Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Inherit Metab Dis ; 47(1): 80-92, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37401651

RESUMO

Phenylketonuria (PKU) or hyperphenylalaninemia is considered a paradigm for an inherited (metabolic) liver defect and is, based on murine models that replicate all human pathology, an exemplar model for experimental studies on liver gene therapy. Variants in the PAH gene that lead to hyperphenylalaninemia are never fatal (although devastating if untreated), newborn screening has been available for two generations, and dietary treatment has been considered for a long time as therapeutic and satisfactory. However, significant shortcomings of contemporary dietary treatment of PKU remain. A long list of various gene therapeutic experimental approaches using the classical model for human PKU, the homozygous enu2/2 mouse, witnesses the value of this model to develop treatment for a genetic liver defect. The list of experiments for proof of principle includes recombinant viral (AdV, AAV, and LV) and non-viral (naked DNA or LNP-mRNA) vector delivery methods, combined with gene addition, genome, gene or base editing, and gene insertion or replacement. In addition, a list of current and planned clinical trials for PKU gene therapy is included. This review summarizes, compares, and evaluates the various approaches for the sake of scientific understanding and efficacy testing that may eventually pave the way for safe and efficient human application.


Assuntos
Fenilalanina Hidroxilase , Fenilcetonúrias , Humanos , Camundongos , Animais , Fenilalanina Hidroxilase/genética , Fenilcetonúrias/genética , Fenilcetonúrias/terapia , Terapia Genética/métodos , Fígado/patologia , DNA
2.
Mol Genet Metab ; 137(1-2): 114-126, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36027720

RESUMO

BACKGROUND: Early treated patients with phenylketonuria (PKU) often become lost to follow-up from adolescence onwards due to the historical focus of PKU care on the pediatric population and lack of programs facilitating the transition to adulthood. As a result, evidence on the management of adolescents and young adults with PKU is limited. METHODS: Two meetings were held with a multidisciplinary international panel of 25 experts in PKU and comorbidities frequently experienced by patients with PKU. Based on the outcomes of the first meeting, a set of statements were developed. During the second meeting, these statements were voted on for consensus generation (≥70% agreement), using a modified Delphi approach. RESULTS: A total of 37 consensus recommendations were developed across five areas that were deemed important in the management of adolescents and young adults with PKU: (1) general physical health, (2) mental health and neurocognitive functioning, (3) blood Phe target range, (4) PKU-specific challenges, and (5) transition to adult care. The consensus recommendations reflect the personal opinions and experiences from the participating experts supported with evidence when available. Overall, clinicians managing adolescents and young adults with PKU should be aware of the wide variety of PKU-associated comorbidities, initiating screening at an early age. In addition, management of adolescents/young adults should be a joint effort between the patient, clinical center, and parents/caregivers supporting adolescents with gradually gaining independent control of their disease during the transition to adulthood. CONCLUSIONS: A multidisciplinary international group of experts used a modified Delphi approach to develop a set of consensus recommendations with the aim of providing guidance and offering tools to clinics to aid with supporting adolescents and young adults with PKU.


Assuntos
Fenilcetonúrias , Criança , Adolescente , Adulto Jovem , Humanos , Adulto , Consenso , Fenilcetonúrias/diagnóstico , Programas de Rastreamento
3.
Mol Genet Metab ; 137(1-2): 9-17, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35868243

RESUMO

BACKGROUND: Tyrosinemia type 1 (HT1) is a rare metabolic disorder caused by a defect in the tyrosine catabolic pathway. Since HT1 patients are treated with NTBC, outcome improved and life expectancy greatly increased. However extensive neurocognitive and behavioural problems have been described, which might be related to treatment with NTBC, the biochemical changes induced by NTBC, or metabolites accumulating due to the enzymatic defect characterizing the disease. OBJECTIVE: To study the possible pathophysiological mechanisms of brain dysfunction in HT1, we assessed blood and brain LNAA, and brain monoamine neurotransmitter metabolite levels in relation to behavioural and cognitive performance of HT1 mice. DESIGN: C57BL/6 littermates were divided in three different experimental groups: HT1, heterozygous and wild-type mice (n = 10; 5 male). All groups were treated with NTBC and underwent cognitive and behavioural testing. One week after behavioural testing, blood and brain material were collected to measure amino acid profiles and brain monoaminergic neurotransmitter levels. RESULTS: Irrespective of the genetic background, NTBC treatment resulted in a clear increase in brain tyrosine levels, whereas all other brain LNAA levels tended to be lower than their reference values. Despite these changes in blood and brain biochemistry, no significant differences in brain monoamine neurotransmitter (metabolites) were found and all mice showed normal behaviour and learning and memory. CONCLUSION: Despite the biochemical changes, NTBC and genotype of the mice were not associated with poorer behavioural and cognitive function of the mice. Further research involving dietary treatment of FAH-/- are warranted to investigate whether this reveals the cognitive impairments that have been seen in treated HT1 patients.


Assuntos
Nitrobenzoatos , Tirosinemias , Animais , Camundongos , Masculino , Cicloexanonas , Camundongos Endogâmicos C57BL , Tirosinemias/tratamento farmacológico , Tirosinemias/genética , Tirosina/metabolismo
4.
Mol Genet Metab ; 131(3): 306-315, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33051130

RESUMO

Phenylalanine hydroxylase (PAH) deficiency, colloquially known as phenylketonuria (PKU), is among the most common inborn errors of metabolism and in the past decade has become a target for the development of novel therapeutics such as gene therapy. PAH deficient mouse models have been key to new treatment development, but all prior existing models natively express liver PAH polypeptide as inactive or partially active PAH monomers, which complicates the experimental assessment of protein expression following therapeutic gene, mRNA, protein, or cell transfer. The mutant PAH monomers are able to form hetero-tetramers with and inhibit the overall holoenzyme activity of wild type PAH monomers produced from a therapeutic vector. Preclinical therapeutic studies would benefit from a PKU model that completely lacks both PAH activity and protein expression in liver. In this study, we employed CRISPR/Cas9-mediated gene editing in fertilized mouse embryos to generate a novel mouse model that lacks exon 1 of the Pah gene. Mice that are homozygous for the Pah exon 1 deletion are viable, severely hyperphenylalaninemic, accurately replicate phenotypic features of untreated human classical PKU and lack any detectable liver PAH activity or protein. This model of classical PKU is ideal for further development of gene and cell biologics to treat PKU.


Assuntos
Fígado/metabolismo , Fenilalanina Hidroxilase/genética , Fenilalanina/genética , Fenilcetonúrias/terapia , Animais , Sistemas CRISPR-Cas/genética , Modelos Animais de Doenças , Éxons/genética , Edição de Genes , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos , Fenilalanina/metabolismo , Fenilalanina Hidroxilase/farmacologia , Fenilcetonúrias/genética , Fenilcetonúrias/patologia
5.
Mol Genet Metab ; 131(1-2): 90-97, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32928639

RESUMO

BACKGROUND: The plasma acylcarnitine profile is frequently used as a biochemical assessment for follow-up in diagnosed patients with fatty acid oxidation disorders (FAODs). Disease specific acylcarnitine species are elevated during metabolic decompensation but there is clinical and biochemical heterogeneity among patients and limited data on the utility of an acylcarnitine profile for routine clinical monitoring. METHODS: We evaluated plasma acylcarnitine profiles from 30 diagnosed patients with long-chain FAODs (carnitine palmitoyltransferase-2 (CPT2), very long-chain acyl-CoA dehydrogenase (VLCAD), and long-chain 3-hydroxy acyl-CoA dehydrogenase or mitochondrial trifunctional protein (LCHAD/TFP) deficiencies) collected after an overnight fast, after feeding a controlled low-fat diet, and before and after moderate exercise. Our purpose was to describe the variability in this biomarker and how various physiologic states effect the acylcarnitine concentrations in circulation. RESULTS: Disease specific acylcarnitine species were higher after an overnight fast and decreased by approximately 60% two hours after a controlled breakfast meal. Moderate-intensity exercise increased the acylcarnitine species but it varied by diagnosis. When analyzed for a genotype/phenotype correlation, the presence of the common LCHADD mutation (c.1528G > C) was associated with higher levels of 3-hydroxyacylcarnitines than in patients with other mutations. CONCLUSIONS: We found that feeding consistently suppressed and that moderate intensity exercise increased disease specific acylcarnitine species, but the response to exercise was highly variable across subjects and diagnoses. The clinical utility of routine plasma acylcarnitine analysis for outpatient treatment monitoring remains questionable; however, if acylcarnitine profiles are measured in the clinical setting, standardized procedures are required for sample collection to be of value.


Assuntos
Cardiomiopatias/sangue , Carnitina O-Palmitoiltransferase/deficiência , Carnitina/análogos & derivados , Síndrome Congênita de Insuficiência da Medula Óssea/sangue , Erros Inatos do Metabolismo Lipídico/sangue , Erros Inatos do Metabolismo/sangue , Doenças Mitocondriais/sangue , Miopatias Mitocondriais/sangue , Proteína Mitocondrial Trifuncional/deficiência , Doenças Musculares/sangue , Doenças do Sistema Nervoso/sangue , Rabdomiólise/sangue , 3-Hidroxiacil-CoA Desidrogenases/genética , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Aciltransferase/genética , Acetil-CoA C-Aciltransferase/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/sangue , Isomerases de Ligação Dupla Carbono-Carbono/genética , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Cardiomiopatias/dietoterapia , Cardiomiopatias/patologia , Cardiomiopatias/terapia , Carnitina/sangue , Carnitina/genética , Carnitina/metabolismo , Carnitina O-Palmitoiltransferase/sangue , Síndrome Congênita de Insuficiência da Medula Óssea/dietoterapia , Síndrome Congênita de Insuficiência da Medula Óssea/patologia , Síndrome Congênita de Insuficiência da Medula Óssea/terapia , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Terapia por Exercício , Jejum , Feminino , Humanos , Erros Inatos do Metabolismo Lipídico/dietoterapia , Erros Inatos do Metabolismo Lipídico/patologia , Erros Inatos do Metabolismo Lipídico/terapia , 3-Hidroxiacil-CoA Desidrogenase de Cadeia Longa/sangue , Masculino , Erros Inatos do Metabolismo/dietoterapia , Erros Inatos do Metabolismo/patologia , Erros Inatos do Metabolismo/terapia , Doenças Mitocondriais/dietoterapia , Doenças Mitocondriais/patologia , Doenças Mitocondriais/terapia , Miopatias Mitocondriais/dietoterapia , Miopatias Mitocondriais/patologia , Miopatias Mitocondriais/terapia , Proteína Mitocondrial Trifuncional/sangue , Doenças Musculares/dietoterapia , Doenças Musculares/patologia , Doenças Musculares/terapia , Doenças do Sistema Nervoso/dietoterapia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/terapia , Racemases e Epimerases/genética , Racemases e Epimerases/metabolismo , Rabdomiólise/dietoterapia , Rabdomiólise/patologia , Rabdomiólise/terapia
6.
J Inherit Metab Dis ; 43(6): 1232-1242, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33448436

RESUMO

Anaplerotic odd-chain fatty acid supplementation has been suggested as an approach to replenish citric acid cycle intermediate (CACi) pools and facilitate adenosine triphosphate (ATP) production in subjects with long-chain fatty acid oxidation disorders, but the evidence that cellular CACi depletion exists and that repletion occurs following anaplerotic substrate supplementation is limited. We exercised very long-chain acyl-CoA dehydrogenase-deficient (VLCAD-/-) and wild-type (WT) mice to exhaustion and collected cardiac tissue for measurement of CACi by targeted metabolomics. In a second experimental group, VLCAD-/- and WT mice that had been fed chow prepared with either medium-chain triglyceride (MCT) oil or triheptanoin for 4 weeks were exercised for 60 minutes. VLCAD-/- mice exhibited lower succinate in cardiac muscle at exhaustion than WT mice suggesting lower CACi in VLCAD-/- with prolonged exercise. In mice fed either MCT or triheptanoin, succinate and malate were greater in VLCAD-/- mice fed triheptanoin compared to VLCAD-/- animals fed MCT but lower than WT mice fed triheptanoin. Long-chain odd acylcarnitines such as C19 were elevated in VLCAD-/- and WT mice fed triheptanoin suggesting some elongation of the heptanoate, but it is unknown what proportion of heptanoate was oxidized vs elongated. Prolonged exercise was associated with decreased cardiac muscle succinate in VLCAD-/- mice in comparison to WT mice. VLCAD-/- fed triheptanoin had increased succinate compared to VLCAD-/- mice fed MCT but lower than WT mice fed triheptanoin. Cardiac CACi were higher following dietary ingestion of an anaplerotic substrate, triheptanoin, in comparison to MCT.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Síndrome Congênita de Insuficiência da Medula Óssea/dietoterapia , Síndrome Congênita de Insuficiência da Medula Óssea/metabolismo , Erros Inatos do Metabolismo Lipídico/dietoterapia , Erros Inatos do Metabolismo Lipídico/metabolismo , Doenças Mitocondriais/dietoterapia , Doenças Mitocondriais/metabolismo , Doenças Musculares/dietoterapia , Doenças Musculares/metabolismo , Triglicerídeos/administração & dosagem , Acil-CoA Desidrogenase de Cadeia Longa/genética , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Animais , Ciclo do Ácido Cítrico , Síndrome Congênita de Insuficiência da Medula Óssea/genética , Gorduras na Dieta/administração & dosagem , Ácidos Graxos/metabolismo , Feminino , Erros Inatos do Metabolismo Lipídico/genética , Fígado/metabolismo , Masculino , Camundongos , Doenças Mitocondriais/genética , Doenças Musculares/genética , Miocárdio/metabolismo , Oxirredução , Triglicerídeos/química
7.
Paediatr Drugs ; 21(6): 413-426, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31667718

RESUMO

Tyrosinemia type 1 (TT1) is a rare metabolic disease caused by a defect in tyrosine catabolism. TT1 is clinically characterized by acute liver failure, development of hepatocellular carcinoma, renal and neurological problems, and consequently an extremely poor outcome. This review showed that the introduction of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) in 1992 has revolutionized the outcome of TT1 patients, especially when started pre-clinically. If started early, NTBC can prevent liver failure, renal problems, and neurological attacks and decrease the risk for hepatocellular carcinoma. NTBC has been shown to be safe and well tolerated, although the long-term effectiveness of treatment with NTBC needs to be awaited. The high tyrosine concentrations caused by treatment with NTBC could result in ophthalmological and skin problems and requires life-long dietary restriction of tyrosine and its precursor phenylalanine, which could be strenuous to adhere to. In addition, neurocognitive problems have been reported since the introduction of NTBC, with hypothesized but as yet unproven pathophysiological mechanisms. Further research should be done to investigate the possible relationship between important clinical outcomes and blood concentrations of biochemical parameters such as phenylalanine, tyrosine, succinylacetone, and NTBC, and to develop clear guidelines for treatment and follow-up with reliable measurements. This all in order to ultimately improve the combined NTBC and dietary treatment and limit possible complications such as hepatocellular carcinoma development, neurocognitive problems, and impaired quality of life.


Assuntos
Cicloexanonas/efeitos adversos , Nitrobenzoatos/efeitos adversos , Tirosinemias/tratamento farmacológico , Cicloexanonas/uso terapêutico , Humanos , Nitrobenzoatos/uso terapêutico , Tirosina/sangue , Tirosinemias/complicações , Tirosinemias/dietoterapia
8.
Hum Gene Ther ; 30(10): 1274-1283, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31364419

RESUMO

Phenylketonuria (PKU) is considered to be a paradigm for a monogenic metabolic disorder but was never thought to be a primary application for human gene therapy due to established alternative treatment. However, somewhat unanticipated improvement in neuropsychiatric outcome upon long-term treatment of adults with PKU with enzyme substitution therapy might slowly change this assumption. In parallel, PKU was for a long time considered to be an excellent test system for experimental gene therapy of a Mendelian autosomal recessive defect of the liver due to an outstanding mouse model and the easy to analyze and well-defined therapeutic end point, that is, blood l-phenylalanine concentration. Lifelong treatment by targeting the mouse liver (or skeletal muscle) was achieved using different approaches, including (1) recombinant adeno-associated viral (rAAV) or nonviral naked DNA vector-based gene addition, (2) genome editing using base editors delivered by rAAV vectors, and (3) by delivering rAAVs for promoter-less insertion of the PAH-cDNA into the Pah locus. In this article we summarize the gene therapeutic attempts of correcting a mouse model for PKU and discuss the future implications for human gene therapy.


Assuntos
Dependovirus/genética , Edição de Genes/métodos , Terapia Genética/métodos , Fenilalanina Hidroxilase/genética , Fenilcetonúrias/terapia , Animais , Biomarcadores/sangue , Ensaios Clínicos como Assunto , Dependovirus/metabolismo , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Fígado/enzimologia , Fígado/patologia , Camundongos , Fenilalanina/sangue , Fenilalanina Hidroxilase/deficiência , Fenilcetonúrias/enzimologia , Fenilcetonúrias/genética , Fenilcetonúrias/patologia , Plasmídeos/química , Plasmídeos/metabolismo
9.
Cell Transplant ; 28(1): 79-88, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30477316

RESUMO

Orthotopic liver transplantation remains the only curative therapy for inborn errors of metabolism. Given the tremendous success for primary immunodeficiencies using ex-vivo gene therapy with lentiviral vectors, there is great interest in developing similar curative therapies for metabolic liver diseases. We have previously generated a pig model of hereditary tyrosinemia type 1 (HT1), an autosomal recessive disorder caused by deficiency of fumarylacetoacetate hydrolase (FAH). Using this model, we have demonstrated curative ex-vivo gene and cell therapy using a lentiviral vector to express FAH in autologous hepatocytes. To further evaluate the long-term clinical outcomes of this therapeutic approach, we continued to monitor one of these pigs over the course of three years. The animal continued to thrive off the protective drug NTBC, gaining weight appropriately, and maintaining sexual fecundity for the course of his life. The animal was euthanized 31 months after transplantation to perform a thorough biochemical and histological analysis. Biochemically, liver enzymes and alpha-fetoprotein levels remained normal and abhorrent metabolites specific to HT1 remained corrected. Liver histology showed no evidence of tumorigenicity and Masson's trichrome staining revealed minimal fibrosis and no evidence of cirrhosis. FAH-immunohistochemistry revealed complete repopulation of the liver by transplanted FAH-positive cells. A complete histopathological report on other organs, including kidney, revealed no abnormalities. This study is the first to demonstrate long-term safety and efficacy of hepatocyte-directed gene therapy in a large animal model. We conclude that hepatocyte-directed ex-vivo gene therapy is a rational choice for further exploration as an alternative therapeutic approach to whole organ transplantation for metabolic liver disease, including HT1.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Genética/métodos , Hidrolases/metabolismo , Tirosinemias/enzimologia , Tirosinemias/terapia , Animais , Biologia Computacional , Modelos Animais de Doenças , Hidrolases/genética , Masculino , Suínos , Tirosinemias/metabolismo
10.
Sci Transl Med ; 8(349): 349ra99, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27464750

RESUMO

We tested the hypothesis that ex vivo hepatocyte gene therapy can correct the metabolic disorder in fumarylacetoacetate hydrolase-deficient (Fah(-/-)) pigs, a large animal model of hereditary tyrosinemia type 1 (HT1). Recipient Fah(-/-) pigs underwent partial liver resection and hepatocyte isolation by collagenase digestion. Hepatocytes were transduced with one or both of the lentiviral vectors expressing the therapeutic Fah and the reporter sodium-iodide symporter (Nis) genes under control of the thyroxine-binding globulin promoter. Pigs received autologous transplants of hepatocytes by portal vein infusion. After transplantation, the protective drug 2-(2-nitro-4-trifluoromethylbenzyol)-1,3 cyclohexanedione (NTBC) was withheld from recipient pigs to provide a selective advantage for expansion of corrected FAH(+) cells. Proliferation of transplanted cells, assessed by both immunohistochemistry and noninvasive positron emission tomography imaging of NIS-labeled cells, demonstrated near-complete liver repopulation by gene-corrected cells. Tyrosine and succinylacetone levels improved to within normal range, demonstrating complete correction of tyrosine metabolism. In addition, repopulation of the Fah(-/-) liver with transplanted cells inhibited the onset of severe fibrosis, a characteristic of nontransplanted Fah(-/-) pigs. This study demonstrates correction of disease in a pig model of metabolic liver disease by ex vivo gene therapy. To date, ex vivo gene therapy has only been successful in small animal models. We conclude that further exploration of ex vivo hepatocyte genetic correction is warranted for clinical use.


Assuntos
Terapia Genética/métodos , Fígado/metabolismo , Tirosinemias/metabolismo , Tirosinemias/terapia , Animais , Cicloexanonas/farmacologia , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hidrolases/genética , Hidrolases/metabolismo , Imuno-Histoquímica , Nitrobenzoatos/farmacologia , Suínos , Transplante Homólogo , Tirosinemias/enzimologia , Tirosinemias/genética
11.
Ophthalmic Genet ; 37(4): 404-414, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26979128

RESUMO

BACKGROUND: Cobalamin C disease (cblC), which leads to methylmalonic acidemia with homocystinuria, is the most common inherited disorder of vitamin B12 metabolism. Reported ocular findings associated with cblC have been maculopathy, pigmentary retinopathy, and optic nerve atrophy. Cobalamin A disease (cblA) which causes an isolated methylmalonic acidemia without homocystinuria is rarer than cblC. This is the first detailed report of the ocular findings associated with cblA. We also describe the spectrum of ocular findings in our cblC patients. MATERIALS AND METHODS: A case series describing the ophthalmologic clinical course of six patients with a diagnosis of cobalamin C type and one patient with cobalamin A type of methylmalonic acidemia. Patients were diagnosed through biochemical laboratory testing and genetic analysis was conducted on most patients. Longitudinal fundus findings, optical coherence tomography (OCT), autofluorescence, and electrophysiology were followed in the patients. RESULTS: The cblA patient demonstrated a relatively mild ocular phenotype with late-onset and slowly progressing temporal disc pallor and peripapillary atrophy in the second decade of life. The patient maintained good visual acuity and central vision, without evidence of maculopathy. The six cblC patients demonstrated a range of ocular findings from unremarkable and mild phenotypes to significant retinopathy, including bull's eye maculopathy, severe maculopathy with punched out chorioretinal atrophy, peripheral bone spicules, and optic nerve atrophy. CONCLUSIONS: The spectrum of ocular manifestations seen with inherited disorders of cobalamin metabolism is wide, ranging from mild optic nerve atrophy to severe macular or retinal degeneration. This heterogeneity may in part reflect the associated biochemical phenotype, such as that observed between our cblA and cblC patients. We also observed heterogeneity within the cblC type in agreement with previous reports.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Homocistinúria/diagnóstico , Doenças do Nervo Óptico/diagnóstico , Degeneração Retiniana/diagnóstico , Transtornos da Visão/diagnóstico , Deficiência de Vitamina B 12/congênito , Erros Inatos do Metabolismo dos Aminoácidos/fisiopatologia , Eletrorretinografia , Feminino , Seguimentos , Fumaratos/sangue , Homocisteína/sangue , Homocistinúria/fisiopatologia , Humanos , Lactente , Recém-Nascido , Masculino , Maleatos/sangue , Doenças do Nervo Óptico/fisiopatologia , Imagem Óptica , Degeneração Retiniana/fisiopatologia , Tomografia de Coerência Óptica , Transtornos da Visão/fisiopatologia , Acuidade Visual/fisiologia , Deficiência de Vitamina B 12/diagnóstico , Deficiência de Vitamina B 12/fisiopatologia
12.
Nutr Res ; 36(1): 101-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26773786

RESUMO

A higher incidence of osteopenia is observed among children with inherited metabolic disorders (inborn errors of metabolism, or IEMs) who consume medical food-based diets that restrict natural vitamin D-containing food sources. We evaluated the vitamin D status of children with IEMs who live in the Pacific Northwest with limited sun exposure and determined whether bone mineral density (BMD) in children with phenylketonuria (PKU), the most common IEM, correlated with diet or biochemical markers of bone metabolism. We hypothesized that children with IEMs would have lower serum vitamin D concentrations than controls and that some children with PKU would have reduced bone mineralization. A retrospective record review of 88 patients with IEMs, and 445 children on unrestricted diets (controls) found the 25-hydroxyvitamin D concentrations were normal and not significantly different between groups (IEM patients, 27.1 ± 10.9; controls, 27.6 ± 11.2). Normal BMD at the hip or spine (-2

Assuntos
25-Hidroxivitamina D 2/sangue , Densidade Óssea , Doenças do Desenvolvimento Ósseo/prevenção & controle , Calcifediol/sangue , Alimentos Formulados , Erros Inatos do Metabolismo/dietoterapia , Deficiência de Vitamina D/prevenção & controle , Centros Médicos Acadêmicos , Adolescente , Adulto , Biomarcadores/sangue , Doenças do Desenvolvimento Ósseo/epidemiologia , Doenças do Desenvolvimento Ósseo/etiologia , Criança , Estudos de Coortes , Estudos Transversais , Registros Eletrônicos de Saúde , Alimentos Formulados/efeitos adversos , Humanos , Incidência , Erros Inatos do Metabolismo/sangue , Erros Inatos do Metabolismo/fisiopatologia , Oregon/epidemiologia , Fenilcetonúrias/sangue , Fenilcetonúrias/dietoterapia , Fenilcetonúrias/fisiopatologia , Estudos Retrospectivos , Risco , Deficiência de Vitamina D/epidemiologia , Deficiência de Vitamina D/etiologia , Adulto Jovem
13.
Mol Genet Metab ; 116(1-2): 29-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26296711

RESUMO

BACKGROUND: Health care outcomes have been increasingly assessed through health-related quality of life (HRQoL) measures. While the introduction of nitrogen-scavenging medications has improved survival in patients with urea cycle disorders (UCDs), they are often associated with side effects that may affect patient compliance and outcomes. METHODS: Symptoms commonly associated with nitrogen-scavenging medications were evaluated in 100 adult and pediatric participants using a non-validated UCD-specific questionnaire. Patients or their caregivers responded to a pre-defined list of symptoms known to be associated with the use of these medications. Responses were collected at baseline (while patients were receiving sodium phenylbutyrate [NaPBA]) and during treatment with glycerol phenylbutyrate (GPB). RESULTS: After 3 months of GPB dosing, there were significant reductions in the proportion of patients with treatment-associated symptoms (69% vs. 46%; p<0.0001), the number of symptoms per patient (2.5 vs. 1.1; p<0.0001), and frequency of the more commonly reported individual symptoms such as body odor, abdominal pain, nausea, burning sensation in mouth, vomiting, and heartburn (p<0.05). The reduction in symptoms was observed in both pediatric and adult patients. The presence or absence of symptoms or change in severity did not correlate with plasma ammonia levels or NaPBA dose. CONCLUSIONS: The reduction in symptoms following 3 months of open-label GPB dosing was similar in pediatric and adult patients and may be related to chemical structure and intrinsic characteristics of the product rather than its effect on ammonia control.


Assuntos
Glicerol/análogos & derivados , Fenilbutiratos/efeitos adversos , Qualidade de Vida , Autorrelato , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Adolescente , Adulto , Idoso , Amônia/sangue , Antineoplásicos/uso terapêutico , Criança , Pré-Escolar , Feminino , Glicerol/efeitos adversos , Glicerol/química , Glicerol/uso terapêutico , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Fenilbutiratos/química , Fenilbutiratos/uso terapêutico , Inquéritos e Questionários , Distúrbios Congênitos do Ciclo da Ureia/sangue , Distúrbios Congênitos do Ciclo da Ureia/psicologia , Adulto Jovem
14.
Mol Ther Nucleic Acids ; 3: e193, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25226162

RESUMO

We have previously demonstrated the efficacy of antisense therapy for splicing defects in cellular models of metabolic diseases, suppressing the use of cryptic splice sites or pseudoexon insertions. To date, no animal models with these defects are available. Here, we propose exon skipping of the phenylalanine hydroxylase (Pah) gene expressed in liver and kidney to generate systemic hyperphenylalaninemia in mice as a sensitive in vivo assay to test splice suppression. Systemic elevation of blood L-Phe can be quantified using tandem MS/MS. Exon 11 and/or 12 skipping for the normal PAH gene was validated in hepatoma cells for comparing two oligonucleotide chemistries, morpholinos and locked nucleic acids. Subsequently, Vivo-morpholinos (VMO) were tested in wild-type and in phenotypically normal Pah(enu2/+) heterozygous mice to target exon 11 and/or 12 of the murine Pah gene using different VMO dosing, mode of injection and treatment regimes. Consecutive intravenous injections of VMO resulted in transient hyperphenylalaninemia correlating with complete exon skipping and absence of PAH protein and enzyme activity. Sustained effect required repeated injection of VMOs. Our results provide not only a sensitive in vivo assay to test for splice-modulating antisense oligonucleotides, but also a simple method to generate murine models for genetic liver diseases.

15.
Stem Cell Res ; 13(1): 144-53, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24879068

RESUMO

Hereditary tyrosinemia type I (HT1) is caused by deficiency in fumarylacetoacetate hydrolase (FAH), an enzyme that catalyzes the last step of tyrosine metabolism. The most severe form of the disease presents acutely during infancy, and is characterized by severe liver involvement, most commonly resulting in death if untreated. Generation of FAH(+/-) pigs was previously accomplished by adeno-associated virus-mediated gene knockout in fibroblasts and somatic cell nuclear transfer. Subsequently, these animals were outbred and crossed to produce the first FAH(-/-) pigs. FAH-deficiency produced a lethal defect in utero that was corrected by administration of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3 cyclohexanedione (NTBC) throughout pregnancy. Animals on NTBC were phenotypically normal at birth; however, the animals were euthanized approximately four weeks after withdrawal of NTBC due to clinical decline and physical examination findings of severe liver injury and encephalopathy consistent with acute liver failure. Biochemical and histological analyses, characterized by diffuse and severe hepatocellular damage, confirmed the diagnosis of severe liver injury. FAH(-/-) pigs provide the first genetically engineered large animal model of a metabolic liver disorder. Future applications of FAH(-/-) pigs include discovery research as a large animal model of HT1 and spontaneous acute liver failure, and preclinical testing of the efficacy of liver cell therapies, including transplantation of hepatocytes, liver stem cells, and pluripotent stem cell-derived hepatocytes.


Assuntos
Hidrolases/deficiência , Hepatopatias/enzimologia , Tirosinemias/enzimologia , Animais , Modelos Animais de Doenças , Feminino , Técnicas de Inativação de Genes , Genótipo , Hepatopatias/metabolismo , Masculino , Gravidez , Suínos
16.
Mol Genet Metab ; 111(4): 484-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24503138

RESUMO

Very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a fatty acid oxidation disorder with widely varying presentations that has presented a significant challenge to newborn screening (NBS). The Western States Regional Genetics Services Collaborative developed a workgroup to study infants with NBS positive for VLCADD. We performed retrospective analysis of newborns with elevated C14:1-acylcarnitine on NBS in California, Oregon, Washington, and Hawai'i including available confirmatory testing and clinical information. Overall, from 2,802,504 children screened, there were 242 cases screen-positive for VLCADD. There were 34 symptomatic true positive cases, 18 asymptomatic true positives, 112 false positives, 55 heterozygotes, 11 lost to follow-up, and 12 other disorders. One in 11,581 newborns had an abnormal NBS for suspected VLCADD. Comparison of analytes and analyte ratios from the NBS demonstrated statistically significant differences between true positive and false positive groups for C14:1, C14, C14:1/C2, and C14:1/C16. The positive predictive value for all true positive cases was 94%, 54%, and 23% when C14:1 was ≥2.0 µM, ≥1.0 µM, and ≥0.7 µM, respectively. Sequential post-analytical analysis could reduce the referral rate in 25.8% of cases. This study is the largest reported follow-up of infants with NBS screen-positive results for suspected VLCADD and demonstrates the necessity of developing comprehensive and consistent long-term follow-up NBS systems. Application of clinical information revealed differences between symptomatic and asymptomatic children with VLCADD. Comparison of NBS analytes and analyte ratios may be valuable in developing more effective diagnostic algorithms.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Erros Inatos do Metabolismo Lipídico/diagnóstico , Doenças Mitocondriais/diagnóstico , Doenças Musculares/diagnóstico , Triagem Neonatal/métodos , Carnitina/análogos & derivados , Carnitina/metabolismo , Síndrome Congênita de Insuficiência da Medula Óssea , Análise Mutacional de DNA , Demografia , Ácidos Graxos/metabolismo , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo , Reprodutibilidade dos Testes
17.
J Inherit Metab Dis ; 33(6): 681-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20495959

RESUMO

Problems with long-term dietary compliance in phenylketonuria (PKU) necessitate the development of alternative treatment approaches. Therapeutic liver repopulation with phenylalanine hydroxylase (PAH)-expressing cells following hepatocyte or haematopoietic stem cell transplantation has been investigated as a possible novel treatment approach for PKU. Successful therapeutic liver repopulation requires both a stimulus for liver regeneration at the time of cell transplantation and a selective growth advantage for the PAH+ donor cells. Unfortunately, wild-type PAH+ hepatocytes do not enjoy any growth advantage over PAH- cells. Successful correction of hyperphenylalaninemia following therapeutic liver repopulation has been accomplished only in an animal model that yields a selective advantage for the donor cells. Haematopoietic stem cell (HSC)-mediated therapeutic liver repopulation has not been reported in any hyperphenylalaninemic system, and the success of HSC-mediated liver repopulation for PKU may be limited by the slow kinetics of this approach. If therapeutic liver repopulation is to be employed successfully in humans with PKU, an effective method of providing a selective growth advantage for the donor cells must be developed. If this can be achieved, liver repopulation with 10-20% wild-type hepatocytes will likely completely normalize Phe clearance in individuals with PKU.


Assuntos
Regeneração Hepática/fisiologia , Transplante de Fígado/métodos , Fenilcetonúrias/terapia , Medicina Regenerativa/métodos , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Mamíferos
18.
Blood ; 115(16): 3374-81, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20177050

RESUMO

Hereditary hemochromatosis is caused by mutations in the hereditary hemochromatosis protein (HFE), transferrin-receptor 2 (TfR2), hemojuvelin, hepcidin, or ferroportin genes. Hepcidin is a key iron regulator, which is secreted by the liver, and decreases serum iron levels by causing the down-regulation of the iron transporter, ferroportin. Mutations in either HFE or TfR2 lower hepcidin levels, implying that both HFE and TfR2 are necessary for regulation of hepcidin expression. In this study, we used a recombinant adeno-associated virus, AAV2/8, for hepatocyte-specific expression of either Hfe or Tfr2 in mice. Expression of Hfe in Hfe-null mice both increased Hfe and hepcidin mRNA and lowered hepatic iron and Tf saturation. Expression of Tfr2 in Tfr2-deficient mice had a similar effect, whereas expression of Hfe in Tfr2-deficient mice or of Tfr2 in Hfe-null mice had no effect on liver or serum iron levels. Expression of Hfe in wild-type mice increased hepcidin mRNA and lowered iron levels. In contrast, expression of Tfr2 had no effect on wild-type mice. These findings suggest that Hfe is limiting in formation of the Hfe/Tfr2 complex that regulates hepcidin expression. In addition, these studies show that the use of recombinant AAV vector to deliver genes is a promising approach for studying physiologic consequences of protein complexes.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/genética , Proteína 2 de Ligação a Repetições Teloméricas/genética , Adenoviridae/genética , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Vetores Genéticos , Proteína da Hemocromatose , Hepcidinas , Antígenos de Histocompatibilidade Classe I/metabolismo , Immunoblotting , Ferro/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
19.
Hum Gene Ther ; 21(4): 463-77, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19916803

RESUMO

Phenylketonuria (PKU) is caused by hepatic phenylalanine hydroxylase (PAH) deficiency and is associated with systemic accumulation of phenylalanine (Phe). Previously we demonstrated correction of murine PKU after intravenous injection of a recombinant type 2 adeno-associated viral vector pseudotyped with type 8 capsid (rAAV2/8), which successfully directed hepatic transduction and Pah gene expression. Here, we report that liver PAH activity and phenylalanine clearance were also restored in PAH-deficient mice after simple intramuscular injection of either AAV2 pseudotype 1 (rAAV2/1) or rAAV2/8 vectors. Serotype 2 AAV vector (rAAV2/2) was also investigated, but long-term phenylalanine clearance has been observed only for pseudotypes 1 and 8. Therapeutic correction was shown in both male and female mice, albeit more effectively in males, in which correction lasted for the entire period of the experiment (>1 year). Although phenylalanine levels began to rise in female mice at about 8-10 months after rAAV2/8 injection they remained only mildly hyperphenylalaninemic thereafter and subsequent supplementation with synthetic tetrahydrobiopterin resulted in a transient decrease in blood phenylalanine. Alternatively, subsequent administration of a second vector with a different AAV pseudotype to avoid immunity against the previously administrated vector was also successful for long-term treatment of female PKU mice. Overall, this relatively less invasive gene transfer approach completes our previous studies and allows comparison of complementary strategies in the development of efficient PKU gene therapy protocols.


Assuntos
Dependovirus/classificação , Dependovirus/fisiologia , Vetores Genéticos/administração & dosagem , Fenilalanina Hidroxilase/metabolismo , Fenilcetonúrias/terapia , Animais , Dependovirus/genética , Feminino , Terapia Genética , Vetores Genéticos/classificação , Vetores Genéticos/genética , Humanos , Injeções Intramusculares , Fígado/enzimologia , Fígado/metabolismo , Fígado/virologia , Masculino , Camundongos , Fenilalanina Hidroxilase/genética , Fenilcetonúrias/virologia , Sorotipagem , Resultado do Tratamento
20.
South Med J ; 101(5): 543-5, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18414167

RESUMO

A young patient with hepatocellular carcinoma receiving chemotherapy presented with encephalopathy. Evaluation of the patient revealed a metabolic profile consistent with ornithine transcarbamoylase (OTC) deficiency, an inherited disorder of the urea cycle. The evaluation yielded a plasma amino acid analysis consistent with OTC deficiency. However, genetic analysis did not reveal a somatic mutation of the OTC gene in this patient. The hyperammonemic encephalopathy was reversed by the infusion of arginine, a common treatment for hereditary OTC deficiency. This case may represent a distinct syndrome of reversible hyperammonemia in patients with hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/complicações , Encefalopatia Hepática/tratamento farmacológico , Hiperamonemia/diagnóstico , Neoplasias Hepáticas/complicações , Doença da Deficiência de Ornitina Carbomoiltransferase/diagnóstico , Adulto , Arginina/sangue , Arginina/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Citrulina/sangue , Feminino , Glutamina/sangue , Encefalopatia Hepática/diagnóstico , Encefalopatia Hepática/etiologia , Humanos , Hiperamonemia/tratamento farmacológico , Hiperamonemia/etiologia , Neoplasias Hepáticas/tratamento farmacológico , Ornitina/sangue , Doença da Deficiência de Ornitina Carbomoiltransferase/genética , Ácido Orótico/urina , Polimorfismo Conformacional de Fita Simples , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA