was performed on a 43-year-old woman with photosensitive skin rash, abnormal liver function, and pancytopenia, which demonstrated prominent hepatomegaly, splenomegaly, and diffuse liver F-FDG avidity. The liver biopsy revealed intrahepatic cholestasis with biliary fibrosis resulting from the deposition of protoporphyrin. X-linked erythroid-specific 5-aminolevulinate synthase gene analysis proved the diagnosis of X-linked protoporphyria.


Asunto(s)
Fluorodesoxiglucosa F18/metabolismo , Hígado/diagnóstico por imagen , Hígado/metabolismo , Porfirias Hepáticas/diagnóstico por imagen , Porfirias Hepáticas/genética , Tomografía Computarizada por Tomografía de Emisión de Positrones , Adulto , Femenino , Humanos , Porfirias Hepáticas/metabolismo
13.
Biochim Biophys Acta ; 1180(2): 201-6, 1992 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-1463771

RESUMEN

Swelling of isolated rat liver mitochondria is shown to be induced by metal-catalyzed 5-aminolevulinic acid (ALA) aerobic oxidation, a putative endogenous source of reactive oxygen species (ROS), at concentrations as low as 50-100 microM. In this concentration range, ALA is estimated to occur in the liver of acute intermittent porphyria patients. Removal of Ca2+ (10 microM) from the suspension of isolated rat liver mitochondria by added EGTA abolishes both the ALA-induced transmembrane-potential collapse and mitochondrial swelling. Prevention of the ALA-induced swelling by addition of ruthenium red prior to mitochondrial energization by succinate demonstrates the deleterious involvement of internal Ca2+. Addition of MgCl2 at concentrations higher than 2.5 mM, prevents the ALA-induced mitochondrial swelling, transmembrane potential collapse and Ca2+ efflux. This indicates that Mg2+ protects against the mitochondrial damage promoted by ALA-generated ROS. The ALA-induced mitochondrial damage might be a key event in the liver mitochondrial damage of acute intermittent porphyria patients reported elsewhere.


Asunto(s)
Ácido Aminolevulínico/farmacología , Calcio/farmacología , Mitocondrias Hepáticas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ácido Aminolevulínico/antagonistas & inhibidores , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Ácido Egtácico , Magnesio/farmacología , Masculino , Mitocondrias Hepáticas/metabolismo , Oxidación-Reducción , Porfirias Hepáticas/metabolismo , Ratas , Ratas Wistar
14.
Eur J Hum Genet ; 10(10): 649-57, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12357337

RESUMEN

Variegate porphyria (VP) is an inherited metabolic disease resulting from the partial deficiency of protoporphyrinogen oxidase, the penultimate enzyme in the heme biosynthetic pathway. We have evaluated the clinical and biochemical outcome of 103 Finnish VP patients diagnosed between 1966 and 2001. Fifty-two per cent of patients had experienced clinical symptoms: 40% had photosensitivity, 27% acute attacks and 14% both manifestations. The proportion of patients with acute attacks has decreased dramatically from 38 to 14% in patients diagnosed before and after 1980, whereas the prevalence of skin symptoms had decreased only subtly from 45 to 34%. We have studied the correlation between PPOX genotype and clinical outcome of 90 patients with the three most common Finnish mutations I12T, R152C and 338G-->C. The patients with the I12T mutation experienced no photosensitivity and acute attacks were rare (8%). Therefore, the occurrence of photosensitivity was lower in the I12T group compared to the R152C group (P=0.001), whereas no significant differences between the R152C and 338G-->C groups could be observed. Biochemical abnormalities were significantly milder suggesting a milder form of the disease in patients with the I12T mutation. In all VP patients, normal excretion of protoporphyrin in faeces in adulthood predicted freedom from both skin symptoms and acute attacks. The most valuable test predicting an increased risk of symptoms was urinary coproporphyrin, but only a substantially increased excretion exceeding 1,000 nmol/day was associated with an increased risk of both skin symptoms and acute attacks. All patients with an excretion of more than 1,000 nmol/day experienced either skin symptoms, acute attacks, or both.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Oxidorreductasas/genética , Porfirias Hepáticas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Coproporfirinas/metabolismo , Coproporfirinas/orina , Heces/química , Femenino , Finlandia , Flavoproteínas , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales , Oxidorreductasas/metabolismo , Fenotipo , Porfirias Hepáticas/metabolismo , Protoporfirinógeno-Oxidasa , Protoporfirinas/metabolismo , Relación Estructura-Actividad , Uroporfirinas/orina
15.
Clin Pharmacol Ther ; 56(1): 9-13, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8033500

RESUMEN

We investigated the effects of heme on metabolism of coumarin, debrisoquin, caffeine, and lidocaine in seven female patients with variegate porphyria and in 10 healthy men. During baseline conditions metabolism of the drugs was identical in the two groups. Compared with the results without heme, a single infusion of heme arginate (3 mg/kg heme) significantly decreased the debrisoquin/4-hydroxy-debrisoquin metabolic ratio in subjects with porphyria (p = 0.016) and in the control subjects (p = 0.016) and increased formation of monoethylglycinexylidide from lidocaine (p = 0.016 and p = 0.004, respectively). Metabolism of coumarin and caffeine was not affected by heme. Our results show that, in patients with porphyria and in healthy subjects, exogenous heme is able to accelerate the reactions mediated by the cytochrome isozymes CYP2D6 (debrisoquin) and CYP3A4 (lidocaine) but not reactions mediated by CYP1A2 (caffeine) and CYP2A6 (coumarin). This suggests that influence of heme on drug metabolism is P450 isozyme-specific.


