Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Am J Hum Genet ; 111(4): 714-728, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38579669

ABSTRACT

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.


Subject(s)
Argininosuccinate Lyase , Argininosuccinic Aciduria , Humans , Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/therapy , Clustered Regularly Interspaced Short Palindromic Repeats , RNA, Guide, CRISPR-Cas Systems , Urea , Gene Editing/methods
2.
Stem Cell Res ; 76: 103365, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422816

ABSTRACT

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.


Subject(s)
Argininosuccinic Aciduria , Induced Pluripotent Stem Cells , Humans , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Argininosuccinic Aciduria/therapy , Induced Pluripotent Stem Cells/metabolism , Argininosuccinate Lyase/genetics , Mutation/genetics , Homozygote
3.
Sci Transl Med ; 16(729): eadh1334, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38198573

ABSTRACT

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.


Subject(s)
Argininosuccinic Aciduria , Liver Diseases , Adult , Humans , Animals , Mice , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/therapy , Cysteine , Glutathione , Metabolomics
4.
Clin Dysmorphol ; 33(1): 43-49, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37865865

ABSTRACT

Argininosuccinate lyase (ASL) deficiency is an autosomal recessive disorder of the urea cycle with a diverse spectrum of clinical presentation that is detectable in newborn screening. We report an 8-year-old girl with ASL deficiency who was detected through newborn screening and was confirmed using biochemical and functional assay. She is compound heterozygous for a likely pathogenic variant NM_000048.4(ASL):c.283C>T (p.Arg95Cys) and a likely benign variant NM_000048.4(ASL): c.1319T>C (p.Leu440Pro). Functional characterisation of the likely benign genetic variant in ASL was performed. Genomic sequencing was performed on the index patient presenting with non-specific symptoms of poor feeding and lethargy and shown to have increased serum and urine argininosuccinic acid. Functional assay using HEK293T cell model was performed. ASL enzymatic activity was reduced for Leu440Pro. This study highlights the role of functional testing of a variant that may appear benign in a patient with a phenotype consistent with ASL deficiency, and reclassifies NM_000048.4(ASL): c.1319T>C (p.Leu440Pro) variant as likely pathogenic.


Subject(s)
Argininosuccinic Aciduria , Infant, Newborn , Female , Humans , Child , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/chemistry , Argininosuccinate Lyase/metabolism , Neonatal Screening , HEK293 Cells , Base Sequence
5.
JCI Insight ; 8(17)2023 09 08.
Article in English | MEDLINE | ID: mdl-37490345

ABSTRACT

Nitric oxide (NO) is a critical signaling molecule that has been implicated in the pathogenesis of neurocognitive diseases. Both excessive and insufficient NO production have been linked to pathology. Previously, we have shown that argininosuccinate lyase deficiency (ASLD) is a novel model system to investigate cell-autonomous, nitric oxide synthase-dependent NO deficiency. Humans with ASLD are at increased risk for developing hyperammonemia due to a block in ureagenesis. However, natural history studies have shown that individuals with ASLD have multisystem disease including neurocognitive deficits that can be independent of ammonia. Here, using ASLD as a model of NO deficiency, we investigated the effects of NO on brain endothelial cells in vitro and the blood-brain barrier (BBB) in vivo. Knockdown of ASL in human brain microvascular endothelial cells (HBMECs) led to decreased transendothelial electrical resistance, indicative of increased cell permeability. Mechanistically, treatment with an NO donor or inhibition of Claudin-1 improved barrier integrity in ASL-deficient HBMECs. Furthermore, in vivo assessment of a hypomorphic mouse model of ASLD showed increased BBB leakage, which was partially rescued by NO supplementation. Our results suggest that ASL-mediated NO synthesis is required for proper maintenance of brain microvascular endothelial cell functions as well as BBB integrity.


Subject(s)
Argininosuccinic Aciduria , Mice , Animals , Humans , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Argininosuccinic Aciduria/pathology , Nitric Oxide/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Claudins/metabolism , Disease Models, Animal
6.
Am J Transplant ; 23(5): 673-675, 2023 05.
Article in English | MEDLINE | ID: mdl-36870389

ABSTRACT

Liver transplantation risks transferring a genetic defect in metabolic pathways, including the urea cycle. We present a case of pediatric liver transplantation complicated by metabolic crisis and early allograft dysfunction (EAD) in a previously healthy unrelated deceased donor. Allograft function improved with supportive care, and retransplantation was avoided. Because hyperammonemia suggested an enzymatic defect in the allograft, genetic testing from donor-derived deoxyribonucleic acid revealed a heterozygous mutation in the ASL gene, which encodes the urea cycle enzyme argininosuccinate lyase. Homozygous ASL mutations precipitate metabolic crises during fasting or postoperative states, whereas heterozygous carriers retain sufficient enzyme activity and are asymptomatic. In the described case, postoperative ischemia/reperfusion injury created a metabolic demand that exceeded the enzymatic capacity of the allograft. To our knowledge, this is the first report of an acquired argininosuccinate lyase deficiency by liver transplantation and underscores the importance of considering occult metabolic variants in the allograft during EAD.


Subject(s)
Argininosuccinic Aciduria , Humans , Child , Mutation , Argininosuccinic Aciduria/genetics , Liver , Allografts , Urea
7.
Epilepsia ; 64(6): 1612-1626, 2023 06.
Article in English | MEDLINE | ID: mdl-36994644

ABSTRACT

OBJECTIVE: Argininosuccinate lyase (ASL) is integral to the urea cycle, which enables nitrogen wasting and biosynthesis of arginine, a precursor of nitric oxide. Inherited ASL deficiency causes argininosuccinic aciduria, the second most common urea cycle defect and an inherited model of systemic nitric oxide deficiency. Patients present with developmental delay, epilepsy, and movement disorder. Here we aim to characterize epilepsy, a common and neurodebilitating comorbidity in argininosuccinic aciduria. METHODS: We conducted a retrospective study in seven tertiary metabolic centers in the UK, Italy, and Canada from 2020 to 2022, to assess the phenotype of epilepsy in argininosuccinic aciduria and correlate it with clinical, biochemical, radiological, and electroencephalographic data. RESULTS: Thirty-seven patients, 1-31 years of age, were included. Twenty-two patients (60%) presented with epilepsy. The median age at epilepsy onset was 24 months. Generalized tonic-clonic and focal seizures were most common in early-onset patients, whereas atypical absences were predominant in late-onset patients. Seventeen patients (77%) required antiseizure medications and six (27%) had pharmacoresistant epilepsy. Patients with epilepsy presented with a severe neurodebilitating disease with higher rates of speech delay (p = .04) and autism spectrum disorders (p = .01) and more frequent arginine supplementation (p = .01) compared to patients without epilepsy. Neonatal seizures were not associated with a higher risk of developing epilepsy. Biomarkers of ureagenesis did not differ between epileptic and non-epileptic patients. Epilepsy onset in early infancy (p = .05) and electroencephalographic background asymmetry (p = .0007) were significant predictors of partially controlled or refractory epilepsy. SIGNIFICANCE: Epilepsy in argininosuccinic aciduria is frequent, polymorphic, and associated with more frequent neurodevelopmental comorbidities. We identified prognostic factors for pharmacoresistance in epilepsy. This study does not support defective ureagenesis as prominent in the pathophysiology of epilepsy but suggests a role of central dopamine deficiency. A role of arginine in epileptogenesis was not supported and warrants further studies to assess the potential arginine neurotoxicity in argininosuccinic aciduria.


Subject(s)
Argininosuccinic Aciduria , Epilepsy , Humans , Argininosuccinic Aciduria/complications , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Retrospective Studies , Nitric Oxide , Arginine/metabolism , Arginine/therapeutic use , Epilepsy/complications , Epilepsy/epidemiology , Epilepsy/drug therapy , Urea , Seizures/drug therapy
8.
Am J Cardiol ; 192: 155-159, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36807131

ABSTRACT

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.


Subject(s)
Argininosuccinic Aciduria , Coronary Vasospasm , Male , Humans , Adult , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/therapy , Argininosuccinate Lyase/genetics , Angina Pectoris , Arginine
9.
BMJ Case Rep ; 14(3)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33789861

ABSTRACT

Argininosuccinate lyase (ASL) deficiency is a rare autosomal recessive urea cycle disorder. The severe neonatal-onset form is characterised by hyperammonaemia in the first days of life and manifests with a variety of severe symptoms. However, an index of suspicion for additional or alternative diagnoses must be maintained when the patient's presentation is out of keeping with expected manifestations and course. We present a case of a neonate with ASL deficiency and concomitant hypotonia, severe respiratory distress, pulmonary hypertension, systemic hypotension and congenital hypothyroidism. The patient was investigated and subsequently diagnosed with brain-lung-thyroid syndrome, caused by a mutation in the NKX2-1 gene.


Subject(s)
Argininosuccinic Aciduria , Chorea , Congenital Hypothyroidism , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Athetosis , Humans , Infant, Newborn , Respiratory Distress Syndrome, Newborn
10.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33373331

ABSTRACT

Previous studies have shown that nitric oxide (NO) supplements may prevent bone loss and fractures in preclinical models of estrogen deficiency. However, the mechanisms by which NO modulates bone anabolism remain largely unclear. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing arginine, the sole precursor for nitric oxide synthase-dependent (NOS-dependent) NO synthesis. Moreover, ASL is also required for channeling extracellular arginine to NOS for NO production. ASL deficiency (ASLD) is thus a model to study cell-autonomous, NOS-dependent NO deficiency. Here, we report that loss of ASL led to decreased NO production and impairment of osteoblast differentiation. Mechanistically, the bone phenotype was at least in part driven by the loss of NO-mediated activation of the glycolysis pathway in osteoblasts that led to decreased osteoblast differentiation and function. Heterozygous deletion of caveolin 1, a negative regulator of NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass in a hypomorphic mouse model of ASLD. The translational significance of these preclinical studies was further reiterated by studies conducted in induced pluripotent stem cells from an individual with ASLD. Taken together, our findings suggest that ASLD is a unique genetic model for studying NO-dependent osteoblast function and that the NO/glycolysis pathway may be a new target to modulate bone anabolism.


Subject(s)
Argininosuccinic Aciduria/metabolism , Bone and Bones/metabolism , Cell Differentiation , Glycolysis , Nitric Acid/metabolism , Osteoblasts/metabolism , Adolescent , Adult , Animals , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/pathology , Bone and Bones/pathology , Child , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Osteoblasts/pathology
11.
Mol Genet Metab ; 131(4): 390-397, 2020 12.
Article in English | MEDLINE | ID: mdl-33288448

ABSTRACT

OBJECTIVE: The implementation of newborn screening (NBS) programs for citrullinemia type 1 (CTLN1) and argininosuccinic aciduria (ASA) is subject to controversial debate. The aim of this study was to assess the impact of NBS on the metabolic disease course and clinical outcome of affected individuals. METHODS: In 115 individuals with CTLN1 and ASA, we compared the severity of the initial hyperammonemic episode (HAE) and the frequency of (subsequent) HAEs with the mode of diagnosis. Based on a recently established functional disease prediction model, individuals were stratified according to their predicted severe or attenuated phenotype. RESULTS: Individuals with predicted attenuated forms of CTLN1 and ASA were overrepresented in the NBS group, while those with a predicted severe phenotype were underrepresented compared to individuals identified after the manifestation of symptoms (SX). Identification by NBS was associated with reduced severity of the initial HAE both in individuals with predicted severe and attenuated phenotypes, while it was not associated with lower frequency of (subsequent) HAEs. Similar results were obtained when including some patients diagnosed presymptomatically (i.e. prenatal testing, and high-risk family screening) in this analysis. CONCLUSION: Since one of the major challenges of NBS outcome studies is the potential overrepresentation of individuals with predicted attenuated phenotypes in NBS cohorts, severity-adjusted evaluation of screened and unscreened individuals is important to avoid overestimation of the NBS effect. NBS enables the attenuation of the initial HAE but does not affect the frequency of subsequent metabolic decompensations in individuals with CTLN1 and ASA. Future long-term studies will need to evaluate the clinical impact of this finding, especially with regard to mortality, as well as cognitive outcome and quality of life of survivors.


Subject(s)
Argininosuccinic Aciduria/diagnosis , Citrullinemia/diagnosis , Metabolic Diseases/genetics , Urea Cycle Disorders, Inborn/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Argininosuccinic Aciduria/pathology , Citrullinemia/genetics , Citrullinemia/metabolism , Citrullinemia/pathology , Female , Humans , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Infant, Newborn , Male , Metabolic Diseases/diagnosis , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Neonatal Screening , Quality of Life , Severity of Illness Index , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology
12.
Mol Genet Genomic Med ; 8(7): e1301, 2020 07.
Article in English | MEDLINE | ID: mdl-32410394

ABSTRACT

BACKGROUND: The urea cycle plays a key role in preventing the accumulation of toxic nitrogenous waste products, including two essential enzymes: ornithine transcarbamylase (OTC) and argininosuccinate lyase (ASL). Ornithine transcarbamylase deficiency (OTCD) results from mutations in the OTC. Meanwhile, argininosuccinate lyase deficiency (ASLD) is caused by mutations in the ASL. METHODS: Blood tandem mass spectrometric analysis and urea organic acidemia screening were performed on five Chinese cases, including three OTCD and two ASLD patients. Next-generation sequencing was then used to make a definite diagnosis, and the related variants were validated by Sanger sequencing. RESULTS: The five patients exhibited severe clinical symptoms, with abnormal biochemical analysis and amino acids profile. Genetic analysis revealed two variants [c.77G>A (p.Arg26Gln); c.116G>T (p.Gly39Val)] in the OTC, as well as two variants [c.1311T>G (p.Tyr437*); c.961T>A (p.Tyr321Asn)] in the ASL. Conservation analysis showed that the amino acids of the two novel mutations were highly conserved in different species and were predicted to be possibly damaging with several in silico prediction programs. 3D-modeling analysis indicated that the two novel missense variants might result in modest distortions of the OTC and ASL protein structures, respectively. CONCLUSIONS: Two novel variants expand the mutational spectrums of the OTC and ASL. All the results may contribute to a better understanding of the clinical course and genetic characteristics of patients with urea cycle disorders.


Subject(s)
Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/genetics , Mutation , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase/genetics , Argininosuccinate Lyase/chemistry , Argininosuccinic Aciduria/pathology , Female , Humans , Infant , Male , Molecular Dynamics Simulation , Ornithine Carbamoyltransferase/chemistry , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Pedigree , Protein Domains
13.
Hum Mutat ; 41(5): 946-960, 2020 05.
Article in English | MEDLINE | ID: mdl-31943503

ABSTRACT

Argininosuccinic aciduria (ASA) is an inherited urea cycle disorder and has a highly variable phenotypic spectrum ranging from individuals with lethal hyperammonemic encephalopathy, liver dysfunction, and cognitive deterioration, to individuals with a mild disease course. As it is difficult to predict the phenotypic severity, we aimed at identifying a reliable disease prediction model. We applied a biallelic expression system to assess the functional impact of pathogenic argininosuccinate lyase (ASL) variants and to determine the enzymatic activity of ASL in 58 individuals with ASA. This cohort represented 42 ASL gene variants and 42 combinations in total. Enzymatic ASL activity was compared with biochemical and clinical endpoints from the UCDC and E-IMD databases. Enzymatic ASL activity correlated with peak plasma ammonium concentration at initial presentation and with the number of hyperammonemic events (HAEs) per year of observation. Individuals with ≤9% of enzymatic activity had more severe initial decompensations and a higher annual frequency of HAEs than individuals above this threshold. Enzymatic ASL activity also correlated with the cognitive outcome and the severity of the liver disease, enabling a reliable severity prediction for individuals with ASA. Thus, enzymatic activity measured by this novel expression system can serve as an important marker of phenotypic severity.


Subject(s)
Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Phenotype , Adolescent , Adult , Argininosuccinate Lyase/blood , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Argininosuccinic Aciduria/metabolism , Biomarkers , Child , Child, Preschool , Enzyme Activation , Female , Gene Expression , Genetic Association Studies/methods , Humans , Kidney/metabolism , Liver/metabolism , Male , Middle Aged , Mutation , RNA, Messenger/genetics , Severity of Illness Index , Young Adult
14.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(9): 926-929, 2019 Sep 10.
Article in Chinese | MEDLINE | ID: mdl-31515792

ABSTRACT

OBJECTIVE: To explore the genetic basis of a neonate with argininosuccinic aciduria (ASA). METHODS: A neonate with lethargy and food refusal was admitted. The patient had myoclonus, myasthenia, uroschesis, irregular breathing and paroxysmal ventricular tachycardia, and died at 75 hours after birth. Laboratory test showed marked increase in blood ammonia (1249.8 µmol/L). Peripheral blood samples of the patient, her parents and sister were collected and subjected to trio whole-exome sequencing. RESULTS: Whole-exome sequencing revealed that the patient has carried compound heterozygous mutations of the argininosuccinate lyase (ASL) gene, namely c.425(exon5)_c.426(exon5) insAGCTCCCAGCT (p.Thr142Thrfs*37) and c.626(exon8)delT (p.Leu209Argfs*42). The patient was diagnosed as ASA caused by ASL gene mutations. Her parents and her elder sister were heterozygous carriers of the above mutations and had a normal phenotype. CONCLUSION: ASA is a severe congenital genetic metabolic disease and can manifest as onset of hyperammonemia in neonates. The clinical diagnosis is difficult and ASL gene testing may be helpful.


Subject(s)
Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Hyperammonemia , Female , Genetic Testing , Humans , Infant, Newborn , Pedigree
15.
Orphanet J Rare Dis ; 14(1): 203, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31426867

ABSTRACT

BACKGROUND: The incidence, prevalence, and molecular epidemiology of urea cycle disorders (UCDs) in Argentina remain underexplored. The present study is the first to thoroughly assess the clinical and molecular profiles of UCD patients examined at a single reference center in Argentina. RESULTS: Forty-nine UCD cases were collected. About half (26/49, 53%) manifested neonatally with classical presentation and had a high mortality (25/26, 96%). Ornithine transcarbamylase deficiency (OTCD) was the most common UCD (26 patients). Argininosuccinate synthetase deficiency (ASSD) was detected in 19 cases, while argininosuccinate lyase deficiency (ASLD) was diagnosed in 4 cases. Molecular genetic analysis revealed 8 private OTC mutations and two large deletion/duplication events in the OTC gene. Most mutations in the ASS1 and ASL genes were recurrent missense changes, and four alterations were novel. The clinical outcome of our UCD cohort was poor, with an overall mortality of 57% (28/49 cases), and a 28% (6/21) disability rate among the survivors. CONCLUSIONS: Most patients in our case series showed severe neonatal onset, with high morbidity/mortality. We detected in total 19 mutations, most of them recurrent and of high frequency worldwide. Noteworthy, we highlight the presence of a geographic cluster with high prevalence of a point mutation in the ASS1 gene. This study suggests that these disorders may be more frequent than commonly assumed, and stresses the need for increased awareness amongst health professionals and greater availability of diagnostic tools for accurate identification, early diagnosis, and timely treatment.


Subject(s)
Urea Cycle Disorders, Inborn/epidemiology , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/pathology , Argentina/epidemiology , Argininosuccinic Aciduria/epidemiology , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/pathology , Child , Child, Preschool , Citrullinemia/epidemiology , Citrullinemia/genetics , Citrullinemia/pathology , Female , Humans , Hyperammonemia/epidemiology , Hyperammonemia/genetics , Hyperammonemia/pathology , Infant , Infant, Newborn , Male , Mutation/genetics , Ornithine Carbamoyltransferase Deficiency Disease/epidemiology , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/pathology
17.
Biomed Res Int ; 2019: 3530198, 2019.
Article in English | MEDLINE | ID: mdl-31183366

ABSTRACT

Pathogenic variants in the argininosuccinate lyase (ASL) gene have been shown to cause argininosuccinate lyase deficiency (ASLD); therefore, sequencing analysis offers advantages for prenatal testing and counseling in families afflicted with this condition. Here, we performed a genetic analysis of an ASLD patient and his family with an aim to offer available information for clinical diagnosis. The research subjects were a 23-month-old patient with a high plasma level of citrulline and his unaffected parents. Whole-exome sequencing identified potential related ASL gene mutations in this trio. Enzymatic activity was detected spectrophotometrically by a coupled assay using arginase and measuring urea production. We identified a novel nonsynonymous mutation (c.206A>G, p.Lys69Arg) and a stop mutation (c.637C>T, p.Arg213∗) in ASL in a Chinese Han patient with ASLD. The enzymatic activity of a p.Lys69Arg ASL construct in human embryonic kidney 293T cells was significantly reduced compared to that of the wild-type construct, and no significant activity was observed for the p.Arg213∗ construct. Compound heterozygous p.Lys69Arg and p.Arg213∗ mutations that resulted in reduced ASL enzyme activity were found in a patient with ASLD. This finding expands the clinical spectrum of ASL pathogenic variants.


Subject(s)
Argininosuccinate Lyase , Argininosuccinic Aciduria , Exome Sequencing , Heterozygote , Mutation, Missense , Amino Acid Substitution , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Argininosuccinic Aciduria/enzymology , Argininosuccinic Aciduria/genetics , Citrulline/blood , Citrulline/genetics , HEK293 Cells , Humans , Infant , Male
18.
J Inherit Metab Dis ; 42(6): 1077-1087, 2019 11.
Article in English | MEDLINE | ID: mdl-30907007

ABSTRACT

The urea cycle disorder (UCD) argininosuccinate lyase (ASL) deficiency, caused by a defective ASL enzyme, exhibits a wide range of phenotypes, from life-threatening neonatal hyperammonemia to asymptomatic patients, with only the biochemical marker argininosuccinic acid (ASA) elevated in body fluids. Remarkably, even without ever suffering from hyperammonemia, patients often develop severe cognitive impairment and seizures. The goal of this study was to understand the effect on the known toxic metabolite ASA and the assumed toxic metabolite guanidinosuccinic acid (GSA) on developing brain cells, and to evaluate the potential role of creatine (Cr) supplementation, as it was described protective for brain cells exposed to ammonia. We used an in vitro model, in which we exposed three-dimensional (3D) organotypic rat brain cell cultures in aggregates to different combinations of the metabolites of interest at two time points (representing two different developmental stages). After harvest and cryopreservation of the cell cultures, the samples were analyzed mainly by metabolite analysis, immunohistochemistry, and western blotting. ASA and GSA were found toxic for astrocytes and neurons. This toxicity could be reverted in vitro by Cr. As well, an antiapoptotic effect of ASA was revealed, which could contribute to the neurotoxicity in ASL deficiency. Further studies in human ASL deficiency will be required to understand the biochemical situation in the brain of affected patients, and to investigate the impact of high or low arginine doses on brain Cr availability. In addition, clinical trials to evaluate the beneficial effect of Cr supplementation in ASL deficiency would be valuable.


Subject(s)
Argininosuccinic Acid/toxicity , Argininosuccinic Aciduria/pathology , Argininosuccinic Aciduria/prevention & control , Brain/pathology , Creatine/pharmacology , Neurotoxicity Syndromes/pathology , Animals , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Brain/drug effects , Brain/metabolism , Cells, Cultured , Humans , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/metabolism , Organ Culture Techniques/methods , Rats , Tissue Scaffolds/chemistry
19.
J Inherit Metab Dis ; 42(6): 1147-1161, 2019 11.
Article in English | MEDLINE | ID: mdl-30723942

ABSTRACT

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.


Subject(s)
Argininosuccinic Aciduria , Animals , Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/pathology , Argininosuccinic Aciduria/therapy , Humans , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Hyperammonemia/therapy , Infant, Newborn , Liver/diagnostic imaging , Liver/pathology , Liver/surgery , Liver Transplantation , Mice , Mice, Transgenic , Neonatal Screening/methods , Neonatal Screening/trends , Oxidative Stress/physiology , Phenotype
20.
Mol Genet Metab ; 125(3): 241-250, 2018 11.
Article in English | MEDLINE | ID: mdl-30253962

ABSTRACT

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.


Subject(s)
Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/therapy , Genetic Therapy , Hyperammonemia/therapy , Animals , Argininosuccinate Lyase/administration & dosage , Argininosuccinic Aciduria/genetics , Dependovirus/genetics , Disease Models, Animal , Humans , Hyperammonemia/genetics , Hyperammonemia/pathology , Mice , Urea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...