Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 507
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.
Clin Dysmorphol ; 33(1): 43-49, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37865865

RESUMO

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.


Assuntos
Acidúria Argininossuccínica , Recém-Nascido , Feminino , Humanos , Criança , Acidúria Argininossuccínica/diagnóstico , Acidúria Argininossuccínica/genética , Argininossuccinato Liase/genética , Argininossuccinato Liase/química , Argininossuccinato Liase/metabolismo , Triagem Neonatal , Células HEK293 , Sequência de Bases
4.
Hum Mol Genet ; 33(1): 33-37, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37738569

RESUMO

Inhaled nitric oxide (NO) therapy has been reported to improve lung growth in premature newborns. However, the underlying mechanisms by which NO regulates lung development remain largely unclear. NO is enzymatically produced by three isoforms of nitric oxide synthase (NOS) enzymes. NOS knockout mice are useful tools to investigate NO function in the lung. Each single NOS knockout mouse does not show obvious lung alveolar phenotype, likely due to compensatory mechanisms. While mice lacking all three NOS isoforms display impaired lung alveolarization, implicating NO plays a pivotal role in lung alveolarization. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing L-arginine, the sole precursor for NOS-dependent NO synthesis. ASL is also required for channeling extracellular L-arginine into a NO-synthetic complex. Thus, ASL deficiency (ASLD) is a non-redundant model for cell-autonomous, NOS-dependent NO deficiency. Here, we assessed lung alveolarization in ASL-deficient mice. Hypomorphic deletion of Asl (AslNeo/Neo) results in decreased lung alveolarization, accompanied with reduced level of S-nitrosylation in the lung. Genetic ablation of one copy of Caveolin-1, which is a negative regulator of NO production, restores total S-nitrosylation as well as lung alveolarization in AslNeo/Neo mice. Importantly, NO supplementation could partially rescue lung alveolarization in AslNeo/Neo mice. Furthermore, endothelial-specific knockout mice (VE-Cadherin Cre; Aslflox/flox) exhibit impaired lung alveolarization at 12 weeks old, supporting an essential role of endothelial-derived NO in the enhancement of lung alveolarization. Thus, we propose that ASLD is a model to study NO-mediated lung alveolarization.


Assuntos
Argininossuccinato Liase , Óxido Nítrico , Animais , Camundongos , Argininossuccinato Liase/genética , Óxido Nítrico Sintase/genética , Arginina/genética , Camundongos Knockout , Pulmão , Isoformas de Proteínas , Mamíferos
5.
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
6.
Mol Cell ; 82(20): 3919-3931.e7, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36270249

RESUMO

Cancer-specific TERT promoter mutations have been linked to the reactivation of epigenetically silenced TERT gene by creating de novo binding motifs for E-Twenty-Six transcription factors, especially GABPA. How these mutations switch on TERT from epigenetically repressed states to expressed states have not been defined. Here, we revealed that EGFR activation induces ERK1/2-dependent phosphorylation of argininosuccinate lyase (ASL) at Ser417 (S417), leading to interactions between ASL and GABPA at the mutant regions of TERT promoters. The ASL-generated fumarate inhibits KDM5C, leading to enhanced trimethylation of histone H3 Lys4 (H3K4me3), which in turn promotes the recruitment of c-Myc to TERT promoters for TERT expression. Expression of ASL S417A, which abrogates its binding with GABPA, results in reduced TERT expression, inhibited telomerase activity, shortened telomere length, and impaired brain tumor growth in mice. This study reveals an unrecognized mechanistic insight into epigenetically activation of mutant TERT promoters where GABPA-interacted ASL plays an instrumental role.


Assuntos
Glioblastoma , Telomerase , Animais , Camundongos , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/genética , Fumaratos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Histonas/genética , Histonas/metabolismo , Mutação , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo , Encurtamento do Telômero , Fatores de Transcrição/metabolismo , Regiões Promotoras Genéticas
7.
Biomed Res Int ; 2022: 9008685, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35782058

RESUMO

Despite significant improvements in treatment and survival in paediatric cancers, outcomes for children with brain tumours remain poor. Novel therapeutic approaches are needed to improve survival and quality of survival. Extracellular arginine dependency (auxotrophy) has been recognised in several tumours as a potential therapeutic target. This dependency is due to the inability of cancer cells to recycle or synthesise intracellular arginine through the urea cycle pathway compared to normal cells. Whilst adult glioblastoma exhibits this dependency, the expression of the arginine pathway enzymes has not been delineated in paediatric brain tumours. We used immunohistochemical (IHC) methods to stain for arginine pathway enzymes in paediatric high-grade glioma (pHGG), low-grade glioma (pLGG), ependymoma (EPN), and medulloblastoma (MB) tumour tissue microarrays (TMAs). The antibodies detected protein expression of the metaboliser arginase (Arg1 and Arg2); recycling enzymes ornithine transcarbamoylase (OTC), argininosuccinate synthetase (ASS1), and argininosuccinate lyase (ASL); and the transporter SLC7A1. Deficiency of OTC, ASS1, and ASL was seen in 87.5%, 94%, and 79% of pHGG samples, respectively, consistent with an auxotrophic signature. Similar result was obtained in pLGG with 96%, 93%, and 91% of tumours being deficient in ASL, ASS1, and OTC, respectively. 79%, 88%, and 85% of MB cases were ASL, ASS1, and OTC deficient whilst ASL and OTC were deficient in 57% and 91% of EPN samples. All tumour types highly expressed SLC7A1 and Arginase, with Arg2 being the main isoform, demonstrating that they could transport and utilise arginine. Our results show that pHGG, pLGG, EPN, and MB demonstrate arginine auxotrophy based on protein expression and are likely to be susceptible to arginine depletion. Pegylated arginase (BCT-100) is currently in phase I/II trials in relapsed pHGG. Our results suggest that therapeutic arginine depletion may also be useful in other tumour types and IHC analysis of patient tumour samples could help identify patients likely to benefit from this treatment.


Assuntos
Neoplasias Encefálicas , Neoplasias Cerebelares , Glioma , Meduloblastoma , Adulto , Arginase/genética , Arginina , Argininossuccinato Liase , Neoplasias Encefálicas/genética , Criança , Ependimoma , Glioma/genética , Humanos , Ornitina Carbamoiltransferase
8.
Plant Mol Biol ; 110(1-2): 13-22, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35583703

RESUMO

KEY MESSAGE: This study revealed different catalytic efficiencies of cyanobacterial argininosuccinate lyases in non-nitrogen-fixing and nitrogen-fixing cyanobacteria, demonstrating that L-arginine inhibition of L-argininosuccinate lyase is conserved among enzymes of three cyanobacterial orders. Arginine is a nitrogen-rich amino acid that uses a nitrogen reservoir, and its biosynthesis is strictly controlled by feedback inhibition. Argininosuccinate lyase (EC 4.3.2.1) is the final enzyme in arginine biosynthesis that catalyzes the conversion of argininosuccinate to L-arginine and fumarate. Cyanobacteria synthesize intracellular cyanophycin, which is a nitrogen reservoir composed of aspartate and arginine. Arginine is an important source of nitrogen for cyanobacteria. We expressed and purified argininosuccinate lyases, ArgHs, from Synechocystis sp. PCC 6803, Nostoc sp. PCC 7120, and Arthrospira platensis NIES-39. The catalytic efficiency of the Nostoc sp. PCC 7120 ArgH was 2.8-fold higher than those of Synechocystis sp. PCC 6803 and Arthrospira platensis NIES-39. All three ArgHs were inhibited in the presence of arginine, and their inhibitory effects were lowered at pH 7.0, compared to those at pH 8.0. These results indicate that arginine inhibition of ArgH is widely conserved among the three cyanobacterial orders. The current results demonstrate the conserved regulation of enzymes in the cyanobacterial aspartase/fumarase superfamily.


Assuntos
Liases , Synechocystis , Arginina/metabolismo , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Liases/metabolismo , Nitrogênio/metabolismo , Spirulina , Synechocystis/genética , Synechocystis/metabolismo
9.
NPJ Syst Biol Appl ; 7(1): 36, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34535676

RESUMO

Epithelial-to-mesenchymal transition (EMT) is fundamental to both normal tissue development and cancer progression. We hypothesized that EMT plasticity defines a range of metabolic phenotypes and that individual breast epithelial metabolic phenotypes are likely to fall within this phenotypic landscape. To determine EMT metabolic phenotypes, the metabolism of EMT was described within genome-scale metabolic models (GSMMs) using either transcriptomic or proteomic data from the breast epithelial EMT cell culture model D492. The ability of the different data types to describe breast epithelial metabolism was assessed using constraint-based modeling which was subsequently verified using 13C isotope tracer analysis. The application of proteomic data to GSMMs provided relatively higher accuracy in flux predictions compared to the transcriptomic data. Furthermore, the proteomic GSMMs predicted altered cholesterol metabolism and increased dependency on argininosuccinate lyase (ASL) following EMT which were confirmed in vitro using drug assays and siRNA knockdown experiments. The successful verification of the proteomic GSMMs afforded iBreast2886, a breast GSMM that encompasses the metabolic plasticity of EMT as defined by the D492 EMT cell culture model. Analysis of breast tumor proteomic data using iBreast2886 identified vulnerabilities within arginine metabolism that allowed prognostic discrimination of breast cancer patients on a subtype-specific level. Taken together, we demonstrate that the metabolic reconstruction iBreast2886 formalizes the metabolism of breast epithelial cell development and can be utilized as a tool for the functional interpretation of high throughput clinical data.


Assuntos
Neoplasias da Mama , Proteômica , Argininossuccinato Liase/genética , Neoplasias da Mama/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Genoma , Humanos
10.
Biomolecules ; 11(8)2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34439753

RESUMO

There is a pressing need for molecular targets and biomarkers in gastric cancer (GC). We aimed at identifying aberrations in L-arginine metabolism with therapeutic and diagnostic potential. Systemic metabolites were quantified using mass spectrometry in 293 individuals and enzymes' gene expression was quantified in 29 paired tumor-normal samples using qPCR and referred to cancer pathology and molecular landscape. Patients with cancer or benign disorders had reduced systemic arginine, citrulline, and ornithine and elevated symmetric dimethylarginine and dimethylamine. Citrulline and ornithine depletion was accentuated in metastasizing cancers. Metabolite diagnostic panel had 91% accuracy in detecting cancer and 70% accuracy in differentiating cancer from benign disorders. Gastric tumors had upregulated NOS2 and downregulated ASL, PRMT2, ORNT1, and DDAH1 expression. NOS2 upregulation was less and ASL downregulation was more pronounced in metastatic cancers. Tumor ASL and PRMT2 expression was inversely related to local advancement. Enzyme up- or downregulation was greater or significant solely in cardia subtype. Metabolic reprogramming in GC includes aberrant L-arginine metabolism, reflecting GC subtype and pathology, and is manifested by altered interplay of its intermediates and enzymes. Exploiting L-arginine metabolic pathways for diagnostic and therapeutic purposes is warranted. Functional studies on ASL, PRMT2, and ORNT1 in GC are needed.


Assuntos
Arginina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Gástricas/metabolismo , Idoso , Argininossuccinato Liase/biossíntese , Diferenciação Celular , Citrulina/metabolismo , DNA Complementar/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Masculino , Espectrometria de Massas , Metabolômica , Pessoa de Meia-Idade , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Metástase Neoplásica , Óxido Nítrico Sintase Tipo II , Ornitina/metabolismo , Reação em Cadeia da Polimerase , Proteína-Arginina N-Metiltransferases/biossíntese , Reprodutibilidade dos Testes , Neoplasias Gástricas/tratamento farmacológico , Transcriptoma
11.
Hum Genet ; 140(10): 1471-1485, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34417872

RESUMO

Argininosuccinate lyase (ASL) is essential for the NO-dependent regulation of tyrosine hydroxylase (TH) and thus for catecholamine production. Using a conditional mouse model with loss of ASL in catecholamine neurons, we demonstrate that ASL is expressed in dopaminergic neurons in the substantia nigra pars compacta, including the ALDH1A1 + subpopulation that is pivotal for the pathogenesis of Parkinson disease (PD). Neuronal loss of ASL results in catecholamine deficiency, in accumulation and formation of tyrosine aggregates, in elevation of α-synuclein, and phenotypically in motor and cognitive deficits. NO supplementation rescues the formation of aggregates as well as the motor deficiencies. Our data point to a potential metabolic link between accumulations of tyrosine and seeding of pathological aggregates in neurons as initiators for the pathological processes involved in neurodegeneration. Hence, interventions in tyrosine metabolism via regulation of NO levels may be therapeutic beneficial for the treatment of catecholamine-related neurodegenerative disorders.


Assuntos
Família Aldeído Desidrogenase 1/genética , Família Aldeído Desidrogenase 1/metabolismo , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Neurônios Dopaminérgicos/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Fenótipo , Retinal Desidrogenase/genética , Retinal Desidrogenase/metabolismo
12.
Front Immunol ; 12: 653571, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054815

RESUMO

Macrophages are indispensable immune cells tasked at eliminating intracellular pathogens. Mycobacterium tuberculosis (Mtb), one of the most virulent intracellular bacterial pathogens known to man, infects and resides within macrophages. While macrophages can be provoked by extracellular stimuli to inhibit and kill Mtb bacilli, these host defense mechanisms can be blocked by limiting nutritional metabolites, such as amino acids. The amino acid L-arginine has been well described to enhance immune function, especially in the context of driving macrophage nitric oxide (NO) production in mice. In this study, we aimed to establish the necessity of L-arginine on anti-Mtb macrophage function independent of NO. Utilizing an in vitro system, we identified that macrophages relied on NO for only half of their L-arginine-mediated host defenses and this L-arginine-mediated defense in the absence of NO was associated with enhanced macrophage numbers and viability. Additionally, we observed macrophage glycolysis to be driven by both L-arginine and mechanistic target of rapamycin (mTOR), and inhibition of glycolysis or mTOR reduced macrophage control of Mtb as well as macrophage number and viability in the presence of L-arginine. Our data underscore L-arginine as an essential nutrient for macrophage function, not only by fueling anti-mycobacterial NO production, but also as a central regulator of macrophage metabolism and additional host defense mechanisms.


Assuntos
Arginina/metabolismo , Suplementos Nutricionais , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/dietoterapia , Animais , Arginina/administração & dosagem , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Argininossuccinato Sintase/genética , Argininossuccinato Sintase/metabolismo , Sobrevivência Celular , Modelos Animais de Doenças , Humanos , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo , Cultura Primária de Células , Células RAW 264.7 , Tuberculose/imunologia , Tuberculose/microbiologia
13.
Mol Genet Metab ; 133(2): 148-156, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846069

RESUMO

BACKGROUND: Urea cycle disorders (UCDs) are among the most common inborn errors of liver metabolism. As therapies for hyperammonemia associated with urea cycle dysfunction have improved, chronic complications, such as liver disease, have become increasingly apparent in individuals with UCDs. Liver disease in UCDs may be associated with hepatic inflammation, hepatic fibrosis, portal hypertension, liver cancer and even liver failure. However, except for monitoring serum aminotransferases, there are no clear guidelines for screening and/or monitoring individuals with UCDs for liver disease. Thus, we systematically evaluated the potential utility of several non-invasive biomarkers for liver fibrosis in UCDs. METHODS: We evaluated grey-scale ultrasonography, liver stiffness obtained from shear wave elastography (SWE), and various serum biomarkers for hepatic fibrosis and necroinflammation, in a cohort of 28 children and adults with various UCDs. RESULTS: Overall, we demonstrate a high burden of liver disease in our participants with 46% of participants having abnormal grey-scale ultrasound pattern of the liver parenchyma, and 52% of individuals having increased liver stiffness. The analysis of serum biomarkers revealed that 32% of participants had elevated FibroTest™ score, a marker for hepatic fibrosis, and 25% of participants had increased ActiTest™ score, a marker for necroinflammation. Interestingly, liver stiffness did not correlate with ultrasound appearance or FibroTest™. CONCLUSION: Overall, our results demonstrate the high overall burden of liver disease in UCDs and highlights the need for further studies exploring new tools for identifying and monitoring individuals with UCDs who are at risk for this complication. TRIAL REGISTRATION: This study has been registered in ClinicalTrials.gov (NCT03721367).


Assuntos
Argininossuccinato Liase/sangue , Doenças Genéticas Inatas/sangue , Cirrose Hepática/sangue , Hepatopatias/sangue , Distúrbios Congênitos do Ciclo da Ureia/sangue , Adolescente , Adulto , Biomarcadores/sangue , Criança , Pré-Escolar , Técnicas de Imagem por Elasticidade , Feminino , Doenças Genéticas Inatas/diagnóstico por imagem , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Humanos , Hiperamonemia/sangue , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Fígado/diagnóstico por imagem , Fígado/patologia , Cirrose Hepática/diagnóstico por imagem , Cirrose Hepática/genética , Cirrose Hepática/patologia , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/patologia , Masculino , Erros Inatos do Metabolismo/genética , Pessoa de Meia-Idade , Ultrassonografia , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/patologia , Adulto Jovem
14.
Am J Med Genet A ; 185(7): 2026-2036, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33851512

RESUMO

Urea cycle disorders (UCDs) are inherited metabolic diseases that lead to hyperammonemia with variable clinical manifestations. Using data from a nationwide study, we investigated the onset time, gene variants, clinical manifestations, and treatment of patients with UCDs in Japan. Of the 229 patients with UCDs diagnosed and/or treated between January 2000 and March 2018, identified gene variants and clinical information were available for 102 patients, including 62 patients with ornithine transcarbamylase (OTC) deficiency, 18 patients with carbamoyl phosphate synthetase 1 (CPS1) deficiency, 16 patients with argininosuccinate synthetase (ASS) deficiency, and 6 patients with argininosuccinate lyase (ASL) deficiency. A total of 13, 10, 4, and 5 variants in the OTC, CPS1, ASS, and ASL genes were respectively identified as novel variants, which were neither registered in ClinVar databases nor previously reported. The onset time and severity in patients with UCD could be predicted based on the identified gene variants in each patient from this nationwide study and previous studies. This genetic information may help in predicting the long-term outcome and determining specific treatment strategies such as liver transplantation in patients with UCDs.


Assuntos
Argininossuccinato Liase/genética , Argininossuccinato Sintase/genética , Carbamoil-Fosfato Sintase (Amônia)/genética , Ornitina Carbamoiltransferase/genética , Distúrbios Congênitos do Ciclo da Ureia/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Variação Genética/genética , Humanos , Hiperamonemia/enzimologia , Hiperamonemia/genética , Hiperamonemia/patologia , Lactente , Masculino , Doenças Metabólicas/enzimologia , Doenças Metabólicas/genética , Doenças Metabólicas/patologia , Distúrbios Congênitos do Ciclo da Ureia/enzimologia , Distúrbios Congênitos do Ciclo da Ureia/patologia , Adulto Jovem
15.
J Clin Invest ; 131(5)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33645546

RESUMO

The involvement of nitric oxide (NO) in preventing bone loss has long been hypothesized, but despite decades of research the mechanisms remain obscure. In this issue of the JCI, Jin et al. explored NO deficiency using human cell and mouse models that lacked argininosuccinate lyase (ASL), the enzyme involved in synthesizing arginine and NO production. Osteoblasts that did not express ASL produced less NO and failed to differentiate. Notably, in the context of Asl deficiency, heterozygous deletion of caveolin 1, which normally inhibits NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass. These experiments suggest that ASL regulates arginine synthesis in osteoblasts, which leads to enhanced NO production and increased glucose metabolism. After a period when research slowed, these studies, like the legendary phoenix, renew the exploration of NO in bone biology, and provide exciting translational potential.


Assuntos
Argininossuccinato Liase , Óxido Nítrico , Animais , Arginina , Modelos Animais de Doenças , Camundongos
16.
Nitric Oxide ; 108: 12-19, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33338599

RESUMO

Despite the saturating concentrations of intracellular l-arginine, nitric oxide (NO) production in endothelial cells (EC) can be stimulated by exogenous arginine. This phenomenon, termed the "arginine paradox" led to the discovery of an arginine recycling pathway in which l-citrulline is recycled to l-arginine by utilizing two important urea cycle enzymes argininosuccinate synthetase (ASS) and argininosuccinate lyase (ASL). Prior work has shown that ASL is present in a NO synthetic complex containing hsp90 and endothelial NO synthase (eNOS). However, it is unclear whether hsp90 forms functional complexes with ASS and ASL and if it is involved regulating their activity. Thus, elucidating the role of hsp90 in the arginine recycling pathway was the goal of this study. Our data indicate that both ASS and ASL are chaperoned by hsp90. Inhibiting hsp90 activity with geldanamycin (GA), decreased the activity of both ASS and ASL and decreased cellular l-arginine levels in bovine aortic endothelial cells (BAEC). hsp90 inhibition led to a time-dependent decrease in ASS and ASL protein, despite no changes in mRNA levels. We further linked this protein loss to a proteasome dependent degradation of ASS and ASL via the E3 ubiquitin ligase, C-terminus of Hsc70-interacting protein (CHIP) and the heat shock protein, hsp70. Transient over-expression of CHIP was sufficient to stimulate ASS and ASL degradation while the over-expression of CHIP mutant proteins identified both TPR- and U-box-domain as essential for ASS and ASL degradation. This study provides a novel insight into the molecular regulation l-arginine recycling in EC and implicates the proteasome pathway as a possible therapeutic target to stimulate NO signaling.


Assuntos
Arginina/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Argininossuccinato Liase/química , Argininossuccinato Liase/metabolismo , Argininossuccinato Sintase/química , Argininossuccinato Sintase/metabolismo , Bovinos , Células Endoteliais , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
17.
Respir Physiol Neurobiol ; 285: 103598, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33326865

RESUMO

Short-term hypoxic states can influence the health and life activities of lowlanders who travel shortly to high altitudes, in transitory situations, such as surgical ischemia-reperfusion (to one or several organs), and in some sporting activities, such as parachuting and extreme skiing, mountain rescue teams, regular commercial flight crews, in which the subject may not even notice the hypoxia. NO is an integral part of the human physiological response to hypoxia. Until recently, the urea cycle (UC) was only considered as an important mechanism for neutralizing ammonia. We are the first to reveal an interrelation in hypoxic states between the activities of NO-synthase and UC enzymes in male rats' liver, kidney and brain. In the presented work, we have shown that during short-term intermittent hypobaric hypoxia (IHH) all enzymes of UC play an important role in the maintenance of NO quantity. The results allow thinking that kidney and brain argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL) and liver ASS and ASL can be different isoenzymes. It is worth mentioning that the results have revealed new sides of l-arginine metabolism in a hypoxic state in male rats.


Assuntos
Encéfalo/enzimologia , Hipóxia/enzimologia , Rim/enzimologia , Fígado/enzimologia , Óxido Nítrico/metabolismo , Ureia/metabolismo , Animais , Argininossuccinato Liase/metabolismo , Argininossuccinato Sintase/metabolismo , Modelos Animais de Doenças , Masculino , Redes e Vias Metabólicas/fisiologia , Ratos , Ratos Wistar
18.
Acta Biochim Pol ; 67(3): 347-351, 2020 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-32931185

RESUMO

Argininosuccinic aciduria is an autosomal, recessive amino acid disorder that is caused by a deficiency of the argininosuccinate lyase enzyme. Citrulline is the most significant marker to detect this disorder. We used the High-performance liquid chromatography with fluorescence detection with 450 nm emission and 330 nm excitation wavelengths, 15 mmol/L potassium dihydrogen phosphate and 5 mmol/L dipotassium hydrogen phosphate as Mobile Phase A, and 50 mL water, 250 mL acetonitrile, and 200 mL methanol as Mobile Phase B in gradient mode with flow rate of 1.2 mL/min. The citrulline concentration was 22 µmol/L in healthy infants and 220 µmol/L in infants suffering from the disorder.


Assuntos
Aminoácidos/sangue , Aminoácidos/química , Acidúria Argininossuccínica/sangue , Acetonitrilas/química , Aminoácidos/classificação , Argininossuccinato Liase/metabolismo , Acidúria Argininossuccínica/diagnóstico , Acidúria Argininossuccínica/enzimologia , Biomarcadores/sangue , Biomarcadores/química , Estudos de Casos e Controles , Cromatografia Líquida de Alta Pressão/métodos , Citrulina/sangue , Citrulina/química , Humanos , Lactente , Recém-Nascido , Irã (Geográfico) , Metanol/química , Fosfatos/química , Compostos de Potássio/química , Espectrometria de Fluorescência/métodos , Água/química
19.
Anal Chem ; 92(17): 11505-11510, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32794704

RESUMO

We developed a simple and rapid method for analyzing nonproteinogenic amino acids that does not require conventional chromatographic equipment. In this technique, nonproteinogenic amino acids were first converted to a proteinogenic amino acid through in vitro metabolism in a cell extract. The proteinogenic amino acid generated from the nonproteinogenic precursors were then incorporated into a reporter protein using a cell-free protein synthesis system. The titers of the nonproteinogenic amino acids could be readily quantified by measuring the activity of reporter proteins. This method, which combines the enzymatic conversion of target amino acids with translational analysis, makes amino acid analysis more accessible while minimizing the cost and time requirements. We anticipate that the same strategy could be extended to the detection of diverse biochemical molecules with clinical and industrial implications.


Assuntos
Extratos Celulares/química , Citrulina/química , Ornitina/química , Proteínas/química , Sequência de Aminoácidos , Arginina/química , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Argininossuccinato Sintase/genética , Argininossuccinato Sintase/metabolismo , Carboxil e Carbamoil Transferases/genética , Carboxil e Carbamoil Transferases/metabolismo , Citrulina/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Ornitina/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica , Estereoisomerismo , Especificidade por Substrato
20.
Mol Genet Genomic Med ; 8(7): e1301, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32410394

RESUMO

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.


Assuntos
Argininossuccinato Liase/genética , Acidúria Argininossuccínica/genética , Mutação , Doença da Deficiência de Ornitina Carbomoiltransferase/genética , Ornitina Carbamoiltransferase/genética , Argininossuccinato Liase/química , Acidúria Argininossuccínica/patologia , Feminino , Humanos , Lactente , Masculino , Simulação de Dinâmica Molecular , Ornitina Carbamoiltransferase/química , Doença da Deficiência de Ornitina Carbomoiltransferase/patologia , Linhagem , Domínios Proteicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...