Asunto(s)
Arginina/farmacología , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Hemo/farmacología , Porfirias Hepáticas/metabolismo , Adulto , Cafeína/metabolismo , Cumarinas/metabolismo , Debrisoquina/metabolismo , Femenino , Humanos , Isoenzimas/efectos de los fármacos , Lidocaína/metabolismo , Masculino , Persona de Mediana Edad , Porfirias Hepáticas/enzimología , Valores de Referencia
16.
Clin Liver Dis ; 2(1): 77-102, vi, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15560047

RESUMEN

The porphyrias are metabolic disorders characterized by abnormal heme biosynthesis with excessive accumulation and excretion of porphyrias or porphyrin precursors. Defects in the enzymes of the heme biosynthetic pathway result in porphyria. Several of the disorders have been classified as hepatic because the major site of the biochemical defect has been localized to the liver. This article describes the enzymes of the heme biosynthetic pathway, the clinical features of the hepatic porphyrias and management of the disorders.


Asunto(s)
Porfirias Hepáticas/metabolismo , Hemo/biosíntesis , Humanos , Hígado/metabolismo , Porfiria Cutánea Tardía/metabolismo , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/terapia
17.
Biochem Pharmacol ; 58(5): 887-96, 1999 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10449201

RESUMEN

When patients with acute porphyrias are treated with antihypertensives and analgesics, they could be placed at increased risk of developing porphyric attacks, since little is known about the potential for many of these drugs to induce these attacks. We used primary chick embryo liver cells, which maintain intact heme synthesis and regulation, to study the effects of antihypertensives and analgesics on porphyrin accumulation. Cells were treated with desferrioxamine to block heme synthesis partially, simulating conditions encountered in porphyric patients. Typically, cells were treated for 20 hr with the test drugs (3.16 to 1000 microM), along with desferrioxamine. Porphyrins were measured spectrofluorometrically, as uro-, copro,- and protoporphyrin. The evaluated drugs included six antihypertensives (two calcium channel blockers, an angiotensin receptor antagonist, and three inhibitors of angiotensin converting enzyme) and eight analgesics. Of the calcium channel blockers tested, nifedipine greatly increased porphyrin accumulation, whereas diltiazem caused only a slight increase. Losartan (an angiotensin receptor antagonist), captopril, or lisinopril (two angiotensin converting enzyme inhibitors) produced only small increases in porphyrin accumulation. In contrast, enalapril (another angiotensin converting enzyme inhibitor) substantially increased porphyrin accumulation when given in high concentrations. Among the analgesics tested, fentanyl and tramadol produced the highest porphyrin accumulations. Nalbuphine, hydrocodone, oxycodone, and dezocine were moderately or weakly porphyrogenic, whereas buprenorphine and morphine did not increase porphyrin accumulation. These studies suggest that patients with acute porphyrias may be at greater risk for developing porphyric attacks when treated with nifedipine (compared with diltiazem), enalapril (compared with captopril or lisinopril), and tramadol (compared with the other analgesics).


Asunto(s)
Analgésicos/farmacología , Antihipertensivos/farmacología , Porfirias Hepáticas/inducido químicamente , Porfirinas/metabolismo , Analgésicos/efectos adversos , Animales , Antihipertensivos/efectos adversos , Células Cultivadas , Embrión de Pollo , Hígado/citología , Hígado/efectos de los fármacos , Porfirias Hepáticas/metabolismo
18.
Clin Biochem ; 33(6): 465-73, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11074238

