Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Orphanet J Rare Dis ; 18(1): 384, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38066651

RESUMO

BACKGROUND: Acute hepatic porphyria (AHP) is a family of rare genetic diseases, including acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, and delta-aminolevulinic acid dehydratase-deficient porphyria. The objective of this retrospective cohort study was to provide information on the clinical features of AHP in Japan-including acute attacks, chronic symptoms, and long-term complications. METHODS: Patients with AHP between April 2008 and June 2020 were selected from Japan's Medical Data Vision claims database. Patients with AHP were matched 1:10, by sex and age, to patients without AHP. The outcomes were evaluated overall, for patients age ≥ 55 years, and for the matched population. RESULTS: A total of 391 patients with AHP were included from the Japanese Medical Data Vision database. During the observation period (April 2008-June 2020), 18.2% (71/391) of patients experienced 1 acute attack and 10.5% (41/391) experienced ≥ 2 attacks. Chronic symptoms with rates ~ 10% or higher in the AHP population compared with the matched population included neurotic, stress-related, and somatoform disorders (21.7% vs. 6.7% [15.0% difference]); sleep disorders (23.0% vs. 9.9% [13.1% difference]); other and unspecified abdominal pain (13.6% vs. 3.7% [9.9% difference]); and nausea and vomiting, excluding chemotherapy-induced emesis (17.9% vs. 8.1% [9.8% difference]). Long-term complications with higher incidence rates in the AHP population compared with the matched population included fibrosis and cirrhosis of liver (15.9% vs. 3.0% [12.9% difference]), polyneuropathies and other disorders of the peripheral nervous system (20.5% vs. 7.9% [12.6% difference]), liver cancer (16.9% vs. 4.7% [12.2% difference]), renal failure (16.4% vs. 4.3% [12.1% difference]), and hypertension (26.1% vs. 18.8% [7.3% difference]). Among AHP patients age ≥ 55 years, the most common long-term complications were hypertension, kidney failure, and liver cancer. CONCLUSIONS: In Japan, patients with AHP experience a high clinical burden in terms of acute attacks, chronic symptoms, and long-term complications. The clinical burden related to chronic symptoms and long-term complications was substantially higher in Japanese patients with AHP compared with a matched population without AHP. Recognizing these signs and symptoms of AHP may aid physicians in making an earlier diagnosis, which may help patients avoid attack triggers, implement disease management, and reduce lifetime disease burden.


Assuntos
Hipertensão , Neoplasias Hepáticas , Porfiria Aguda Intermitente , Porfirias Hepáticas , Humanos , Pessoa de Meia-Idade , Sintase do Porfobilinogênio , Japão/epidemiologia , Estudos Retrospectivos , Porfirias Hepáticas/complicações , Porfirias Hepáticas/epidemiologia , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/genética
2.
Arch Biochem Biophys ; 738: 109540, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36746260

RESUMO

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.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Porfiria Aguda Intermitente , Humanos , Ácido Aminolevulínico/metabolismo , Ácido Aminolevulínico/farmacologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Hepáticas/genética , Transcriptoma , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/metabolismo , Carcinogênese
3.
Gastroenterology ; 164(3): 484-491, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36642627

RESUMO

DESCRIPTION: The acute hepatic porphyrias (AHP) are rare, inborn errors of heme-metabolism and include acute intermittent porphyria, hereditary coproporphyria, variegate porphyria, and porphyria due to severe deficiency of 5-aminolevulinic acid dehydratase. Acute intermittent porphyria is the most common type of AHP, with an estimated prevalence of patients with symptoms of approximately 1 in 100,000. The major clinical presentation involves attacks of severe pain, usually abdominal and generalized, without peritoneal signs or abnormalities on cross-sectional imaging. Acute attacks occur mainly in women in their childbearing years. AHP should be considered in the evaluation of all patients, and especially women aged 15-50 years with recurrent severe abdominal pain not ascribable to common causes. The screening tests of choice include random urine porphobilinogen and δ-aminolevulinic acid corrected to creatinine. All patients with elevations in urinary porphobilinogen and/or δ-aminolevulinic acid should initially be presumed to have AHP. The cornerstones of management include discontinuation of porphyrinogenic drugs and chemicals, administration of oral or intravenous dextrose and intravenous hemin, and use of analgesics and antiemetics. Diagnosis of AHP type can be confirmed after initial treatment by genetic testing for pathogenic variants in HMBS, CPOX, PPOX, and ALAD genes. AHP is also associated with chronic symptoms and long-term risk of systemic arterial hypertension, chronic renal and liver disease, and hepatocellular carcinoma. Patients who have recurrent acute attacks (4 or more per year) should be considered for prophylactic therapy with intravenous hemin or subcutaneous givosiran. Liver transplantation is curative and reserved for patients with intractable symptoms who have failed other treatment options. METHODS: This expert review was commissioned and approved by the American Gastroenterological Association (AGA) Institute Clinical Practice Updates Committee (CPUC) and the AGA Governing Board to provide timely guidance on a topic of high clinical importance to the AGA membership, and underwent internal peer review by the CPUC and external peer review through standard procedures of Gastroenterology. These Best Practice Advice (BPA) statements were drawn from a review of the published literature and from expert opinion. Because systematic reviews were not performed, these BPA statements do not carry formal ratings of the quality of evidence or strength of the presented considerations. Best Practice Advice Statements BEST PRACTICE ADVICE 1: Women aged 15-50 years with unexplained, recurrent severe abdominal pain without a clear etiology after an initial workup should be considered for screening for an AHP. BEST PRACTICE ADVICE 2: Initial diagnosis of AHP should be made by biochemical testing measuring δ-aminolevulinic acid, porphobilinogen, and creatinine on a random urine sample. BEST PRACTICE ADVICE 3: Genetic testing should be used to confirm the diagnosis of AHP in patients with positive biochemical testing. BEST PRACTICE ADVICE 4: Acute attacks of AHP that are severe enough to require hospital admission should be treated with intravenous hemin, given daily, preferably into a high-flow central vein. BEST PRACTICE ADVICE 5: In addition to intravenous hemin, management of acute attacks of AHP should include pain control, antiemetics, management of systemic arterial hypertension, tachycardia, and hyponatremia, and hypomagnesemia, if present. BEST PRACTICE ADVICE 6: Patients should be counseled to avoid identifiable triggers that may precipitate acute attacks, such as alcohol and porphyrinogenic medications. BEST PRACTICE ADVICE 7: Prophylactic heme therapy or givosiran, administered in an outpatient setting, should be considered in patients with recurrent attacks (4 or more per year). BEST PRACTICE ADVICE 8: Liver transplantation for AHP should be limited to patients with intractable symptoms and significantly decreased quality of life who are refractory to pharmacotherapy. BEST PRACTICE ADVICE 9: Patients with AHP should be monitored annually for liver disease. BEST PRACTICE ADVICE 10: Patients with AHP, regardless of the severity of symptoms, should undergo surveillance for hepatocellular carcinoma, beginning at age 50 years, with liver ultrasound every 6 months. BEST PRACTICE ADVICE 11: Patients with AHP on treatment should undergo surveillance for chronic kidney disease annually with serum creatinine and estimated glomerular filtration rate. BEST PRACTICE ADVICE 12: Patients should be counseled on the chronic and long-term complications of AHP, including neuropathy, chronic kidney disease, hypertension, and hepatocellular carcinoma, and need for long-term monitoring.


Assuntos
Antieméticos , Carcinoma Hepatocelular , Hipertensão , Neoplasias Hepáticas , Porfiria Aguda Intermitente , Porfirias Hepáticas , Insuficiência Renal Crônica , Humanos , Feminino , Estados Unidos , Pessoa de Meia-Idade , Porfiria Aguda Intermitente/diagnóstico , Porfiria Aguda Intermitente/genética , Sintase do Porfobilinogênio , Porfobilinogênio/urina , Hemina , Ácido Aminolevulínico/urina , Creatinina , Qualidade de Vida , Heme , Dor Abdominal
4.
J Cancer Res Clin Oncol ; 149(6): 2647-2655, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36245063

RESUMO

INTRODUCTION: Acute intermittent porphyria (AIP) is a very rare (orphan) metabolic disorder of porphyrin biosynthesis which is characterized by elevated plasma and urine levels of 5-aminolevulinic acid (5-ALA) and porphobilinogen (PBG). Patients with this disorder which is caused by a germline mutation of the hydroxymethylbilan-synthase (HMBS)-gene have a high risk of primary liver cancer which may be determined by disease activity. The exact mechanism of carcinogenesis of this rare tumor is unknown, however. MATERIALS AND METHODS: We analyzed paraffin-embedded formalin-fixed liver tumor and normal liver specimens of two female AIP patients treated at the Munich EPNET center. One patient had developed hepatocellular carcinoma (HCC), the other intrahepatic cholangiocarcinoma (CCA). Since biallelic inactivation of HMBS had been observed in one study, we used Sanger and next-generation sequencing with a 8 gene porphyria panel plus 6 potential modifier loci to search for mutations in DNA extractions. RESULTS: In the patient with the HCC, we found a second inactivating mutation in the HMBS gene in the tumor but not in the adjacent normal liver tissue. No mutation could be found in the liver tissues of the patient with CCA, however. CONCLUSIONS: Biallelic inactivation of HMBS or protoporphyrinogen-oxidase (PPOX), another enzyme of porphyrin biosynthesis, has been observed in patients with acute porphyrias and liver tumors. We could confirm this in our patient with HCC with a mutation in HMBS but not in the one with CCA. Since 5-ALA can be converted into carcinogenic substances such as 4,5-dioxovaleric acid (DOVA) or 3,6-dihydropyrazine-2,5-dipropanoic acid (= cyclic dimerization product of 5-ALA), local production of these metabolites in hepatic areas with complete loss of HMBS activity may contribute to liver carcinogenesis.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Porfiria Aguda Intermitente , Porfirinas , Feminino , Humanos , Ácido Aminolevulínico/urina , Carcinogênese , Carcinoma Hepatocelular/genética , Flavoproteínas , Neoplasias Hepáticas/genética , Proteínas Mitocondriais , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/patologia , Protoporfirinogênio Oxidase/genética , Adulto
5.
FEBS Open Bio ; 12(12): 2136-2146, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36115019

RESUMO

Hydroxymethylbilane synthase (HMBS) is the third enzyme involved in haem biosynthesis, in which it catalyses the formation of tetrapyrrole 1-hydroxymethylbilane (HMB). In this process, HMBS binds four consecutive substrate molecules, creating the enzyme-intermediate complexes ES, ES2 , ES3 and ES4 . Pathogenic variants in the HMBS gene are associated with the dominantly inherited disorder acute intermittent porphyria. In this study, we have characterised the p.R26H variant to shed light on the role of Arg26 in the elongation mechanism of HMBS and to provide insights into its effect on the enzyme. With selected biophysical methods, we have been able to show that p.R26H forms a single enzyme-intermediate complex in the ES2 -state. We were also able to demonstrate that the p.R26H variant results in an inactive enzyme, which is unable to produce the HMB product.


Assuntos
Hidroximetilbilano Sintase , Porfiria Aguda Intermitente , Humanos , Hidroximetilbilano Sintase/genética , Hidroximetilbilano Sintase/química , Hidroximetilbilano Sintase/metabolismo , Porfiria Aguda Intermitente/genética
6.
J Investig Med High Impact Case Rep ; 10: 23247096221109206, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35762500

RESUMO

Acute intermittent porphyria (AIP) is a rare autosomal dominant inherited disease, predominantly seen in female patients, caused by mutations in the hydroxymethylbilane synthase gene. When impaired, elevated heme biosynthesis precursor levels accumulate in the liver, resulting in neurological symptoms, psychiatric disturbances, darkened urine color, abdominal pain, nausea, vomiting, and ileus. We present a 22-year-old Hispanic female with diffuse abdominal pain and no bowel movements for 8 days. She reported recent antibiotic and oral contraceptive pill use. Computerized tomography of her abdomen revealed a dilated small bowel and marked colonic distension. A colonoscopy found mild nonspecific inflammation in the rectosigmoid and terminal ileum. Her abdominal pain persisted despite interventions and improvements in appetite, bowel movements, abdominal imaging, and treatment of an identified Clostridium difficile infection. A random urine porphobilinogen was then obtained and found to be elevated. Fractionation of plasma and urine porphyrins was suggestive of AIP. Her symptoms improved with 3 days of intravenous (IV) hematin and IV dextrose. This is a unique case of a rare disease due to her clinical presentation with ileus, unremarkable past medical history, family history, and the prehospitalization and intrahospitalization factors that likely exacerbated the patient AIP.


Assuntos
Íleus , Porfiria Aguda Intermitente , Dor Abdominal/etiologia , Feminino , Humanos , Hidroximetilbilano Sintase/genética , Mutação , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/diagnóstico , Porfiria Aguda Intermitente/genética , Adulto Jovem
7.
J Hepatol ; 77(4): 1038-1046, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35636578

RESUMO

BACKGROUND & AIMS: Acute intermittent porphyria (AIP), caused by heterozygous germline mutations of the heme synthesis pathway enzyme HMBS (hydroxymethylbilane synthase), confers a high risk of hepatocellular carcinoma (HCC) development. Yet, the role of HMBS in liver tumorigenesis remains unclear. METHODS: Herein, we explore HMBS alterations in a large series of 758 HCC cases, including 4 patients with AIP. We quantify the impact of HMBS mutations on heme biosynthesis pathway intermediates and we investigate the molecular and clinical features of HMBS-mutated tumors. RESULTS: We identify recurrent bi-allelic HMBS inactivation, both in patients with AIP acquiring a second somatic HMBS mutation and in sporadic HCC with 2 somatic hits. HMBS alterations are enriched in truncating mutations, in particular in splice regions, leading to abnormal transcript structures. Bi-allelic HMBS inactivation results in a massive accumulation of its toxic substrate porphobilinogen and synergizes with CTNNB1-activating mutations, leading to the development of well-differentiated tumors with a transcriptomic signature of Wnt/ß-catenin pathway activation and a DNA methylation signature related to ageing. HMBS-inactivated HCC mostly affects females, in the absence of fibrosis and classical HCC risk factors. CONCLUSIONS: These data identify HMBS as a tumor suppressor gene whose bi-allelic inactivation defines a homogenous clinical and molecular HCC subtype. LAY SUMMARY: Heme (the precursor to hemoglobin, which plays a key role in oxygen transport around the body) synthesis occurs in the liver and involves several enzymes including hydroxymethylbilane synthase (HMBS). HMBS mutations cause acute intermittent porphyria, a disease caused by the accumulation of toxic porphyrin precursors. Herein, we show that HMBS inactivation is also involved in the development of liver cancers with distinct clinical and molecular characteristics.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Porfiria Aguda Intermitente , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/genética , Feminino , Heme , Humanos , Hidroximetilbilano Sintase/genética , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/genética , Mutação , Oxigênio , Porfobilinogênio , Porfiria Aguda Intermitente/etiologia , Porfiria Aguda Intermitente/genética , beta Catenina/genética
8.
J Intern Med ; 291(6): 824-836, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35112415

RESUMO

BACKGROUND: The acute hepatic porphyrias (AHP) are associated with a risk of primary liver cancer (PLC), but risk estimates are unclear, and what AHP characteristics that predict PLC risk are unknown. In this register-based, matched cohort study, we assessed the PLC risk in relation to biochemical and clinical porphyria severity, genotype, age, and sex. METHODS: All patients in the Swedish porphyria register with acute intermittent porphyria (AIP), variegate porphyria (VP), or hereditary coproporphyria (HCP) during 1987-2015 were included. This AHP cohort was compared with age-, sex-, and county-matched reference individuals from the general population. National register-based hospital admissions for AHP were used to indicate the clinical severity. For AIP, the most common AHP type, patients were stratified by genotype and urinary porphobilinogen (U-PBG). Incident PLC data were collected from national health registers. RESULTS: We identified 1244 individuals with AHP (1063 [85%] AIP). During a median follow-up of 19.5 years, we identified 108 incident PLC cases, including 83 AHP patients (6.7%) and 25 of 12,333 reference individuals (0.2%). The adjusted hazard ratio for AHP-PLC was 38.0 (95% confidence interval: 24.3-59.3). Previously elevated U-PBG and hospitalizations for porphyria, but not AIP genotype or sex, were associated with increased PLC risk. Patients aged >50 years with previously elevated U-PBG (n = 157) had an annual PLC incidence of 1.8%. CONCLUSION: This study confirmed a high PLC risk and identified a strong association with clinical and biochemical AIP activity. Regular PLC surveillance is motivated in patients older than 50 years with a history of active AIP.


Assuntos
Neoplasias Hepáticas , Porfiria Aguda Intermitente , Porfirias Hepáticas , Porfirias , Estudos de Coortes , Humanos , Neoplasias Hepáticas/epidemiologia , Sintase do Porfobilinogênio/deficiência , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/epidemiologia , Porfiria Aguda Intermitente/genética , Porfirias/genética , Porfirias Hepáticas/complicações , Porfirias Hepáticas/epidemiologia
9.
Ann Hematol ; 100(7): 1685-1693, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34050373

RESUMO

Givosiran is a novel approach to treat patients with acute intermittent porphyrias (AIP) by silencing of ∂-ALA-synthase 1, the first enzyme of heme biosynthesis in the liver. We included two patients in the Envision study who responded clinically well to this treatment. However, in both patients, therapy had to be discontinued because of severe adverse effects: One patient (A) developed local injection reactions which continued to spread all over her body with increasing number of injections and eventually caused a severe systemic allergic reaction. Patient B was hospitalized because of a fulminant pancreatitis. Searching for possible causes, we also measured the patients plasma homocysteine (Hcy) levels in fluoride-containing collection tubes: by LC-MS/MS unexpectedly, plasma Hcy levels were 100 and 200 in patient A and between 100 and 400 µmol/l in patient B. Searching for germline mutations in 10 genes that are relevant for homocysteine metabolism only revealed hetero- and homozygous polymorphisms in the MTHFR gene. Alternatively, an acquired inhibition of cystathionine-beta-synthase which is important for homocysteine metabolism could explain the plasma homocysteine increase. This enzyme is heme-dependent: when we gave heme arginate to our patients, Hcy levels rapidly dropped. Hence, we conclude that inhibition of ∂-ALA-synthase 1 by givosiran causes a drop of free heme in the hepatocyte and therefore the excessive increase of plasma homocysteine. Hyperhomocysteinemia may contribute to the adverse effects seen in givosiran-treated patients which may be due to protein-N-homocysteinylation.


Assuntos
5-Aminolevulinato Sintetase/antagonistas & inibidores , Acetilgalactosamina/análogos & derivados , Heme/deficiência , Hiper-Homocisteinemia/etiologia , Porfiria Aguda Intermitente/tratamento farmacológico , Pirrolidinas/uso terapêutico , Acetilgalactosamina/efeitos adversos , Acetilgalactosamina/uso terapêutico , Adulto , Arginina/uso terapêutico , Colite/etiologia , Colo Sigmoide/patologia , Ensaios Clínicos Controlados como Assunto , Hipersensibilidade a Drogas/etiologia , Feminino , Fibrose , Heme/análise , Heme/uso terapêutico , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Homocisteína/metabolismo , Humanos , Hidroximetilbilano Sintase/sangue , Hidroximetilbilano Sintase/genética , Masculino , Modelos Biológicos , Pancreatite/etiologia , Porfiria Aguda Intermitente/sangue , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/genética , Pirrolidinas/efeitos adversos
10.
J Inherit Metab Dis ; 44(4): 961-971, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33861472

RESUMO

Acute intermittent porphyria (AIP) is a rare metabolic disease caused by mutations within the hydroxymethylbilane synthase gene. Previous studies have reported increased levels of plasma total homocysteine (tHcy) in symptomatic AIP patients. In this study, we present long-term data for tHcy and related parameters for an AIP patient cohort (n = 37) in different clinical disease-states. In total, 25 patients (68%) presented with hyperhomocysteinemia (HHcy; tHcy > 15 µmol/L) during the observation period. HHcy was more frequent in AIP patients with recurrent disease receiving heme arginate, than in nonrecurrent (median tHcy: 21.6 µmol/L; range: 10-129 vs median tHcy: 14.5 µmol/L; range 6-77). Long-term serial analyses showed a high within-person tHcy variation, especially among the recurrent patients (coefficient of variation: 16.4%-78.8%). HHcy was frequently associated with low blood concentrations of pyridoxal-5'-phosphate and folate, while cobalamin concentration and the allele distribution of the methylene-tetrahydrofolate-reductase gene were normal. Strikingly, 6 out of the 9 recurrent patients who were later included in a regime of givosiran, a small-interfering RNA that effectively reduced recurrent attacks, showed further increased tHcy (median tHcy in 9 patients: 105 µmol/L; range 16-212). Screening of amino acids in plasma by liquid-chromatography showed co-increased levels of methionine (median 71 µmol/L; range 23-616; normal <40), suggestive of acquired deficiency of cystathionine-ß-synthase. The kynunerine/tryptophan ratio in plasma was, however, normal, indicating a regular metabolism of tryptophan by heme-dependent enzymes. In conclusion, even if HHcy was observed in AIP patients receiving heme arginate, givosiran induced an aggravation of the dysregulation, causing a co-increase of tHcy and methionine resembling classic homocystinuria.


Assuntos
Acetilgalactosamina/análogos & derivados , Arginina/deficiência , Heme/deficiência , Hiper-Homocisteinemia/etiologia , Porfiria Aguda Intermitente/tratamento farmacológico , Pirrolidinas/uso terapêutico , Acetilgalactosamina/efeitos adversos , Acetilgalactosamina/uso terapêutico , Adulto , Arginina/uso terapêutico , Cistationina beta-Sintase/genética , Feminino , Ácido Fólico/sangue , Heme/uso terapêutico , Homeostase , Homocisteína/metabolismo , Homocistinúria/complicações , Humanos , Hidroximetilbilano Sintase/sangue , Hidroximetilbilano Sintase/genética , Masculino , Metionina/sangue , Pessoa de Meia-Idade , Porfiria Aguda Intermitente/sangue , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/genética , Fosfato de Piridoxal/sangue , Pirrolidinas/efeitos adversos , Adulto Jovem
11.
Mol Genet Metab ; 131(4): 418-423, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33199206

RESUMO

BACKGROUND: 5-Aminolevulinic acid dehydratase (ALAD) porphyria (ADP) is an ultrarare autosomal recessive disease, with only eight documented cases, all of whom were males. Although classified as an acute hepatic porphyria (AHP), induction of the rate limiting hepatic enzyme 5-aminolevulinic acid synthase-1 (ALAS1) has not been demonstrated, and the marrow may also contribute excess 5-aminolevulinic acid (ALA). Two patients have died and reported follow up for the others is limited, so the natural history of this disease is poorly understood and treatment experience limited. METHODS: We report new molecular findings and update the clinical course and treatment of the sixth reported ADP patient, now 31 years old and the only known case in the Americas, and review published data regarding genotype-phenotype correlation and treatment. RESULTS: Circulating hepatic 5-aminolevulinic acid synthase-1 (ALAS1) mRNA was elevated in this case, as in other AHPs. Gain of function mutation of erythroid specific ALAS2 - an X-linked modifying gene in some other porphyrias - was not found. Seven reported ADP cases had compound heterozygous ALAD mutations resulting in very low residual ALAD activity and symptoms early in life or adolescence. One adult with a germline ALAD mutant allele developed ADP in association with a clonal myeloproliferative disorder, polycythemia vera. CONCLUSIONS: Elevation in circulating hepatic ALAS1 and response to treatment with hemin indicate that the liver is an important source of excess ALA in ADP, although the marrow may also contribute. Intravenous hemin was effective in most reported cases for treatment and prevention of acute attacks of neurological symptoms.


Assuntos
5-Aminolevulinato Sintetase/genética , Sintase do Porfobilinogênio/deficiência , Sintase do Porfobilinogênio/genética , Porfiria Aguda Intermitente/genética , Porfirias Hepáticas/genética , 5-Aminolevulinato Sintetase/sangue , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Heme/genética , Hemina/administração & dosagem , Humanos , Lactente , Recém-Nascido , Fígado/metabolismo , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Mutação/genética , Porfobilinogênio/metabolismo , Sintase do Porfobilinogênio/sangue , Porfiria Aguda Intermitente/sangue , Porfiria Aguda Intermitente/tratamento farmacológico , Porfiria Aguda Intermitente/patologia , Porfirias Hepáticas/sangue , Porfirias Hepáticas/tratamento farmacológico , Porfirias Hepáticas/patologia , RNA Mensageiro/sangue , Adulto Jovem
12.
Hum Gene Ther ; 30(10): 1180-1189, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31179759

RESUMO

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.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/terapia , Doença de Fabry/terapia , Doenças Genéticas Inatas/terapia , Terapia Genética/métodos , Porfiria Aguda Intermitente/terapia , RNA Mensageiro/genética , Doenças Raras/terapia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Doença de Fabry/genética , Doença de Fabry/metabolismo , Técnicas de Transferência de Genes , Doenças Genéticas Inatas/metabolismo , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Haplorrinos , Humanos , Nanopartículas/administração & dosagem , Nanopartículas/química , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/metabolismo , Estudo de Prova de Conceito , RNA Mensageiro/administração & dosagem , RNA Mensageiro/metabolismo , Doenças Raras/genética , Doenças Raras/metabolismo , Roedores
13.
Am J Med Genet B Neuropsychiatr Genet ; 180(1): 46-54, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30556376

RESUMO

Many genetic conditions can mimic mental health disorders, with psychiatric symptoms that are difficult to treat with standard psychotropic medications. This study tests the hypothesis that psychiatric populations are enriched for pathogenic variants associated with selected inborn errors of metabolism (IEMs). Using next-generation sequencing, 2046 psychiatric patients were screened for pathogenic variants in genes associated with four IEMs, Niemann-Pick disease type C (NPC), Wilson disease (WD), homocystinuria (HOM), and acute intermittent porphyria (AIP). Among the 2046 cases, carrier rates of 0.83, 0.98, and 0.20%, for NPC, WD and HOM, and affected rates of 0.10 and 0.24% for NPC and AIP were seen, respectively. An enrichment of known and predicted pathogenic variants in the genes associated with NPC and AIP was found in the psychiatric cohort and especially in schizophrenia patients. The results of this study support that pathogenic variants in genes associated with IEMs are enriched in psychiatric populations. Underlying undiagnosed IEMs could account for the psychiatric symptomatology in a subset of psychiatric patients. Further studies are warranted to investigate the possibility that carriers for IEMs may have an increased risk for psychiatric disorders, particularly in the context of poor treatment response.


Assuntos
Transtornos Mentais/genética , Transtornos Mentais/metabolismo , Erros Inatos do Metabolismo/genética , Adulto , Transtorno Bipolar/genética , Transtorno Bipolar/metabolismo , Estudos de Coortes , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/metabolismo , Feminino , Variação Genética/genética , Degeneração Hepatolenticular/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Homocistinúria/genética , Humanos , Masculino , Transtornos Mentais/fisiopatologia , Erros Inatos do Metabolismo/complicações , Erros Inatos do Metabolismo/metabolismo , Pessoa de Meia-Idade , Doença de Niemann-Pick Tipo C/genética , Porfiria Aguda Intermitente/genética , Esquizofrenia/genética , Esquizofrenia/metabolismo
14.
Proc Natl Acad Sci U S A ; 115(17): E4071-E4080, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29632172

RESUMO

Hydroxymethylbilane synthase (HMBS), the third enzyme in the heme biosynthetic pathway, catalyzes the head-to-tail condensation of four molecules of porphobilinogen (PBG) to form the linear tetrapyrrole 1-hydroxymethylbilane (HMB). Mutations in human HMBS (hHMBS) cause acute intermittent porphyria (AIP), an autosomal-dominant disorder characterized by life-threatening neurovisceral attacks. Although the 3D structure of hHMBS has been reported, the mechanism of the stepwise polymerization of four PBG molecules to form HMB remains unknown. Moreover, the specific roles of each of the critical active-site residues in the stepwise enzymatic mechanism and the dynamic behavior of hHMBS during catalysis have not been investigated. Here, we report atomistic studies of HMB stepwise synthesis by using molecular dynamics (MD) simulations, mutagenesis, and in vitro expression analyses. These studies revealed that the hHMBS active-site loop movement and cofactor turn created space for the elongating pyrrole chain. Twenty-seven residues around the active site and water molecules interacted to stabilize the large, negatively charged, elongating polypyrrole. Mutagenesis of these active-site residues altered the binding site, hindered cofactor binding, decreased catalysis, impaired ligand exit, and/or destabilized the enzyme. Based on intermediate stages of chain elongation, R26 and R167 were the strongest candidates for proton transfer to deaminate the incoming PBG molecules. Unbiased random acceleration MD simulations identified R167 as a gatekeeper and facilitator of HMB egress through the space between the enzyme's domains and the active-site loop. These studies identified the specific active-site residues involved in each step of pyrrole elongation, thereby providing the molecular bases of the active-site mutations causing AIP.


Assuntos
Hidroximetilbilano Sintase/química , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto , Porfiria Aguda Intermitente/enzimologia , Pirróis/química , Substituição de Aminoácidos , Humanos , Hidroximetilbilano Sintase/genética , Hidroximetilbilano Sintase/metabolismo , Porfiria Aguda Intermitente/genética , Estrutura Secundária de Proteína , Pirróis/metabolismo
15.
Hum Gene Ther ; 29(4): 480-491, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28990424

RESUMO

Porphobilinogen deaminase (PBGD) gene therapy represents a promising therapeutic option for acute intermittent porphyria (AIP) patients suffering recurrent acute attacks. A first-in-human Phase I clinical trial confirmed the safety and tolerability of adeno-associated virus (AAV)-AAT-PBGD gene therapy, but higher doses and/or more efficient vectors are needed to achieve therapeutic expression of the transgene. This study assayed the insertion into the promoter of a short enhancer element able to induce transgene expression during exposure to endogenous and exogenous stimuli related to the pathology of the disease. The inclusion in tandem of two elements of the minimal functional sequence of human δ-aminolevulinic acid synthase drug-responsive enhancing sequence (ADRES) positioned upstream of the promoter strongly induced transgene expression in the presence of estrogens, starvation, and certain drugs known to trigger attacks in porphyria patients. The inclusion of two ADRES motives in an AAV vector improved therapeutic efficacy, reducing 10-fold the effective dose in AIP mice. In conclusion, the inclusion of specific enhancer elements in the promoter of gene therapy vectors for AIP was able to overexpress the therapeutic transgene when it is most needed, at the time when porphyrinogenic factors increase the demand for hepatic heme and precipitate acute porphyria attacks.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Porfiria Aguda Intermitente/terapia , 5-Aminolevulinato Sintetase/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Vetores Genéticos/genética , Humanos , Hidroximetilbilano Sintase/genética , Fígado/metabolismo , Fígado/patologia , Camundongos , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/patologia , Regiões Promotoras Genéticas
16.
Hum Mol Genet ; 24(17): 5015-23, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26071363

RESUMO

Acute intermittent porphyria (AIP), an autosomal dominant metabolic disease (MIM #176000), is due to a deficiency of hydroxymethylbilane synthase (HMBS), which catalyzes the third step of the heme biosynthetic pathway. The clinical expression of the disease is mainly neurological, involving the autonomous, central and peripheral nervous systems. We explored mitochondrial oxidative phosphorylation (OXPHOS) in the brain and skeletal muscle of the Hmbs(-/-) mouse model first in the basal state (BS), and then after induction of the disease with phenobarbital and treatment with heme arginate (HA). The modification of the respiratory parameters, determined in mice in the BS, reflected a spontaneous metabolic energetic adaptation to HMBS deficiency. Phenobarbital induced a sharp alteration of the oxidative metabolism with a significant decrease of ATP production in skeletal muscle that was restored by treatment with HA. This OXPHOS defect was due to deficiencies in complexes I and II in the skeletal muscle whereas all four respiratory chain complexes were affected in the brain. To date, the pathogenesis of AIP has been mainly attributed to the neurotoxicity of aminolevulinic acid and heme deficiency. Our results show that mitochondrial energetic failure also plays an important role in the expression of the disease.


Assuntos
Encéfalo/metabolismo , Hidroximetilbilano Sintase/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Músculos/metabolismo , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/metabolismo , Trifosfato de Adenosina/biossíntese , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Músculos/efeitos dos fármacos , Fenobarbital/farmacologia
17.
J Hepatol ; 62(3): 734-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25445397

RESUMO

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


Assuntos
Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/genética , Flavoproteínas/genética , Hidroximetilbilano Sintase/genética , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/genética , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Mutação , Porfiria Aguda Intermitente/complicações , Porfiria Aguda Intermitente/genética , Porfiria Variegada/complicações , Porfiria Variegada/genética , Protoporfirinogênio Oxidase/genética , Idoso , Idoso de 80 Anos ou mais , Carcinoma Hepatocelular/enzimologia , Feminino , Mutação em Linhagem Germinativa , Humanos , Neoplasias Hepáticas/enzimologia , Porfiria Aguda Intermitente/enzimologia , Porfiria Variegada/enzimologia , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
18.
Hum Gene Ther ; 24(12): 1007-17, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24070415

RESUMO

Acute intermittent porphyria (AIP) results from haplo-insufficient activity of porphobilinogen deaminase (PBGD) and is characterized clinically by life-threatening, acute neurovisceral attacks. To date, liver transplantation is the only curative option for AIP. The aim of the present preclinical nonhuman primate study was to determine the safety and transduction efficacy of an adeno-associated viral vector encoding PBGD (recombinant AAV serotype 5-codon-optimized human porphobilinogen deaminase, rAAV5-cohPBGD) administered intravenously as part of a safety program to start a clinical study in patients with AIP. Macaques injected with either 1 × 10(13) or 5 × 10(13) vector genomes/kg of clinical-grade rAAV5-cohPBGD were monitored by standardized clinical parameters, and vector shedding was analyzed. Liver transduction efficacy, biodistribution, vector integration, and histopathology at day 30 postvector administration were determined. There was no evidence of acute toxicity, and no adverse effects were observed. The vector achieved efficient and homogenous hepatocellular transduction, reaching transgenic PBGD expression levels equivalent to 50% of the naturally expressed PBGD mRNA. No cellular immune response was detected against the human PBGD or AAV capsid proteins. Integration site analysis in transduced liver cells revealed an almost random integration pattern supporting the good safety profile of rAAV5-cohPBGD. Together, data obtained in nonhuman primates indicate that rAAV5-cohPBGD represents a safe therapy to correct the metabolic defect present in AIP patients.


Assuntos
Terapia Genética , Haploinsuficiência/genética , Hidroximetilbilano Sintase/genética , Porfiria Aguda Intermitente/terapia , Animais , Dependovirus , Vetores Genéticos , Hepatócitos/metabolismo , Humanos , Hidroximetilbilano Sintase/uso terapêutico , Macaca , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/patologia , Distribuição Tecidual/genética , Transdução Genética
19.
Hum Mol Genet ; 22(14): 2929-40, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23562909

RESUMO

Acute intermittent porphyria (AIP) is a hepatic metabolic disease that results from haplo-insufficient activity of porphobilinogen deaminase (PBGD). The dominant clinical feature is acute intermittent attacks when hepatic heme synthesis is activated by endocrine or exogenous factors. Gene therapy vectors over-expressing PBGD protein in the liver offers potential as a cure for AIP. Here, we developed a helper-dependent adenovirus (HDA) encoding human PBGD (hPBGD) and assessed its therapeutic efficacy in a murine model of AIP. Intravenous or intrahepatic administration of HDA-hPBGD to AIP mice resulted in a sustained hepatic hPBGD expression in a dose-dependent manner. Intrahepatic administration conveyed full protection against induced porphyria attacks at a significantly lower viral dose than intravenous injection. Transgenic hPBGD accumulated only in the cytosol of hepatocytes as the endogenous protein. Characterization of PBGD-deficient mouse strains revealed that a strong PBGD deficiency causes the chronic disturbance of cytosolic and endoplasmic reticulum folding machineries. This disturbance was completely restored over time by the over-expression of hPBGD. HDA-hPBGD is a promising vector that protects against porphyria attacks and resolves the chronic folding stress associated with low levels of PBGD activity.


Assuntos
Adenoviridae/genética , Terapia Genética , Hidroximetilbilano Sintase/genética , Porfiria Aguda Intermitente/genética , Porfiria Aguda Intermitente/terapia , Adenoviridae/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Hepatócitos/enzimologia , Hepatócitos/virologia , Humanos , Hidroximetilbilano Sintase/metabolismo , Fígado/enzimologia , Fígado/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Porfiria Aguda Intermitente/enzimologia , Porfiria Aguda Intermitente/prevenção & controle , Dobramento de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA