Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 85
1.
Arch Biochem Biophys ; 738: 109540, 2023 04.
Article En | MEDLINE | ID: mdl-36746260

5-aminolevulinic acid (5-ALA) is the first precursor of the heme biosynthesis pathway, accumulated in acute intermittent porphyria (AIP), an inherited metabolic disease characterized by porphobilinogen deaminase deficiency. An increased incidence of hepatocellular carcinoma (HCC) has been reported as a long-term manifestation in symptomatic AIP patients. 5-ALA is an α-aminoketone prone to oxidation, yielding reactive oxygen species and 4,5-dioxovaleric acid. A high concentration of 5-ALA presents deleterious pro-oxidant potential. It can induce apoptosis, DNA damage, mitochondrial dysfunction, and altered expression of carcinogenesis-related proteins. Several hypotheses of the increased risk of HCC rely on the harmful effect of elevated 5-ALA in the liver of AIP patients, which could promote a pro-carcinogenic environment. We investigated the global transcriptional changes and perturbed molecular pathways in HepG2 cells following exposure to 5-ALA 25 mM for 2 h and 24 h using DNA microarray. Distinct transcriptome profiles were observed. 5-ALA '25 mM-2h' upregulated 10 genes associated with oxidative stress response and carcinogenesis. Enrichment analysis of differentially expressed genes by KEGG, Reactome, MetaCore™, and Gene Ontology, showed that 5-ALA '25 mM-24h' enriched pathways involved in drug detoxification, oxidative stress, DNA damage, cell death/survival, cell cycle, and mitochondria dysfunction corroborating the pro-oxidant properties of 5-ALA. Furthermore, our results disclosed other possible processes such as senescence, immune responses, endoplasmic reticulum stress, and also some putative effectors, such as sequestosome, osteopontin, and lon peptidase 1. This study provided additional knowledge about molecular mechanisms of 5-ALA toxicity which is essential to a deeper understanding of AIP and HCC pathophysiology. Furthermore, our findings can contribute to improving the efficacy of current therapies and the development of novel biomarkers and targets for diagnosis, prognosis, and therapeutic strategies for AHP/AIP and associated HCC.


Carcinoma, Hepatocellular , Liver Neoplasms , Porphyria, Acute Intermittent , Humans , Aminolevulinic Acid/metabolism , Aminolevulinic Acid/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Reactive Oxygen Species/metabolism , Liver Neoplasms/genetics , Transcriptome , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Carcinogenesis
2.
Biochemistry ; 61(21): 2409-2416, 2022 11 01.
Article En | MEDLINE | ID: mdl-36241173

Patients with major forms of acute hepatic porphyria present acute neurological attacks with overproduction of porphobilinogen (PBG) and δ-aminolevulinic acid (ALA). Even if ALA is considered the most likely agent inducing the acute symptoms, the mechanism of its accumulation has not been experimentally demonstrated. In the most frequent form, acute intermittent porphyria (AIP), inherited gene mutations induce a deficiency in PBG deaminase; thus, accumulation of the substrate PBG is biochemically obligated but not that of ALA. A similar scenario is observed in other forms of acute hepatic porphyria (i.e., porphyria variegate, VP) in which PBG deaminase is inhibited by metabolic intermediates. Here, we have investigated the molecular basis of δ-aminolevulinate accumulation using in vitro fluxomics monitored by NMR spectroscopy and other biophysical techniques. Our results show that porphobilinogen, the natural product of δ-aminolevulinate deaminase, effectively inhibits its anabolic enzyme at abnormally low concentrations. Structurally, this high affinity can be explained by the interactions that porphobilinogen generates with the active site, most of them shared with the substrate. Enzymatically, our flux analysis of an altered heme pathway demonstrates that a minimum accumulation of porphobilinogen will immediately trigger the accumulation of δ-aminolevulinate, a long-lasting observation in patients suffering from acute porphyrias.


Porphyria, Acute Intermittent , Porphyrias, Hepatic , Humans , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Porphobilinogen , Hydroxymethylbilane Synthase/genetics , Hydroxymethylbilane Synthase/metabolism , Porphyrias, Hepatic/genetics
3.
Br J Pharmacol ; 179(14): 3815-3830, 2022 07.
Article En | MEDLINE | ID: mdl-35170015

BACKGROUND AND PURPOSE: Acute intermittent porphyria (AIP) is a rare disease caused by a genetic mutation in the hepatic activity of the porphobilinogen-deaminase. We aimed to develop a mechanistic model of the enzymatic restoration effects of a novel therapy based on the administration of different formulations of recombinant human-PBGD (rhPBGD) linked to the ApoAI lipoprotein. This fusion protein circulates in blood, incorporating into HDL and penetrating hepatocytes. EXPERIMENTAL APPROACH: Single i.v. dose of different formulations of rhPBGD linked to ApoAI were administered to AIP mice in which a porphyric attack was triggered by i.p. phenobarbital. Data consist on 24 h urine excreted amounts of heme precursors, 5-aminolevulinic acid (ALA), PBG and total porphyrins that were analysed using non-linear mixed-effects analysis. KEY RESULTS: The mechanistic model successfully characterized over time the amounts excreted in urine of the three heme precursors for different formulations of rhPBGD and unravelled several mechanisms in the heme pathway, such as the regulation in ALA synthesis by heme. Treatment with rhPBGD formulations restored PBGD activity, increasing up to 51 times the value of the rate of tPOR formation estimated from baseline. Model-based simulations showed that several formulation prototypes provided efficient protective effects when administered up to 1 week prior to the occurrence of the AIP attack. CONCLUSION AND IMPLICATIONS: The model developed had excellent performance over a range of doses and formulation type. This mechanistic model warrants use beyond ApoAI-conjugates and represents a useful tool towards more efficient drug treatments of other enzymopenias as well as for acute intermittent porphyria.


Porphyria, Acute Intermittent , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/urine , Animals , Disease Models, Animal , Heme , Mice , Mice, Inbred C57BL , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Recombinant Proteins
4.
Sci Transl Med ; 14(627): eabc0700, 2022 01 12.
Article En | MEDLINE | ID: mdl-35020410

Correction of enzymatic deficits in hepatocytes by systemic administration of a recombinant protein is a desired therapeutic goal for hepatic enzymopenic disorders such as acute intermittent porphyria (AIP), an inherited porphobilinogen deaminase (PBGD) deficiency. Apolipoprotein A-I (ApoAI) is internalized into hepatocytes during the centripetal transport of cholesterol. Here, we generated a recombinant protein formed by linking ApoAI to the amino terminus of human PBGD (rhApoAI-PBGD) in an attempt to transfer PBGD into liver cells. In vivo experiments showed that, after intravenous injection, rhApoAI-PBGD circulates in blood incorporated into high-density lipoprotein (HDL), penetrates into hepatocytes, and crosses the blood-brain barrier, increasing PBGD activity in both the liver and brain. Consistently, the intravenous administration of rhApoAI-PBGD or the hyperfunctional rApoAI-PBGD-I129M/N340S (rApoAI-PBGDms) variant efficiently prevented and abrogated phenobarbital-induced acute attacks in a mouse model of AIP. One month after a single intravenous dose of rApoAI-PBGDms, the protein was still detectable in the liver, and hepatic PBGD activity remained increased above control values. A long-lasting therapeutic effect of rApoAI-PBGDms was observed after either intravenous or subcutaneous administration. These data describe a method to deliver PBGD to hepatocytes with resulting enhanced hepatic enzymatic activity and protection against AIP attacks in rodent models, suggesting that the approach might be an effective therapy for AIP.


Hydroxymethylbilane Synthase , Porphyria, Acute Intermittent , Animals , Disease Models, Animal , Genetic Therapy/methods , Hydroxymethylbilane Synthase/metabolism , Hydroxymethylbilane Synthase/therapeutic use , Mice , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/metabolism
7.
Hum Mol Genet ; 29(19): 3211-3223, 2020 11 25.
Article En | MEDLINE | ID: mdl-32916704

The morphological changes that occur in the central nervous system of patients with severe acute intermittent porphyria (AIP) have not yet been clearly established. The aim of this work was to analyze brain involvement in patients with severe AIP without epileptic seizures or clinical posterior reversible encephalopathy syndrome, as well as in a mouse model receiving or not liver-directed gene therapy aimed at correcting the metabolic disorder. We conducted neuroradiologic studies in 8 severely affected patients (6 women) and 16 gender- and age-matched controls. Seven patients showed significant enlargement of the cerebral ventricles and decreased brain perfusion was observed during the acute attack in two patients in whom perfusion imaging data were acquired. AIP mice exhibited reduced cerebral blood flow and developed chronic dilatation of the cerebral ventricles even in the presence of slightly increased porphyrin precursors. While repeated phenobarbital-induced attacks exacerbated ventricular dilation in AIP mice, correction of the metabolic defect using liver-directed gene therapy restored brain perfusion and afforded protection against ventricular enlargement. Histological studies revealed no signs of neuronal loss but a denser neurofilament pattern in the periventricular areas, suggesting compression probably caused by imbalance in cerebrospinal fluid dynamics. In conclusion, severely affected AIP patients exhibit cerebral ventricular enlargement. Liver-directed gene therapy protected against the morphological consequences of the disease seen in the brain of AIP mice. The observational study was registered at Clinicaltrial.gov as NCT02076763.


Brain/pathology , Cerebral Ventricles/pathology , Disease Models, Animal , Hydroxymethylbilane Synthase/genetics , Porphyria, Acute Intermittent/physiopathology , Adult , Animals , Brain/metabolism , Case-Control Studies , Cerebral Ventricles/metabolism , Clinical Trials, Phase I as Topic , Female , Genetic Therapy , Humans , Male , Mice , Middle Aged , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Prospective Studies
8.
Genes (Basel) ; 11(8)2020 08 12.
Article En | MEDLINE | ID: mdl-32806544

Acute intermittent porphyria (AIP) results from a decreased activity of hepatic hydroxymethylbilane synthase (HMBS), the third enzyme in the heme biosynthetic pathway. AIP is an autosomal dominant disorder with incomplete penetrance, characterized by acute neurovisceral attacks precipitated by several factors that induce the hepatic 5-aminolevulinic acid synthase, the first enzyme in the heme biosynthesis. Thus, a deficiency in HMBS activity results in an overproduction of porphyrin precursors and the clinical manifestation of the disease. Early diagnosis and counselling are essential to prevent attacks, and mutation analysis is the most accurate method to identify asymptomatic carriers in AIP families. In the present study, we have investigated the molecular defects in 55 unrelated Spanish patients with AIP, identifying 32 HMBS gene mutations, of which six were novel and ten were found in more than one patient. The novel mutations included a missense, an insertion, two deletions, and two splice site variants. Prokaryotic expression studies demonstrated the detrimental effect for the missense mutation, whereas reverse transcription-PCR and sequencing showed aberrant splicing caused by each splice site mutation. These results will allow for an accurate diagnosis of carriers of the disease in these families. Furthermore, they increase the knowledge about the molecular heterogeneity of AIP in Spain.


Biomarkers , Porphyria, Acute Intermittent/etiology , Porphyria, Acute Intermittent/metabolism , Adolescent , Adult , DNA Mutational Analysis , Disease Susceptibility , Female , Genotype , Humans , Hydroxymethylbilane Synthase/genetics , Hydroxymethylbilane Synthase/metabolism , Male , Middle Aged , Mutation , Polymerase Chain Reaction , Porphyria, Acute Intermittent/diagnosis , RNA Splicing , Spain , Young Adult
9.
Blood ; 136(21): 2457-2468, 2020 11 19.
Article En | MEDLINE | ID: mdl-32678895

Congenital erythropoietic porphyria (CEP) is an inborn error of heme synthesis resulting from uroporphyrinogen III synthase (UROS) deficiency and the accumulation of nonphysiological porphyrin isomer I metabolites. Clinical features are heterogeneous among patients with CEP but usually combine skin photosensitivity and chronic hemolytic anemia, the severity of which is related to porphyrin overload. Therapeutic options include symptomatic strategies only and are unsatisfactory. One promising approach to treating CEP is to reduce the erythroid production of porphyrins through substrate reduction therapy by inhibiting 5-aminolevulinate synthase 2 (ALAS2), the first and rate-limiting enzyme in the heme biosynthetic pathway. We efficiently reduced porphyrin accumulation after RNA interference-mediated downregulation of ALAS2 in human erythroid cellular models of CEP disease. Taking advantage of the physiological iron-dependent posttranscriptional regulation of ALAS2, we evaluated whether iron chelation with deferiprone could decrease ALAS2 expression and subsequent porphyrin production in vitro and in vivo in a CEP murine model. Treatment with deferiprone of UROS-deficient erythroid cell lines and peripheral blood CD34+-derived erythroid cultures from a patient with CEP inhibited iron-dependent protein ALAS2 and iron-responsive element-binding protein 2 expression and reduced porphyrin production. Furthermore, porphyrin accumulation progressively decreased in red blood cells and urine, and skin photosensitivity in CEP mice treated with deferiprone (1 or 3 mg/mL in drinking water) for 26 weeks was reversed. Hemolysis and iron overload improved upon iron chelation with full correction of anemia in CEP mice treated at the highest dose of deferiprone. Our findings highlight, in both mouse and human models, the therapeutic potential of iron restriction to modulate the phenotype in CEP.


Anemia, Hemolytic/drug therapy , Deferiprone/therapeutic use , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Photosensitivity Disorders/drug therapy , Porphyria, Erythropoietic/drug therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/biosynthesis , 5-Aminolevulinate Synthetase/genetics , Adult , Anemia, Hemolytic/etiology , Animals , CRISPR-Cas Systems , Cell Line , Cell Line, Tumor , Disease Models, Animal , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Iron/metabolism , Iron Overload/etiology , Leukemia, Erythroblastic, Acute/pathology , Mice , Peripheral Blood Stem Cells/drug effects , Peripheral Blood Stem Cells/metabolism , Photosensitivity Disorders/etiology , Porphyria, Acute Intermittent/metabolism , Porphyria, Erythropoietic/complications , Porphyrins/biosynthesis , RNA Interference , RNA, Small Interfering/pharmacology
10.
Acta Neuropathol Commun ; 8(1): 38, 2020 03 20.
Article En | MEDLINE | ID: mdl-32197664

Acute intermittent porphyria (AIP) is an autosomal dominant inborn error of heme biosynthesis due to a pathogenic mutation in the Hmbs gene, resulting in half-normal activity of hydroxymethylbilane synthase. Factors that induce hepatic heme biosynthesis induce episodic attacks in heterozygous patients. The clinical presentation of acute attacks involves the signature neurovisceral pain and may include psychiatric symptoms. Here we used a knock-in mouse line that is biallelic for the Hmbs c.500G > A (p.R167Q) mutation with ~ 5% of normal hydroxymethylbilane synthase activity to unravel the consequences of severe HMBS deficiency on affective behavior and brain physiology. Hmbs knock-in mice (KI mice) model the rare homozygous dominant form of AIP and were used as tool to elucidate the hitherto unknown pathophysiology of the behavioral manifestations of the disease and its neural underpinnings. Extensive behavioral analyses revealed a selective depression-like phenotype in Hmbs KI mice; transcriptomic and immunohistochemical analyses demonstrated aberrant myelination. The uncovered compromised mitochondrial function in the hippocampus of knock-in mice and its ensuing neurogenic and neuroplastic deficits lead us to propose a mechanistic role for disrupted mitochondrial energy production in the pathogenesis of the behavioral consequences of severe HMBS deficiency and its neuropathological sequelae in the brain.


Behavior, Animal , Depression/metabolism , Hippocampus/metabolism , Hydroxymethylbilane Synthase/genetics , Mitochondria/metabolism , Porphyria, Acute Intermittent/metabolism , Anhedonia , Animals , Cell Proliferation , Depression/genetics , Depression/physiopathology , Depression/psychology , Disease Models, Animal , Elevated Plus Maze Test , Gene Knock-In Techniques , Hindlimb Suspension , Hippocampus/physiopathology , Homozygote , Immunohistochemistry , Long-Term Potentiation , Mice , Microscopy, Fluorescence , Myelin Sheath/metabolism , Neural Inhibition , Neural Stem Cells , Neurogenesis , Neuronal Plasticity , Patch-Clamp Techniques , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/physiopathology , Porphyria, Acute Intermittent/psychology
11.
Mol Ther ; 28(2): 677-689, 2020 02 05.
Article En | MEDLINE | ID: mdl-31810863

Mutations in hydroxymethylbilane synthase (HMBS) cause acute intermittent porphyria (AIP), an autosomal dominant disease where typically only one HMBS allele is mutated. In AIP, the accumulation of porphyrin precursors triggers life-threatening neurovisceral attacks and at long-term, entails an increased risk of hepatocellular carcinoma, kidney failure, and hypertension. Today, the only cure is liver transplantation, and a need for effective mechanism-based therapies, such as pharmacological chaperones, is prevailing. These are small molecules that specifically stabilize a target protein. They may be developed into an oral treatment, which could work curatively during acute attacks, but also prophylactically in asymptomatic HMBS mutant carriers. With the use of a 10,000 compound library, we identified four binders that further increased the initially very high thermal stability of wild-type HMBS and protected the enzyme from trypsin digestion. The best hit and a selected analog increased steady-state levels and total HMBS activity in human hepatoma cells overexpressing HMBS, and in an Hmbs-deficient mouse model with a low-expressed wild-type-like allele, compared to untreated controls. Moreover, the concentration of porphyrin precursors decreased in liver of mice treated with the best hit. Our findings demonstrate the great potential of these hits for the development of a pharmacological chaperone-based corrective treatment of AIP by enhancing wild-type HMBS function independently of the patients' specific mutation.


Biomarkers , Drug Discovery , Porphyria, Acute Intermittent/metabolism , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Mice , Mice, Knockout , Molecular Targeted Therapy , Porphyria, Acute Intermittent/etiology , Porphyria, Acute Intermittent/therapy , Protein Folding , Proteins/antagonists & inhibitors , Proteins/chemistry , Proteins/metabolism , Reproducibility of Results , Small Molecule Libraries , Structure-Activity Relationship
12.
Ann Hematol ; 98(12): 2683-2691, 2019 Dec.
Article En | MEDLINE | ID: mdl-31745600

In Germany, analyses of clinical and laboratory features of patients with acute porphyrias are only available for hereditary coproporphyria (HCP) but not with other acute porphyrias, acute intermittent porphyria (AIP) and variegate porphyria (VP). The aim of the study was to analyze a large cohort of patients with particular focus upon quality of life aspects. Sixty-two individuals from separate families with acute porphyrias (57 AIP, 5 VP) were included into an observational study collecting biochemical, genetic, and clinical data. A questionnaire was designed to complete anamnestic information and to assess the influence on quality of life. Most frequent signs and symptoms or laboratory abnormalities were abdominal colicky pain, red coloration of urine, and hyponatremia. Depression or anxiety was reported by 61% or 52% individuals, respectively. Fatigue was mentioned as the most quality of life-limiting symptom. In 59/61 patients, mutations could be identified. 44% (20/45) had to be admitted to an intensive care unit. Heme arginate was used in 64% (29/45) of patients for treatment of acute attacks at least once and in 33% for long-term treatment with high frequency of administration. Serum creatinine values increased in 47% (7/17) of the patients with recurrent attacks. Our analysis confirms a substantial influence of the diseases on the quality of life on patients. Percentages of urine discoloration and intensive care unit admissions were much higher than in other reports. Long-term treatment with heme arginate requires careful monitoring of iron status and renal values.


Arginine/administration & dosage , Family , Heme/administration & dosage , Hospitalization , Porphyria, Acute Intermittent , Quality of Life , Surveys and Questionnaires , Adult , Anxiety/drug therapy , Anxiety/genetics , Anxiety/metabolism , Anxiety/psychology , Depression/drug therapy , Depression/genetics , Depression/metabolism , Depression/psychology , Female , Germany , Humans , Male , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Porphyria, Acute Intermittent/psychology , Prospective Studies
13.
Hum Gene Ther ; 30(10): 1180-1189, 2019 10.
Article En | MEDLINE | ID: mdl-31179759

Exogenous delivery of messenger RNA (mRNA) is emerging as a new class of medicine with broad applicability including the potential to treat rare monogenic disorders. Recent advances in mRNA technology, including modifications to the mRNA itself along with improvements to the delivery vehicle, have transformed the utility of mRNA as a potential therapy to restore or replace different types of therapeutic proteins. Preclinical proof-of-concept has been demonstrated for mRNA therapy for three different rare metabolic disorders: methylmalonic acidemia, acute intermittent porphyria, and Fabry disease. Herein, we review those preclinical efficacy and safety studies in multiple animal models. For all three disorders, mRNA therapy restored functional protein to therapeutically relevant levels in target organs, led to sustained and reproducible pharmacology following each dose administration of mRNA, and was well tolerated as supported by liver function tests evaluated in animal models including nonhuman primates. These data provide compelling support for the clinical development of mRNA therapy as a treatment for various rare metabolic disorders.


Amino Acid Metabolism, Inborn Errors/therapy , Fabry Disease/therapy , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Porphyria, Acute Intermittent/therapy , RNA, Messenger/genetics , Rare Diseases/therapy , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Clinical Trials as Topic , Disease Models, Animal , Fabry Disease/genetics , Fabry Disease/metabolism , Gene Transfer Techniques , Genetic Diseases, Inborn/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Haplorhini , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Proof of Concept Study , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , Rare Diseases/genetics , Rare Diseases/metabolism , Rodentia
14.
PLoS One ; 13(12): e0208753, 2018.
Article En | MEDLINE | ID: mdl-30571766

Activation induced deaminase (AID) has two distinct and well defined roles, both relying on its deoxycytidine (dC) deaminating function: one as a DNA mutator and another in DNA demethylation. In chronic lymphocytic leukemia (CLL), AID was previously shown to be an independent negative prognostic factor. While there is substantial impact on DNA mutations, effects of AID on gene expression by promoter demethylation of disease related target genes in leukemia has not been addressed. To shed light on this question, we aimed at determining genome wide methylation changes as well as gene expression changes in response to AID expression in CLL. Although we found minor differences in individual methylation variable positions following AID expression, we could not find recurrent methylation changes of specific target sites or changes in global methylation.


Epigenesis, Genetic , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Porphyria, Acute Intermittent/metabolism , Computational Biology , DNA Methylation/physiology , Gene Expression Regulation, Neoplastic , Humans , Leukocytes, Mononuclear/enzymology
15.
Biochim Biophys Acta Gen Subj ; 1862(6): 1296-1305, 2018 06.
Article En | MEDLINE | ID: mdl-29476795

BACKGROUND: Acute Intermittent Porphyria (AIP) is an inherited disease produced by a deficiency of Porphobilinogen deaminase (PBG-D). The aim of this work was to evaluate the effects of Isoflurane and Sevoflurane on heme metabolism in a mouse genetic model of AIP to further support our previous proposal for avoiding their use in porphyric patients. A comparative study was performed administering the porphyrinogenic drugs allylisopropylacetamide (AIA), barbital and ethanol, and also between sex and mutation using AIP (PBG-D activity 70% reduced) and T1 (PBG-D activity 50% diminished) mice. METHODS: The activities of 5-Aminolevulinic synthetase (ALA-S), PBG-D, Heme oxygenase (HO) and CYP2E1; the expression of ALA-S and the levels of 5-aminolevulinic acid (ALA) were measured in different tissues of mice treated with the drugs mentioned. RESULTS: Isoflurane increased liver, kidney and brain ALA-S activity of AIP females but only affected kidney AIP males. Sevoflurane induced ALA-S activity in kidney and brain of female AIP group. PBG-D activity was further reduced by Isoflurane in liver male T1; in AIP male mice activity remained in its low basal levels. Ethanol and barbital also caused biochemical alterations. Only AIA triggered neurological signs similar to those observed during human acute attacks in male AIP being the symptoms less pronounced in females although ALA-S induction was greater. Heme degradation was affected. DISCUSSION: Biochemical alterations caused by the porphyrinogenic drugs assayed were different in male and female mice and also between T1 and AIP being more affected the females of AIP group. GENERAL SIGNIFICANCE: This is the first study using volatile anaesthetics in an AIP genetic model confirming Isoflurane and Sevoflurane porphyrinogenicity.


Anesthetics/pharmacology , Heme/metabolism , Hydroxymethylbilane Synthase/physiology , Models, Genetic , Porphobilinogen/pharmacology , Porphyria, Acute Intermittent/drug therapy , Volatile Organic Compounds/pharmacokinetics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Porphobilinogen/chemistry , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Porphyria, Acute Intermittent/pathology
16.
Clin Chim Acta ; 479: 1-6, 2018 Apr.
Article En | MEDLINE | ID: mdl-29317194

BACKGROUND: Acute intermittent porphyria (AIP) is caused by diminished activity of porphobilinogen deaminase (PBGD). The purpose of this study was to validate and compare two assays for PBGD activity. The clinical sensitivity of the PBGD activity assays in AIP diagnosis was also evaluated. METHODS: This study included 74 subjects from 18 Taiwanese families including symptomatic patients with AIP, asymptomatic carriers, and healthy family members. The specific mutations in AIP patients were identified by DNA sequencing. PBGD activity was measured in erythrocytes by quantifying formation of coproporphyrin or uroporphyrin by the enzyme using porphobilinogen (PBG) as a substrate and fluorimetry for detection. RESULTS: The calibration curves obtained with pure coproporphyrin or uroporphyrin were linear with correlation coefficients >0.99 in the range of 0-200nM for coproporphyrin and 0-150nM for uroporphyrin. The coefficients of variation for within-run and between-day imprecision were <9.8% for both assays. The three groups of subjects were used to establish the best cut-off of PBGD activity for identifying symptomatic AIP patients by using area under receiver operating characteristic curve analysis. The symptomatic AIP patients and asymptomatic carriers had significantly lower PBGD activity compared with the healthy family members (all p<.001). CONCLUSION: Two different PBGD activity assays were validated. The best cut-off for coproporphyrin was derived as 46.4nmol/h/mL RBC with corresponding sensitivity of 100% and specificity of 100% and the best cut-off for uroporphyrin was derived as 43.7nkat/L RBC with corresponding sensitivity of 100% and specificity of 97.4%.


Hydroxymethylbilane Synthase/metabolism , Porphyria, Acute Intermittent/diagnosis , Adolescent , Adult , Aged , Child , DNA/analysis , Female , Fluorometry , Hematocrit , Hemoglobins/analysis , Humans , Hydroxymethylbilane Synthase/blood , Hydroxymethylbilane Synthase/genetics , Male , Middle Aged , Molecular Structure , Mutation , Porphobilinogen/analysis , Porphobilinogen/metabolism , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/metabolism , Substrate Specificity , Taiwan , Young Adult
17.
Article En | MEDLINE | ID: mdl-28666226

BACKGROUND: Acute intermittent porphyria (AIP) is a rare metabolic disorder due to a deficiency of porphobilinogen deaminase, the third enzyme of the heme biosynthetic pathway. This low enzymatic activity may predispose to the appearance of acute neurological attacks. Seminal studies suggested that AIP was associated with changes in tryptophan homeostasis with inconclusive results. Therefore, the aim of this study was to analyze the urinary metabolome of AIP patients focusing on tryptophan metabolism using state-of-the-art technology. METHODS: This was a case-control study including a group of 25 AIP patients with active biochemical disease and increased excretion of heme-precursors and 25 healthy controls. Tryptophan and related compounds and metabolites including: large neutral amino acids (LNAAs), serotonin, kynurenine, kynurenic acid and anthranilic acid were quantified in urine by liquid chromatography tandem-mass spectrometry (LC-MS/MS). Twenty-nine biological markers (including metabolic ratios and absolute concentrations) were compared between patients and controls. RESULTS: Significant differences were found in the tryptophan-kynurenine metabolic pathway. Compared to controls, AIP patients showed: (a) increased urinary excretion of kynurenine and anthranilic acid (P<0.005); (b): elevation of the kynurenine/tryptophan ratio (P<0.001) and (c): decrease of the kynurenic acid/kynurenine ratio (P=0.001). In contrast, no differences were found in the serotonin metabolic pathway independently of the markers and ratios used. CONCLUSIONS: The results of the study demonstrate that there is an imbalance in the kynurenine metabolic pathway in AIP patients, with an increase of the kynurenine/tryptophan ratio in urine and a reduction of the kynurenic acid/kynurenine ratio. The modified ratios suggest induction of indoleamine 2,3-deoxygenase and decreased activity of kynurenine aminotransferase in the liver. The results confirm that LC-MS/MS is useful for the characterization of the urinary metabolome of hepatic porphyrias.


Metabolome/physiology , Metabolomics/methods , Porphyria, Acute Intermittent/urine , Tryptophan/metabolism , Tryptophan/urine , Adult , Biomarkers/metabolism , Biomarkers/urine , Cohort Studies , Female , Humans , Kynurenine/metabolism , Male , Middle Aged , Porphyria, Acute Intermittent/metabolism , Serotonin/metabolism , Young Adult
18.
Clin Exp Immunol ; 187(3): 466-479, 2017 03.
Article En | MEDLINE | ID: mdl-27859020

This study aimed to examine whether acute intermittent porphyria (AIP) is associated with systemic inflammation and whether the inflammation correlates with disease activity. A case-control study with 50 AIP cases and age-, sex- and place of residence-matched controls was performed. Plasma cytokines, insulin and C-peptide were analysed after an overnight fast using multiplex assay. Long pentraxin-3 (PTX3) and complement activation products (C3bc and TCC) were analysed using enzyme-linked immunosorbent assay (ELISA). Urine porphobilinogen ratio (U-PBG, µmol/mmol creatinine), haematological and biochemical tests were performed using routine methods. Questionnaires were used to register AIP symptoms, medication and other diseases. All 27 cytokines, chemokines and growth factors investigated were increased significantly in symptomatic AIP cases compared with controls (P < 0·0004). Hierarchical cluster analyses revealed a cluster with high visfatin levels and several highly expressed cytokines including interleukin (IL)-17, suggesting a T helper type 17 (Th17) inflammatory response in a group of AIP cases. C3bc (P = 0·002) and serum immunoglobulin (Ig)G levels (P = 0·03) were increased significantly in cases with AIP. The U-PBG ratio correlated positively with PTX3 (r = 0·38, P = 0·006), and with terminal complement complex (TCC) levels (r = 0·33, P = 0·02). PTX3 was a significant predictor of the biochemical disease activity marker U-PBG in AIP cases after adjustment for potential confounders in multiple linear regression analyses (P = 0·032). Prealbumin, C-peptide, insulin and kidney function were all decreased in the symptomatic AIP cases, but not in the asymptomatic cases. These results indicate that AIP is associated with systemic inflammation. Decreased C-peptide levels in symptomatic AIP cases indicate that reduced insulin release is associated with enhanced disease activity and reduced kidney function.


Inflammation/blood , Porphyria, Acute Intermittent/blood , Biomarkers/blood , C-Peptide/blood , Case-Control Studies , Cytokines/blood , Female , Humans , Immunoglobulin G/blood , Inflammation/immunology , Inflammation/metabolism , Insulin/blood , Kidney/immunology , Kidney/physiopathology , Male , Middle Aged , Porphyria, Acute Intermittent/immunology , Porphyria, Acute Intermittent/metabolism , Prealbumin/metabolism , T-Lymphocytes, Helper-Inducer/immunology
19.
Lakartidningen ; 1132016 09 09.
Article Sv | MEDLINE | ID: mdl-27622758

The attack of acute porphyria Based on in silico evidence of pharmacokinetic, pharmacodynamic, and physiologic properties, have approximately 1 300 medicinal drugs been assessed with regard to the specific risk to carriers of acute porphyria. The classifications have been published in booklet form, together with prophylactic advice to the carriers and suggestions for doctors in charge of their care. The risk-classifications rest on the behavior of the drug in an extended molecular model of the attack of acute porphyria. In this, symptoms are effects of 5-aminolevulinate (ALA) produced in surplus after acceleration of enzyme-deficient heme biosynthesis, taking place during induction of drug-metabolizing cytochromes P450 and triggered by hepatocellular nuclear receptors, activated by the drug. The process is enhanced by glucagon- and sirtuin-dependent molecular processes activated in stress and cellular energy deficit, and enhanced and prolonged by auto-generating ALA.


Drug-Related Side Effects and Adverse Reactions , Porphyria, Acute Intermittent , Algorithms , Humans , Liver/drug effects , Liver/metabolism , Porphyria, Acute Intermittent/chemically induced , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism
20.
Elife ; 52016 05 31.
Article En | MEDLINE | ID: mdl-27240733

Porphyrias are disorders of heme metabolism frequently characterized by extreme photosensitivity. This symptom results from accumulation of porphyrins, tetrapyrrole intermediates in heme biosynthesis that generate reactive oxygen species when exposed to light, in the skin of affected individuals. Here we report that in addition to producing an ommochrome body pigment, the planarian flatworm Schmidtea mediterranea generates porphyrins in its subepithelial pigment cells under physiological conditions, and that this leads to pigment cell loss when animals are exposed to intense visible light. Remarkably, porphyrin biosynthesis and light-induced depigmentation are enhanced by starvation, recapitulating a common feature of some porphyrias - decreased nutrient intake precipitates an acute manifestation of the disease. Our results establish planarians as an experimentally tractable animal model for research into the pathophysiology of acute porphyrias, and potentially for the identification of novel pharmacological interventions capable of alleviating porphyrin-mediated photosensitivity or decoupling dieting and fasting from disease pathogenesis.


Helminth Proteins/genetics , Pigments, Biological/genetics , Planarians/radiation effects , Porphyria, Acute Intermittent/physiopathology , Porphyrins/genetics , Animals , Disease Models, Animal , Gene Expression Regulation , Helminth Proteins/metabolism , Heme/genetics , Heme/metabolism , Humans , Light , Phenothiazines/metabolism , Pigments, Biological/antagonists & inhibitors , Pigments, Biological/biosynthesis , Planarians/genetics , Planarians/metabolism , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Porphyrins/antagonists & inhibitors , Porphyrins/biosynthesis , Skin Pigmentation/genetics , Skin Pigmentation/radiation effects , Starvation/genetics
...