Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Drug Metab Dispos ; 50(6): 781-797, 2022 06.
Article in English | MEDLINE | ID: mdl-34154993

ABSTRACT

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.


Subject(s)
Acetylgalactosamine , Porphyrias, Hepatic , Acetylgalactosamine/pharmacokinetics , Asialoglycoprotein Receptor/metabolism , Hepatocytes/metabolism , Humans , Porphyrias, Hepatic/metabolism , RNA, Small Interfering/genetics
2.
Clin Pharmacol Ther ; 110(5): 1250-1260, 2021 11.
Article in English | MEDLINE | ID: mdl-34510420

ABSTRACT

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.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions/physiology , Enzyme Activation/physiology , Liver/metabolism , Pyrrolidines/metabolism , RNA, Small Interfering/metabolism , 5-Aminolevulinate Synthetase/metabolism , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/metabolism , Adult , Caffeine/administration & dosage , Caffeine/metabolism , Cross-Over Studies , Enzyme Activation/drug effects , Female , Humans , Liver/drug effects , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Midazolam/administration & dosage , Midazolam/metabolism , Middle Aged , Omeprazole/administration & dosage , Omeprazole/metabolism , Porphyrias, Hepatic/drug therapy , Porphyrias, Hepatic/metabolism , Pyrrolidines/administration & dosage
3.
Dtsch Med Wochenschr ; 146(15): 955-958, 2021 08.
Article in German | MEDLINE | ID: mdl-34344029

ABSTRACT

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.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/drug therapy , Pyrrolidines , RNA, Small Interfering , RNAi Therapeutics , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/therapeutic use , Humans , Porphobilinogen Synthase/metabolism , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/physiopathology , Porphyrias, Hepatic/prevention & control , Pyrrolidines/administration & dosage , Pyrrolidines/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , RNA, Small Interfering/ultrastructure , Randomized Controlled Trials as Topic
4.
Methods Mol Biol ; 2282: 57-75, 2021.
Article in English | MEDLINE | ID: mdl-33928570

ABSTRACT

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.


Subject(s)
Liver Diseases/therapy , RNA Interference , RNA, Small Interfering/therapeutic use , RNAi Therapeutics , Acetylgalactosamine/analogs & derivatives , Acetylgalactosamine/therapeutic use , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/metabolism , Amyloid Neuropathies, Familial/therapy , Animals , Gene Transfer Techniques , Humans , Liver Diseases/genetics , Liver Diseases/metabolism , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/therapy , Pyrrolidines/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
5.
Drugs ; 80(3): 335-339, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32034693

ABSTRACT

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.


Subject(s)
5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/analogs & derivatives , Drug Approval , Enzyme Inhibitors/pharmacology , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/drug therapy , Pyrrolidines/pharmacology , 5-Aminolevulinate Synthetase/metabolism , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/pharmacology , Enzyme Inhibitors/administration & dosage , Humans , Porphobilinogen Synthase/metabolism , Porphyrias, Hepatic/metabolism , Pyrrolidines/administration & dosage , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism
6.
Med Hypotheses ; 131: 109314, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31443750

ABSTRACT

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.


Subject(s)
5-Aminolevulinate Synthetase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Heme/metabolism , Molecular Targeted Therapy , Porphyrias, Hepatic/drug therapy , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan/therapeutic use , 5-Aminolevulinate Synthetase/genetics , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Enzyme Induction/drug effects , Fasting/metabolism , Feedback, Physiological , Gene Silencing , Glucose/metabolism , Glucose/therapeutic use , Guinea Pigs , Heme/therapeutic use , Humans , Kynurenine/metabolism , Liver/drug effects , Liver/enzymology , Models, Biological , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/genetics , Porphyrias, Hepatic/metabolism , Rodentia , Species Specificity , Tryptophan/adverse effects , Tryptophan/pharmacology
7.
Clin Nucl Med ; 43(8): 617-618, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29894336

ABSTRACT

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.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Liver/diagnostic imaging , Liver/metabolism , Porphyrias, Hepatic/diagnostic imaging , Porphyrias, Hepatic/genetics , Positron Emission Tomography Computed Tomography , Adult , Female , Humans , Porphyrias, Hepatic/metabolism
8.
Hepatology ; 66(4): 1314-1322, 2017 10.
Article in English | MEDLINE | ID: mdl-28605040

ABSTRACT

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).


Subject(s)
Porphyrias, Hepatic/therapy , Disease Management , Humans , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/metabolism
9.
Curr Opin Hematol ; 24(3): 198-207, 2017 May.
Article in English | MEDLINE | ID: mdl-28118224

ABSTRACT

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.


Subject(s)
Porphobilinogen Synthase/deficiency , Porphyria, Erythropoietic/etiology , Porphyria, Erythropoietic/metabolism , Porphyrias, Hepatic/etiology , Porphyrias, Hepatic/metabolism , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , 5-Aminolevulinate Synthetase/therapeutic use , Age Factors , Animals , Biomarkers , Chronic Pain/etiology , Enzyme Activation , Erythrocytes/metabolism , Gene Expression Regulation , Genes, X-Linked , Genetic Association Studies , Genetic Predisposition to Disease , Heme/biosynthesis , Humans , Mutation , Phenotype , Porphobilinogen Synthase/metabolism , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/therapy , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/therapy , RNA, Small Interfering/genetics
10.
Mol Genet Metab ; 119(3): 278-283, 2016 11.
Article in English | MEDLINE | ID: mdl-27595545

ABSTRACT

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.


Subject(s)
Patients/psychology , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/physiopathology , Porphyrias, Hepatic/psychology , Adult , Aged , Female , Heme/biosynthesis , Heme/metabolism , Humans , Male , Middle Aged , Physician-Patient Relations , Porphobilinogen Synthase/metabolism , Porphyrias, Hepatic/epidemiology , Porphyrias, Hepatic/metabolism , Quality of Life
11.
Eksp Klin Gastroenterol ; (7): 69-77, 2015.
Article in English, Russian | MEDLINE | ID: mdl-26817125

ABSTRACT

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.


Subject(s)
Abdominal Pain , Neuralgia , Porphyrias, Hepatic , Abdominal Pain/diagnosis , Abdominal Pain/metabolism , Abdominal Pain/pathology , Abdominal Pain/physiopathology , Adult , Female , Humans , Neuralgia/diagnosis , Neuralgia/metabolism , Neuralgia/pathology , Neuralgia/physiopathology , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , Porphyrias, Hepatic/physiopathology
12.
J Cell Sci ; 126(Pt 14): 3105-12, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23641075

ABSTRACT

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.


Subject(s)
Fatty Liver/metabolism , Lamin Type A/metabolism , Lamin Type B/metabolism , Porphyrias, Hepatic/metabolism , Animals , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/genetics , Ferrochelatase/genetics , GTP-Binding Proteins/antagonists & inhibitors , Hep G2 Cells , Humans , Mallory Bodies/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Oxidative Stress , Porphyrias, Hepatic/complications , Porphyrias, Hepatic/genetics , Protein Glutamine gamma Glutamyltransferase 2 , Protein Multimerization/drug effects , Protein Transport/drug effects , Protoporphyrins/pharmacology , Pyridines/toxicity , Transglutaminases/antagonists & inhibitors
13.
J Toxicol Sci ; 33(5): 599-608, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19043281

ABSTRACT

Most patients with hepatic porphyria exhibit neuropsychiatric symptoms, including abdominal pain, peripheral neuropathy, confusion, insomnia and mental disturbances such as anxiety and depression. Although heme deficiency and accumulation of heme precursors are thought to be responsible for neuropsychiatric manifestations in patients with acute porphyria, the pathogenetic mechanisms remain poorly understood. In the present study, we observed psychiatric behaviors in mice with hepatic porphyria induced by the ingestion of a griseofulvin (GF)-containing diet over a period of 12 weeks. GF ingestion by the mice caused an accumulation of porphyrins in the feces and a decrease in heme in the liver; these effects were observed throughout the entire duration of the experiment, with maximum levels observed after circa 1 week of ingestion of this diet. In addition, the mice developed enlargement of the liver, hepatocyte injury, and cholestasis. Mice with hepatic porphyria manifested an anxiety-like behavior by the long-term treatment (over 5 weeks) in a GF-dose and duration dependent manner. The hepatic porphyria mice also manifested depression-like behaviors by the short-term treatment (3 weeks) of GF2.0, which was reversed by administration of anti-depressant, imipramine. In conclusion, this study for the first time demonstrated psychiatric manifestations in GF-induced hepatic porphyria mice. The present results suggest that model animals could be useful for elucidating the mechanisms underlying psychiatric manifestations in syndromes such as hepatic porphyria and hepatic encephalopathy that are associated with the impairment of hepatic function.


Subject(s)
Anxiety/psychology , Behavior, Animal , Depression/psychology , Griseofulvin/toxicity , Heme/biosynthesis , Porphyrias, Hepatic/psychology , Animals , Anxiety/etiology , Anxiety/metabolism , Behavior, Animal/drug effects , Cytochrome P-450 Enzyme System/metabolism , Depression/etiology , Depression/metabolism , Disease Models, Animal , Enzymes/blood , Feces/chemistry , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred ICR , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/complications , Porphyrias, Hepatic/metabolism , Swimming
14.
Toxicol Pathol ; 33(6): 720-5, 2005.
Article in English | MEDLINE | ID: mdl-16263697

ABSTRACT

As part of regulatory safety testing program, a 13-week oral toxicity study with a new antipsychotic drug candidate was performed in beagle dogs. During this study, dark red/brown feces were recorded in treated dogs and increases in liver parameters (alanine aminotransferase, alkaline phosphatase, bilirubin) were measured biochemically. At the end of the study, livers of high-dose (50 mg/kg) animals were (mottled) dark brown, sometimes with pale foci. Histopathological examination of these livers showed dark globular pigment deposits in the hepatocellular cytoplasm and within the bile canaliculi. Varying numbers of inflammatory cell infiltrates were additionally present in association with the deposits. These pigment deposits showed birefringency with characteristic "Maltese Cross"-like structures under polarized light. Electronmicroscopy revealed the typical, so-called "sunburst" pattern with radiating double-lined crystalline structures. These morphologic characteristics strongly indicated at the presence of porphyrins, which was definitely confirmed biochemically. Published reports of drug-induced hepatic porphyria in dogs are rare. The possible underlying mechanism in the dog and man is discussed.


Subject(s)
Antipsychotic Agents/toxicity , Liver/drug effects , Porphyrias, Hepatic/chemically induced , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Antipsychotic Agents/administration & dosage , Bilirubin/blood , Blood Coagulation/drug effects , Dogs , Dose-Response Relationship, Drug , Feces/chemistry , Female , Hyperplasia , Liver/enzymology , Liver/pathology , Male , Organ Size/drug effects , Partial Thromboplastin Time , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , Protoporphyrins/analysis , Protoporphyrins/metabolism , Thymus Gland/drug effects , Thymus Gland/pathology , Toxicity Tests
15.
Br J Dermatol ; 149(6): 1266-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14674906

ABSTRACT

BACKGROUND: Porphyrinogens are the obligate intracellular precursors of haem. These compounds are, however, unstable and are easily oxidized to the corresponding porphyrins, which are the form in which they are usually measured in the laboratory. A substantial enterohepatic cycling of porphyrins has been shown. Administration of oral activated charcoal, by interrupting this cycle, may reduce plasma and urine porphyrin levels in patients with some forms of porphyria. The effect of charcoal in subjects with variegate porphyria (VP) has not been reported. OBJECTIVES: To determine the clinical and biochemical effects of the administration of oral activated charcoal in patients with VP. METHODS: Oral activated charcoal was administered to eight subjects with VP. Clinical activity was assessed by skin lesion counts fortnightly for 6 weeks, 6 weeks after cessation of therapy, and during a subsequent 6-week control period during which no charcoal was taken. Urine and plasma porphyrins and urine precursors were measured by standard techniques. RESULTS: Treatment resulted in a significant increase in skin disease, urine and plasma porphyrins. CONCLUSIONS: Oral charcoal administration results in a paradoxical aggravation of VP, suggesting a complex and as yet undefined interaction of hepatic porphyrin metabolism and bowel porphyrin reabsorption. Oral sorbents should not be prescribed to subjects with VP.


Subject(s)
Charcoal/administration & dosage , Porphyrias, Hepatic/drug therapy , Administration, Oral , Adolescent , Adult , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Analysis of Variance , Female , Humans , Male , Porphobilinogen/blood , Porphobilinogen/urine , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , Porphyrins/blood , Porphyrins/urine , Prospective Studies , South Africa , Treatment Failure
16.
Cell Mol Biol (Noisy-le-grand) ; 49(4): 493-500, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12899439

ABSTRACT

The porphyrias are a group of inherited metabolic disorders of heme biosynthesis which result from a partial deficiency in one of its seven specific enzymes, after its first and rate limiting enzyme, delta-aminolevulinic acid synthetase. They can be classified on the basis of their clinical manifestations into cutaneous, acute and mixed disorders. Acute intermittent porphyria (AIP) is the most common type of hepatic acute porphyrias, inherited as an autosomal dominant trait, caused by a defect in the gene which codifies for the heme enzyme porphobilinogen deaminase. Its prevalence in the Argentinean population is about 1:125,000. A partial deficiency in another enzyme, protoporphyrinogen oxidase, produces variegate porphyria (VP), the second acute porphyria most frequent in the Argentinean population (1:600,000). Here, we review all the mutations we have found in 46 AIP and 9 VP unrelated Argentinean patients. To screen for mutations in symptomatic patients, we have proposed a geneticresearch strategy.


Subject(s)
Porphyria, Acute Intermittent/genetics , Porphyrias, Hepatic/genetics , Argentina , Humans , Mutation , Porphyria, Acute Intermittent/epidemiology , Porphyria, Acute Intermittent/metabolism , Porphyrias, Hepatic/epidemiology , Porphyrias, Hepatic/metabolism
17.
Toxicol Appl Pharmacol ; 189(1): 28-38, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12758057

ABSTRACT

Hepatic uroporphyria can be readily induced by a variety of treatments in mice of the C57BL strains, whereas DBA/2 mice are almost completely resistant. However, feeding of the protoporphyrinogen oxidase-inhibiting herbicide fomesafen (0.25% in the diet for 18 weeks) induced hepatic uroporphyria in male DBA/2N mice (liver porphyrin content up to 150 nmol/g, control animals 1 nmol/g), whereas fomesafen-treated male C57BL/6N mice displayed only a slight elevation of liver porphyrins (approximately 5 nmol/g). The profile of accumulated hepatic porphyrins in fomesafen-treated DBA/2N mice resembled the well-characterised uroporphyria induced by polyhalogenated aromatic hydrocarbons, while histological examination confirmed the presence of uroporphyria-specific cytoplasmic inclusions in the hepatocytes. Uroporphyrinogen decarboxylase activity decreased to about 30% of control values in fomesafen-treated DBA/2N mice; microsomal methoxyresorufin O-dealkylase activity was slightly reduced. The amount of CYP1A1 and CYP1A2 mRNA, as determined by real-time PCR, was not significantly changed; mRNA encoding the housekeeping 5-aminolevulinic acid synthase was elevated 10-fold. Total liver iron was slightly increased. A similar uroporphyria was induced by the herbicide formulation Blazer, containing a structurally related herbicide acifluorfen, when fed to DBA/2N mice at a dose corresponding to 0.25% of acifluorfen in the diet. Since DBA/2 mice are almost completely resistant to all well-characterised porphyrogenic chemicals, the results suggest the possible existence of a yet unknown mechanism of uroporphyria induction, to which the DBA/2 mouse strain is more sensitive than the C57BL strain.


Subject(s)
Benzamides/toxicity , Herbicides/toxicity , Phenyl Ethers/toxicity , Porphyrias, Hepatic/chemically induced , Uroporphyrins/biosynthesis , Animals , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Polymerase Chain Reaction , Porphyrias, Hepatic/enzymology , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , RNA, Messenger/biosynthesis , Species Specificity , Uroporphyrins/analysis
18.
Eur J Hum Genet ; 10(10): 649-57, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12357337

ABSTRACT

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.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/genetics , Porphyrias, Hepatic/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Coproporphyrins/metabolism , Coproporphyrins/urine , Feces/chemistry , Female , Finland , Flavoproteins , Genotype , Humans , Male , Middle Aged , Mitochondrial Proteins , Oxidoreductases/metabolism , Phenotype , Porphyrias, Hepatic/metabolism , Protoporphyrinogen Oxidase , Protoporphyrins/metabolism , Structure-Activity Relationship , Uroporphyrins/urine
19.
Toxicol Lett ; 122(1): 97-102, 2001 May 31.
Article in English | MEDLINE | ID: mdl-11397561

ABSTRACT

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.


Subject(s)
Deoxyguanosine/metabolism , Hexachlorobenzene/toxicity , Iron/toxicity , Porphyrias, Hepatic/prevention & control , Vitamin E/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Deoxyguanosine/analogs & derivatives , Iron/metabolism , Lipid Peroxides/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/metabolism , Porphyrins/metabolism , Vitamin E/metabolism
20.
Epilepsia ; 42(6): 793-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11422339

ABSTRACT

PURPOSE: Despite the development of new antiepileptic agents (AEDs), the therapy of epilepsies along with hepatic porphyrias remains difficult. Most AEDs such as carbamazepine (CBZ), phenytoin (PHT), valproate (VPA), and lamotrigine (LTG) may precipitate clinically latent porphyria by inducing hepatic metabolism and increasing hepatic heme synthesis. Actually, only gabapentin (GBP), an AED without any hepatic metabolism, is known as a potential therapy for partial seizures in patients having hepatic forms of porphyria. METHODS: We present the case of a 28-year-old man with porphyria cutanea tarda (PCT) who has had pharmacoresistant epilepsy with complex partial and secondarily generalized seizures since early childhood. Despite having undergone several AED therapies over the years, no seizure-free interval had been observed. Only CBZ could cause a seizure reduction, but this treatment had to be discontinued as an elevation of the transaminases as well as pruritus and erythema were noted. The patient was then started on oxcarbazepine (OCBZ), a ketoanalogue of CBZ similar in its pharmacologic mechanism as well as its clinical use, but which, in contrast to CBZ, has only a low hepatic induction of microsomal enzymes. A final maintenance dose four times higher than that of CBZ was prescribed. RESULTS: In the follow-up, the patient stopped having seizures, and his liver functions became normal. CONCLUSIONS: It can be concluded that OCBZ can successfully be administered to patients with hepatic porphyria and focal epilepsy who did not respond to treatment with GBP.


Subject(s)
Amines , Anticonvulsants/therapeutic use , Carbamazepine/therapeutic use , Cyclohexanecarboxylic Acids , Epilepsies, Partial/drug therapy , Porphyrias, Hepatic/epidemiology , gamma-Aminobutyric Acid , Acetates/therapeutic use , Adult , Anticonvulsants/adverse effects , Carbamazepine/adverse effects , Carbamazepine/analogs & derivatives , Comorbidity , Drug Therapy, Combination , Enzyme Induction/drug effects , Epilepsies, Partial/epidemiology , Epilepsies, Partial/metabolism , Gabapentin , Germany/epidemiology , Humans , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Oxcarbazepine , Porphyria Cutanea Tarda/epidemiology , Porphyria Cutanea Tarda/metabolism , Porphyrias, Hepatic/metabolism , Transaminases/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...