RESUMEN

OBJECTIVES: To describe the biochemical and clinical features in hereditary coproporphyria (HCP). DESIGN AND METHOD: Within the last 20 years, we investigated 53 patients (male:female = 1:2.5; age = 8-86 years) suffering from HCP. We describe the characteristic levels of urine, and fecal porphyrins and their precursors in hereditary coproporphyria and present the clinical features. Especially, we measured the coproporphyrin isomers I and III. RESULTS AND CONCLUSION: The group of hereditary coproporphyria patients exhibited a significantly higher (p<0.0001) excretion of urinary porphyrin precursors, delta-aminolevulinic acid (median = 84 micromol/24 h) and porphobilinogen (median = 39 micromol/24 h), as compared to controls (delta-aminolevulinic acid: 22 micromol/24 h, porphobilinogen: 3 micromol/24 h; median, n = 20). The median of coproporphyrin in urine (1315 nmol/24 h) and feces (1855 nmol/g) were enhanced 12- and 168-fold, as compared to healthy subjects (urinary coproporphyrin: 106 nmol/24 h, fecal coproporphyrin: 11 nmol/g; median, n = 20). During therapy on one female patient, with IV application of heme arginate, a considerable decline of porphyrin precursors and porphyrin excretion was observed. The examination of urinary and fecal coproporphyrin isomers I and III revealed an excessive elevation of the coproporphyrin isomer III of 87% in urine and 94% in feces, respectively (normal: urinary isomer III = 69-83% and fecal isomer III = 25-40%). In feces the increase of isomer III caused an inversion of the physiologic coproporphyrin isomer III:I ratio that could be recognized in all various stages in hereditary coproporphyria and in children. Acute attacks of hereditary coproporphyria are accompanied by an acute polysymptomatic clinical syndrome, and this is associated with high levels of urinary porphyrin precursors. On review of our patients, the highest percentage had abdominal pain (89%), followed by neurologic (33%), psychiatric (28%), cardiovascular (25%), and skin symptoms (14%).


Asunto(s)
Ácido Aminolevulínico/análisis , Coproporfirinas/análisis , Heces/química , Porfobilinógeno/análisis , Porfirias Hepáticas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Ácido Aminolevulínico/orina , Arginina/uso terapéutico , Niño , Cromatografía Líquida de Alta Presión , Coproporfirinógeno Oxidasa/genética , Coproporfirinas/orina , Femenino , Alemania , Hemo/uso terapéutico , Heterocigoto , Humanos , Isomerismo , Masculino , Persona de Mediana Edad , Porfobilinógeno/orina , Porfirias Hepáticas/diagnóstico , Porfirias Hepáticas/tratamiento farmacológico , Porfirias Hepáticas/genética , Uroporfirinas/análisis , Uroporfirinas/orina
19.
Toxicology ; 84(1-3): 199-215, 1993 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-8266338

RESUMEN

2-[1-(Ethoxyimino)propyl]-3-hydroxy-5-(2,4,6-trimethylphenyl) cyclohex-2-enone (ETC) is a novel alkyl ketone herbicide. Continuous administration of ETC to mice for 28 days resulted in marked liver enlargement and severe intrahepatic cholestasis. These effects have been shown to result directly from a rapid and marked accumulation of porphyrin in the liver. The porphyrin which accumulates in the liver has been identified as protoporphyrin IX and dose response and time course studies confirm prior inhibition of mitochondrial ferrochelatase as the causal lesion. ETC was a very potent porphyrinogenic compound in mice, with a no-effect level for a single oral dose of 1 mg/kg. Rats and hamsters were insensitive to this type of hepatotoxicity following single oral doses of up to 750 mg/kg or following repeated, and indeed prolonged administration. The sensitivity of different species to ETC-induced porphyria correlated with the effect of ETC on hepatic ferrochelatase activity. The inhibition of ferrochelatase activity and the hepatic porphyria in mice were both found to be readily reversible upon withdrawal of ETC.


Asunto(s)
Ciclohexanonas/toxicidad , Hemo/biosíntesis , Herbicidas/toxicidad , Hígado/efectos de los fármacos , Porfirias Hepáticas/metabolismo , Administración Oral , Animales , Cricetinae , Femenino , Ferroquelatasa/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Mesocricetus , Ratones , Porfirias Hepáticas/inducido químicamente , Porfirinas/química , Ratas , Ratas Wistar , Especificidad de la Especie
20.
Toxicol Lett ; 122(1): 97-102, 2001 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-11397561

RESUMEN

The effect of vitamin E treatment on total porphyrin content, lipid peroxidation (LOOH) and 8-hydroxydeoxyguanosine (8-OHdG) was studied in the livers of C57BL/10ScSn mice following hexachlorobenzene (HCB) and iron treatment. HCB was administered i.p. (totalling 300 mg/kg) twice, with 1 week interval. Three days after the first HCB injection iron-dextran was given i.p. (500 mg Fe per kg). Vitamin E was administered weekly (20 mg/kg) by s.c. injection. Both total hepatic porphyrin and LOOH levels were significantly (P<0.001) increased in the HCB-iron treated group as compared with the control group. Mice treated additionally with vitamin E had significant (P<0.001) lower levels as compared with the HCB-iron group. Similarly, the levels of 8-OHdG were significantly (P<0.001) increased above controls after HCB-iron treatment and this increase was reduced after co-treatment with vitamin E (P<0.02). The data support the hypothesis that the mechanism of hepatic porphyrinogenicity of HCB with iron overload is an oxidative free radical process.


Asunto(s)
Desoxiguanosina/metabolismo , Hexaclorobenceno/toxicidad , Hierro/toxicidad , Porfirias Hepáticas/prevención & control , Vitamina E/farmacología , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Desoxiguanosina/análogos & derivados , Hierro/metabolismo , Peróxidos Lipídicos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Porfirias Hepáticas/inducido químicamente , Porfirias Hepáticas/metabolismo , Porfirinas/metabolismo , Vitamina E/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda