Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Am J Hum Genet ; 111(4): 714-728, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38579669

RESUMO

Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production.


Assuntos
Argininossuccinato Liase , Acidúria Argininossuccínica , Humanos , Argininossuccinato Liase/genética , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/terapia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , RNA Guia de Sistemas CRISPR-Cas , Ureia , Edição de Genes/métodos
2.
Stem Cell Res ; 76: 103365, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38422816

RESUMO

Argininosuccinic aciduria (ASA) is a rare inherited metabolic disease caused by argininosuccinate lyase (ASL) deficiency. Patients with ASA present with hyperammonaemia due to an impaired urea cycle pathway in the liver, and systemic disease with epileptic encephalopathy, chronic liver disease, and arterial hypertension. A human induced pluripotent stem cell (iPSC) line from the fibroblasts of a patient with ASA with homozygous pathogenic c.437G > A mutation of hASL was generated. Characterization of the cell line demonstrated pluripotency, differentiation potential and normal karyotype. This cell line, called UCLi024-A, can be utilized for in vitro disease modelling of ASA, and design of novel therapeutics.


Assuntos
Acidúria Argininossuccínica , Células-Tronco Pluripotentes Induzidas , Humanos , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/metabolismo , Acidúria Argininossuccínica/terapia , Células-Tronco Pluripotentes Induzidas/metabolismo , Argininossuccinato Liase/genética , Mutação/genética , Homozigoto
3.
Sci Transl Med ; 16(729): eadh1334, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38198573

RESUMO

The urea cycle enzyme argininosuccinate lyase (ASL) enables the clearance of neurotoxic ammonia and the biosynthesis of arginine. Patients with ASL deficiency present with argininosuccinic aciduria, an inherited metabolic disease with hyperammonemia and a systemic phenotype coinciding with neurocognitive impairment and chronic liver disease. Here, we describe the dysregulation of glutathione biosynthesis and upstream cysteine utilization in ASL-deficient patients and mice using targeted metabolomics and in vivo positron emission tomography (PET) imaging using (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG). Up-regulation of cysteine metabolism contrasted with glutathione depletion and down-regulated antioxidant pathways. To assess hepatic glutathione dysregulation and liver disease, we present [18F]FSPG PET as a noninvasive diagnostic tool to monitor therapeutic response in argininosuccinic aciduria. Human hASL mRNA encapsulated in lipid nanoparticles improved glutathione metabolism and chronic liver disease. In addition, hASL mRNA therapy corrected and rescued the neonatal and adult Asl-deficient mouse phenotypes, respectively, enhancing ureagenesis. These findings provide mechanistic insights in liver glutathione metabolism and support clinical translation of mRNA therapy for argininosuccinic aciduria.


Assuntos
Acidúria Argininossuccínica , Hepatopatias , Adulto , Humanos , Animais , Camundongos , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/terapia , Cisteína , Glutationa , Metabolômica
4.
Am J Cardiol ; 192: 155-159, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36807131

RESUMO

A 39-year-old male was referred for treatment of hypertension. He had been treated for argininosuccinic aciduria since 8 months of age. Therapeutic drugs, including l-arginine, sodium phenylbutyrate, and antiepileptic drugs, had been prescribed. A detailed medical history revealed that he complained of chest discomfort under psychologic stress. A 12-lead electrocardiogram showed abnormal q waves in lead III and aVF. Transthoracic echocardiography showed hypokinesia of the left ventricular posterior wall. The patient was diagnosed with myocardial infarction because of coronary vasospastic angina by intracoronary acetylcholine provocation test. Argininosuccinic aciduria is a genetic disorder of the urea cycle caused by a deficiency of argininosuccinate lyase. Reduction of the enzymatic activity leads to a decrease in nitric oxide production, even if arginine is supplemented. Our case report supports the significance of endothelial function in the pathogenesis of coronary vasospasm.


Assuntos
Acidúria Argininossuccínica , Vasoespasmo Coronário , Masculino , Humanos , Adulto , Acidúria Argininossuccínica/diagnóstico , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/terapia , Argininossuccinato Liase/genética , Angina Pectoris , Arginina
5.
Stem Cell Rev Rep ; 16(1): 186-197, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31792768

RESUMO

Previous studies have shown that human liver stem-like cells (HLSCs) may undergo differentiation in vitro into urea producing hepatocytes and in vivo may sustain liver function in models of experimentally induced acute liver injury. The aim of this study was to assess the safety of HLSCs intrahepatic administration in inherited neonatal-onset hyperammonemia. The study was approved by the Agenzia Italiana del Farmaco on favorable opinion of the Italian Institute of Health as an open-label, prospective, uncontrolled, monocentric Phase I study (HLSC 01-11, EudraCT-No. 2012-002120-33). Three patients affected by argininosuccinic aciduria (patient 1) and methylmalonic acidemia (patients 2 and 3) and included in the liver transplantation list were enrolled. In all patients, HLSCs were administered by percutaneous intrahepatic injections (once a week for two consecutive weeks) within the first months of life. The first patient received 125,000 HLSCs x gram of liver/dose while the other two patients received twice this dose. No immunosuppression was administered since HLSCs possess immunomodulatory activities. None of the patients experienced infections, hyperammonemia decompensation, or other adverse events during the whole observation period. No donor specific antibodies (DSA) against HLSCs were detected. Patients were metabolic stable despite an increase (~30%) in protein intake. Two patients underwent liver transplantation after 19 and 11 months respectively, and after explantation, the native livers showed no histological alterations. In conclusion, percutaneous intrahepatic administration of HLSCs was safe in newborn with inherited neonatal-onset hyperammonemia. These data pave the way for Phase II studies in selected inherited and acquired liver disorders.


Assuntos
Hiperamonemia/terapia , Transplante de Fígado , Fígado/metabolismo , Erros Inatos do Metabolismo/terapia , Transplante de Células-Tronco , Idade de Início , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Amônia/metabolismo , Acidúria Argininossuccínica/metabolismo , Acidúria Argininossuccínica/patologia , Acidúria Argininossuccínica/terapia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Recém-Nascido , Fígado/crescimento & desenvolvimento , Fígado/patologia , Masculino , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Células-Tronco/metabolismo , Ureia/metabolismo
7.
J Inherit Metab Dis ; 42(6): 1147-1161, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30723942

RESUMO

The first patients affected by argininosuccinic aciduria (ASA) were reported 60 years ago. The clinical presentation was initially described as similar to other urea cycle defects, but increasing evidence has shown overtime an atypical systemic phenotype with a paradoxical observation, that is, a higher rate of neurological complications contrasting with a lower rate of hyperammonaemic episodes. The disappointing long-term clinical outcomes of many of the patients have challenged the current standard of care and therapeutic strategy, which aims to normalize plasma ammonia and arginine levels. Interrogations have raised about the benefit of newborn screening or liver transplantation on the neurological phenotype. Over the last decade, novel discoveries enabled by the generation of new transgenic argininosuccinate lyase (ASL)-deficient mouse models have been achieved, such as, a better understanding of ASL and its close interaction with nitric oxide metabolism, ASL physiological role outside the liver, and the pathophysiological role of oxidative/nitrosative stress or excessive arginine treatment. Here, we present a collaborative review, which highlights these recent discoveries and novel emerging concepts about ASL role in human physiology, ASA clinical phenotype and geographic prevalence, limits of current standard of care and newborn screening, pathophysiology of the disease, and emerging novel therapies. We propose recommendations for monitoring of ASA patients. Ongoing research aims to better understand the underlying pathogenic mechanisms of the systemic disease to design novel therapies.


Assuntos
Acidúria Argininossuccínica , Animais , Argininossuccinato Liase/genética , Acidúria Argininossuccínica/diagnóstico , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/patologia , Acidúria Argininossuccínica/terapia , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Hiperamonemia/terapia , Recém-Nascido , Fígado/diagnóstico por imagem , Fígado/patologia , Fígado/cirurgia , Transplante de Fígado , Camundongos , Camundongos Transgênicos , Triagem Neonatal/métodos , Triagem Neonatal/tendências , Estresse Oxidativo/fisiologia , Fenótipo
8.
Mol Genet Metab ; 125(3): 241-250, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30253962

RESUMO

Argininosuccinic aciduria (ASA) is the second most common genetic disorder affecting the urea cycle. The disease is caused by deleterious mutations in the gene encoding argininosuccinate lyase (ASL); total loss of ASL activity results in severe neonatal onset of the disease, which is characterized by hyperammonemia within a few days of birth that can rapidly progress to coma and death. The long-term complications of ASA, such as hypertension and neurocognitive deficits, appear to be resistant to the current treatment options of dietary restriction, arginine supplementation, and nitrogen scavenging drugs. Treatment-resistant disease is currently being managed by orthotopic liver transplant, which shows variable improvement and requires lifetime immunosuppression. Here, we developed a gene therapy strategy for ASA aimed at alleviating the symptoms associated with urea cycle disruption by providing stable expression of ASL protein in the liver. We designed a codon-optimized human ASL gene packaged within adeno-associated virus serotype 8 (AAV8) as a vector for targeted delivery to the liver. To evaluate the therapeutic efficacy of this approach, we utilized a murine hypomorphic model of ASA. Neonatal administration of AAV8 via the temporal facial vein extended survival in ASA hypomorphic mice, although not to wild-type levels. Intravenous injection into adolescent hypomorphic mice led to increased survival and body weight and correction of metabolites associated with the disease. Our results demonstrate that AAV8 gene therapy is a viable approach for the treatment of ASA.


Assuntos
Argininossuccinato Liase/genética , Acidúria Argininossuccínica/terapia , Terapia Genética , Hiperamonemia/terapia , Animais , Argininossuccinato Liase/administração & dosagem , Acidúria Argininossuccínica/genética , Dependovirus/genética , Modelos Animais de Doenças , Humanos , Hiperamonemia/genética , Hiperamonemia/patologia , Camundongos , Ureia/metabolismo
9.
Nat Commun ; 9(1): 3505, 2018 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-30158522

RESUMO

Argininosuccinate lyase (ASL) belongs to the hepatic urea cycle detoxifying ammonia, and the citrulline-nitric oxide (NO) cycle producing NO. ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia, multiorgan disease and neurocognitive impairment despite treatment aiming to normalise ammonaemia without considering NO imbalance. Here we show that cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress independent of hyperammonaemia. Intravenous injection of AAV8 vector into adult or neonatal ASL-deficient mice demonstrates long-term correction of the hepatic urea cycle and the cerebral citrulline-NO cycle, respectively. Cerebral disease persists if ammonaemia only is normalised but is dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This correlates with behavioural improvement and reduced cortical cell death. Thus, neuronal oxidative/nitrosative stress is a distinct pathophysiological mechanism from hyperammonaemia. Disease amelioration by simultaneous brain and liver gene transfer with one vector, to treat both metabolic pathways, provides new hope for hepatocerebral metabolic diseases.


Assuntos
Argininossuccinato Liase/metabolismo , Acidúria Argininossuccínica/metabolismo , Acidúria Argininossuccínica/terapia , Animais , Argininossuccinato Liase/genética , Acidúria Argininossuccínica/genética , Encefalopatias/genética , Encefalopatias/metabolismo , Encefalopatias/terapia , Citrulina/metabolismo , Terapia Genética , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/terapia , Fígado/citologia , Camundongos , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Estresse Nitrosativo/genética , Estresse Nitrosativo/fisiologia
10.
J Inherit Metab Dis ; 40(3): 357-368, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28251416

RESUMO

OBJECTIVES: This UK-wide study defines the natural history of argininosuccinic aciduria and compares long-term neurological outcomes in patients presenting clinically or treated prospectively from birth with ammonia-lowering drugs. METHODS: Retrospective analysis of medical records prior to March 2013, then prospective analysis until December 2015. Blinded review of brain MRIs. ASL genotyping. RESULTS: Fifty-six patients were defined as early-onset (n = 23) if symptomatic < 28 days of age, late-onset (n = 23) if symptomatic later, or selectively screened perinatally due to a familial proband (n = 10). The median follow-up was 12.4 years (range 0-53). Long-term outcomes in all groups showed a similar neurological phenotype including developmental delay (48/52), epilepsy (24/52), ataxia (9/52), myopathy-like symptoms (6/52) and abnormal neuroimaging (12/21). Neuroimaging findings included parenchymal infarcts (4/21), focal white matter hyperintensity (4/21), cortical or cerebral atrophy (4/21), nodular heterotopia (2/21) and reduced creatine levels in white matter (4/4). 4/21 adult patients went to mainstream school without the need of additional educational support and 1/21 lives independently. Early-onset patients had more severe involvement of visceral organs including liver, kidney and gut. All early-onset and half of late-onset patients presented with hyperammonaemia. Screened patients had normal ammonia at birth and received treatment preventing severe hyperammonaemia. ASL was sequenced (n = 19) and 20 mutations were found. Plasma argininosuccinate was higher in early-onset compared to late-onset patients. CONCLUSIONS: Our study further defines the natural history of argininosuccinic aciduria and genotype-phenotype correlations. The neurological phenotype does not correlate with the severity of hyperammonaemia and plasma argininosuccinic acid levels. The disturbance in nitric oxide synthesis may be a contributor to the neurological disease. Clinical trials providing nitric oxide to the brain merit consideration.


Assuntos
Acidúria Argininossuccínica/patologia , Acidúria Argininossuccínica/terapia , Adolescente , Adulto , Amônia/metabolismo , Ácido Argininossuccínico/sangue , Acidúria Argininossuccínica/sangue , Acidúria Argininossuccínica/genética , Criança , Pré-Escolar , Feminino , Seguimentos , Genótipo , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Mutação/genética , Fenótipo , Estudos Prospectivos , Estudos Retrospectivos , Adulto Jovem
12.
Mol Genet Metab ; 107(1-2): 10-4, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22841516

RESUMO

Argininosuccinic aciduria (ASA) is a urea cycle disorder with a complex phenotype. In spite of a lower risk for recurrent hyperammonemic episodes as compared to the proximal disorders of ureagenesis, subjects with ASA are at risk for long-term complications including, poor neurocognitive outcome, hepatic disease and systemic hypertension. These complications can occur in spite of current standard therapy that includes dietary modifications and arginine supplementation suggesting that the presently available therapy is suboptimal. In this article, we discuss the natural history of ASA and the recent mechanistic insights from animal studies that have shown the requirement of argininosuccinate lyase, the enzyme deficient in ASA, for systemic nitric oxide production. These findings may have therapeutic implications and may help optimize therapy in ASA.


Assuntos
Acidúria Argininossuccínica/terapia , Arginina/uso terapêutico , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/metabolismo , Radicais Livres/metabolismo , Estudos de Associação Genética , Humanos , Fenótipo
14.
Genet Med ; 14(5): 501-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22241104

RESUMO

The urea cycle consists of six consecutive enzymatic reactions that convert waste nitrogen into urea. Deficiencies of any of these enzymes of the cycle result in urea cycle disorders (UCDs), a group of inborn errors of hepatic metabolism that often result in life-threatening hyperammonemia. Argininosuccinate lyase (ASL) catalyzes the fourth reaction in this cycle, resulting in the breakdown of argininosuccinic acid to arginine and fumarate. ASL deficiency (ASLD) is the second most common UCD, with a prevalence of ~1 in 70,000 live births. ASLD can manifest as either a severe neonatal-onset form with hyperammonemia within the first few days after birth or as a late-onset form with episodic hyperammonemia and/or long-term complications that include liver dysfunction, neurocognitive deficits, and hypertension. These long-term complications can occur in the absence of hyperammonemic episodes, implying that ASL has functions outside of its role in ureagenesis and the tissue-specific lack of ASL may be responsible for these manifestations. The biochemical diagnosis of ASLD is typically established with elevation of plasma citrulline together with elevated argininosuccinic acid in the plasma or urine. Molecular genetic testing of ASL and assay of ASL enzyme activity are helpful when the biochemical findings are equivocal. However, there is no correlation between the genotype or enzyme activity and clinical outcome. Treatment of acute metabolic decompensations with hyperammonemia involves discontinuing oral protein intake, supplementing oral intake with intravenous lipids and/or glucose, and use of intravenous arginine and nitrogen-scavenging therapy. Dietary restriction of protein and dietary supplementation with arginine are the mainstays in long-term management. Orthotopic liver transplantation (OLT) is best considered only in patients with recurrent hyperammonemia or metabolic decompensations resistant to conventional medical therapy.


Assuntos
Acidúria Argininossuccínica/diagnóstico , Acidúria Argininossuccínica/genética , Arginina/metabolismo , Arginina/uso terapêutico , Argininossuccinato Liase/genética , Ácido Argininossuccínico/sangue , Ácido Argininossuccínico/metabolismo , Ácido Argininossuccínico/urina , Acidúria Argininossuccínica/terapia , Pré-Escolar , Citrulina/sangue , Transtornos Cognitivos/enzimologia , Transtornos Cognitivos/genética , Dieta com Restrição de Proteínas , Fumaratos/metabolismo , Testes Genéticos , Glucose/uso terapêutico , Humanos , Hiperamonemia/enzimologia , Hiperamonemia/genética , Hipertensão/enzimologia , Hipertensão/genética , Lactente , Recém-Nascido , Lipídeos/uso terapêutico , Hepatopatias/enzimologia , Hepatopatias/genética , Transplante de Fígado , Triagem Neonatal , Fenilbutiratos/uso terapêutico , Benzoato de Sódio/uso terapêutico
16.
Am J Med Genet C Semin Med Genet ; 157C(1): 45-53, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21312326

RESUMO

The urea cycle consists of six consecutive enzymatic reactions that convert waste nitrogen into urea. Deficiencies of any of these enzymes of the cycle result in urea cycle disorders (UCD), a group of inborn errors of hepatic metabolism that often result in life threatening hyperammonemia. Argininosuccinate lyase (ASL) is a cytosolic enzyme which catalyzes the fourth reaction in the cycle and the first degradative step, that is, the breakdown of argininosuccinic acid to arginine and fumarate. Deficiency of ASL results in an accumulation of argininosuccinic acid in tissues, and excretion of argininosuccinic acid in urine leading to the condition argininosuccinic aciduria (ASA). ASA is an autosomal recessive disorder and is the second most common UCD. In addition to the accumulation of argininosuccinic acid, ASL deficiency results in decreased synthesis of arginine, a feature common to all UCDs except argininemia. Arginine is not only the precursor for the synthesis of urea and ornithine as part of the urea cycle but it is also the substrate for the synthesis of nitric oxide, polyamines, proline, glutamate, creatine, and agmatine. Hence, while ASL is the only enzyme in the body able to generate arginine, at least four enzymes use arginine as substrate: arginine decarboxylase, arginase, nitric oxide synthetase (NOS) and arginine/glycine aminotransferase. In the liver, the main function of ASL is ureagenesis, and hence, there is no net synthesis of arginine. In contrast, in most other tissues, its role is to generate arginine that is designated for the specific cell's needs. While patients with ASA share the acute clinical phenotype of hyperammonemia, encephalopathy, and respiratory alkalosis common to other UCD, they also present with unique chronic complications most probably caused by a combination of tissue specific deficiency of arginine and/or elevation of argininosuccinic acid. This review article summarizes the clinical characterization, biochemical, enzymatic, and molecular features of this disorder. Current treatment, prenatal diagnosis, diagnosis through the newborn screening as well as hypothesis driven future treatment modalities are discussed.


Assuntos
Argininossuccinato Liase/genética , Acidúria Argininossuccínica/diagnóstico , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/metabolismo , Arginase/genética , Arginase/metabolismo , Arginina/genética , Arginina/metabolismo , Ácido Argininossuccínico/metabolismo , Acidúria Argininossuccínica/terapia , Carboxiliases/genética , Carboxiliases/metabolismo , Humanos , Hiperamonemia/enzimologia , Hiperamonemia/genética , Hiperamonemia/metabolismo , Recém-Nascido , Hepatopatias/enzimologia , Hepatopatias/genética , Hepatopatias/metabolismo , Triagem Neonatal , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Ornitina/genética , Ornitina/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/enzimologia , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA