Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Clin Chem ; 70(6): 865-877, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38597162

RESUMO

BACKGROUND: Mucopolysaccharidosis (MPS) and glycoproteinosis are 2 groups of heterogenous lysosomal storage disorders (LSDs) caused by defective degradation of glycosaminoglycans (GAGs) and glycoproteins, respectively. Oligosaccharides and glycoamino acids have been recognized as biomarkers for MPS and glycoproteinosis. Given that both groups of LSDs have overlapping clinical features, a multiplexed assay capable of unambiguous subtyping is desired for accurate diagnosis, and potentially for severity stratification and treatment monitoring. METHODS: Urinary oligosaccharides were derivatized with 3-methyl-1-phenyl-2-pyrazoline-5-one (PMP) and analyzed by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) together with the underivatized glycoamino acids. Novel biomarkers were identified with a semi-targeted approach with precursor mass scanning, the fragmentation pattern (if applicable), and the biochemical basis of the condition. RESULTS: A UPLC-MS/MS analysis with improved chromatographic separation was developed. Novel biomarkers for MPS-IIIA, IIIB, IIIC, and VII were identified and validated. A total of 28 oligosaccharides, 2 glycoamino acids, and 2 ratios were selected as key diagnostic biomarkers. Validation studies including linearity, lower limit of quantitation (LLOQ), and precision were carried out with the assay performance meeting the required criteria. Age-specific reference ranges were collected. In the 76 untreated patients, unambiguous diagnosis was achieved with 100% sensitivity and specificity. Additionally, the levels of disease-specific biomarkers were substantially reduced in the treated patients. CONCLUSIONS: A multiplexed UPLC-MS/MS assay for urinary oligosaccharides and glycoamino acids measurement was developed and validated. The assay is suitable for the accurate diagnosis and subtyping of MPS and glycoproteinosis, and potentially for severity stratification and monitoring response to treatment.


Assuntos
Biomarcadores , Glicoproteínas , Mucopolissacaridoses , Oligossacarídeos , Espectrometria de Massas em Tandem , Humanos , Espectrometria de Massas em Tandem/métodos , Oligossacarídeos/urina , Criança , Cromatografia Líquida de Alta Pressão/métodos , Pré-Escolar , Biomarcadores/urina , Mucopolissacaridoses/urina , Mucopolissacaridoses/diagnóstico , Adolescente , Glicoproteínas/urina , Lactente , Masculino , Feminino , Adulto , Aminoácidos/urina , Adulto Jovem
2.
Mol Genet Metab ; 141(1): 108105, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38128203

RESUMO

Previously we developed a multiplex liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay using dried blood spots for all subtypes of mucopolysaccharidoses (MPS) except MPS-IIID. Here we show that the MPS-IIID enzyme N-acetylglucosamine-6-sulfatase (GNS) is inhibited in dried blood spot (DBS) extracts, but activity can be recovered if the extract is diluted to reduce the concentrations of endogenous inhibitors. The new GNS assay displays acceptable characteristics including linearity in product formation with incubation time and amount of enzyme, low variability, and ability to distinguish MPS-IIID-affected from healthy patients using DBS. The assay can be added to the LC-MS/MS multiplex panel for all MPS subtypes requiring ∼2 min per newborn for the LC-MS/MS run.


Assuntos
Mucopolissacaridoses , Mucopolissacaridose VI , Recém-Nascido , Humanos , Espectrometria de Massas em Tandem/métodos , Cromatografia Líquida , Sulfatases , Teste em Amostras de Sangue Seco/métodos
3.
Mol Genet Metab ; 142(4): 108517, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38908075

RESUMO

GM2 gangliosidosis is a group of rare lysosomal storage disorders (LSDs) including Tay-Sachs disease (TSD) and Sandhoff disease (SD), caused by deficiency in activity of either ß-hexosaminidase A (HexA) or both ß-hexosaminidase A and ß-hexosaminidase B (HexB). Methods for screening and diagnosis of TSD and SD include measurement and comparison of the activity of these two enzymes. Here we report a novel method for duplex screening of dried blood spots (DBS) for TSD and SD by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The method requires incubation of a single 3 mm DBS punch with the assay cocktail followed by the injection into the LC-MS/MS. The performance of the method was evaluated by comparing the confirmed TSD and SD patient DBS to random healthy newborn DBS which showed easy discrimination between the three cohorts. The method is multiplexable with other LSD MS/MS enzyme assays which is critical to the continued expansion of the NBS panels.


Assuntos
Teste em Amostras de Sangue Seco , Triagem Neonatal , Doença de Sandhoff , Espectrometria de Massas em Tandem , Doença de Tay-Sachs , Humanos , Doença de Tay-Sachs/diagnóstico , Doença de Tay-Sachs/sangue , Doença de Tay-Sachs/enzimologia , Recém-Nascido , Espectrometria de Massas em Tandem/métodos , Triagem Neonatal/métodos , Teste em Amostras de Sangue Seco/métodos , Doença de Sandhoff/diagnóstico , Doença de Sandhoff/sangue , Cromatografia Líquida/métodos , Ensaios Enzimáticos/métodos , Cadeia alfa da beta-Hexosaminidase/sangue , Hexosaminidase A/sangue , Hexosaminidase B/sangue
4.
Mol Genet Metab ; 142(2): 108489, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38718669

RESUMO

BACKGROUND: Canavan disease is a devastating neurometabolic disorder caused by accumulation of N acetylaspartate in brain and body fluids due to genetic defects in the aspartoacylase gene (ASPA). New gene therapies are on the horizon but will require early presymptomatic diagnosis to be fully effective. METHODS: We therefore developed a fast and highly sensitive liquid chromatography mass spectrometry (LC-MS/MS)-based method for quantification of N-acetylaspartate in dried blood spots and established reference ranges for neonates and older controls. With this test, we investigated 45 samples of 25 Canavan patients including 8 with a neonatal sample. RESULTS: Measuring N-acetylaspartate concentration in dried blood with this novel test, all Canavan patients (with variable severity) were well separated from the control group (median; range: 5.7; 1.6-13.6 µmol/L [n = 45] vs 0.44; 0.24-0.99 µmol/L [n = 59] (p < 0.05)). There was also no overlap when comparing neonatal samples of Canavan patients (7.3; 5.1-9.9 µmol/L [n = 8]) and neonatal controls (0.93; 0.4-1.8 µmol/L [n = 784]) (p < 0.05). CONCLUSIONS: We have developed a new LC-MS/MS-based screening test for early postnatal diagnosis of Canavan disease that should be further evaluated in a population-based study once a promising treatment becomes available. The method meets the general requirements of newborn screening and should be appropriate for multiplexing with other screening approaches that combine chromatographic and mass spectrometry techniques.


Assuntos
Ácido Aspártico , Doença de Canavan , Teste em Amostras de Sangue Seco , Triagem Neonatal , Espectrometria de Massas em Tandem , Humanos , Doença de Canavan/diagnóstico , Doença de Canavan/sangue , Doença de Canavan/genética , Recém-Nascido , Triagem Neonatal/métodos , Teste em Amostras de Sangue Seco/métodos , Espectrometria de Massas em Tandem/métodos , Ácido Aspártico/análogos & derivados , Ácido Aspártico/sangue , Cromatografia Líquida , Feminino , Masculino , Lactente , Pré-Escolar , Espectrometria de Massa com Cromatografia Líquida , Amidoidrolases
5.
Mol Genet Metab ; 142(1): 108436, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38552449

RESUMO

Newborn screening (NBS) for metachromatic leukodystrophy (MLD) is based on first-tier measurement of sulfatides in dried blood spots (DBS) followed by second-tier measurement of arylsulfatase A in the same DBS. This approach is very precise with 0-1 false positives per ∼30,000 newborns tested. Recent data reported here shows that the sulfatide molecular species with an α-hydroxyl, 16­carbon, mono-unsaturated fatty acyl group (16:1-OH-sulfatide) is superior to the original biomarker 16:0-sulfatide in reducing the number of first-tier false positives. This result is consistent across 4 MLD NBS centers. By measuring 16:1-OH-sulfatide alone or together with 16:0-sulfatide, the estimated false positive rate is 0.048% and is reduced essentially to zero with second-tier arylsulfatase A activity assay. The false negative rate is predicted to be extremely low based on the demonstration that 40 out of 40 newborn DBS from clinically-confirmed MLD patients are detected with these methods. The work shows that NBS for MLD is extremely precise and ready for deployment. Furthermore, it can be multiplexed with several other inborn errors of metabolism already tested in NBS centers worldwide.


Assuntos
Cerebrosídeo Sulfatase , Teste em Amostras de Sangue Seco , Leucodistrofia Metacromática , Triagem Neonatal , Sulfoglicoesfingolipídeos , Humanos , Leucodistrofia Metacromática/diagnóstico , Leucodistrofia Metacromática/sangue , Recém-Nascido , Sulfoglicoesfingolipídeos/sangue , Triagem Neonatal/métodos , Cerebrosídeo Sulfatase/sangue , Cerebrosídeo Sulfatase/genética , Teste em Amostras de Sangue Seco/métodos , Reações Falso-Positivas , Biomarcadores/sangue
6.
Mol Genet Metab ; 141(2): 108116, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38161139

RESUMO

Multiple sulfatase deficiency (MSD) is an ultra-rare, inherited lysosomal storage disease caused by mutations in the gene sulfatase modifying factor 1 (SUMF1). MSD is characterized by the functional deficiency of all sulfatase enzymes, leading to the storage of sulfated substrates including glycosaminoglycans (GAGs), sulfolipids, and steroid sulfates. Patients with MSD experience severe neurological impairment, hearing loss, organomegaly, corneal clouding, cardiac valve disease, dysostosis multiplex, contractures, and ichthyosis. Here, we generated a novel human model of MSD by reprogramming patient peripheral blood mononuclear cells to establish an MSD induced pluripotent stem cell (iPSC) line (SUMF1 p.A279V). We also generated an isogenic control iPSC line by correcting the pathogenic variant with CRISPR/Cas9 gene editing. We successfully differentiated these iPSC lines into neural progenitor cells (NPCs) and NGN2-induced neurons (NGN2-iN) to model the neuropathology of MSD. Mature neuronal cells exhibited decreased SUMF1 gene expression, increased lysosomal stress, impaired neurite outgrowth and maturation, reduced sulfatase activities, and GAG accumulation. Interestingly, MSD iPSCs and NPCs did not exhibit as severe of phenotypes, suggesting that as neurons differentiate and mature, they become more vulnerable to loss of SUMF1. In summary, we demonstrate that this human iPSC-derived neuronal model recapitulates the cellular and biochemical features of MSD. These cell models can be used as tools to further elucidate the mechanisms of MSD pathology and for the development of therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença da Deficiência de Múltiplas Sulfatases , Humanos , Leucócitos Mononucleares/metabolismo , Neurônios/patologia , Sulfatases , Oxirredutases atuantes sobre Doadores de Grupo Enxofre
8.
Mol Genet Metab Rep ; 38: 101037, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38173711

RESUMO

The increasing availability of novel therapies highlights the importance of screening newborns for rare genetic disorders so that they may benefit from early therapy, when it is most likely to be effective. Pilot newborn screening (NBS) studies are a way to gather objective evidence about the feasibility and utility of screening, the accuracy of screening assays, and the incidence of disease. They are also an optimal way to evaluate the complex ethical, legal and social implications (ELSI) that accompany NBS expansion for disorders. ScreenPlus is a consented pilot NBS program that aims to enroll over 100,000 infants across New York City. The initial ScreenPlus panel includes 14 disorders and uses an analyte-based, multi-tiered screening platform in an effort to enhance screening accuracy. Infants who receive an abnormal result are referred to a ScreenPlus provider for confirmatory testing, management, and therapy as needed, along with longitudinal capture of outcome data. Participation in ScreenPlus requires parental consent, which is obtained in active and passive manners. Patient-facing documents are translated into the ten most common languages spoken at our nine pilot hospitals, all of which serve diverse communities. At the time of consent, parents are invited to receive a series of online surveys to capture their opinions about specific ELSI-related topics, such as NBS policy, residual dried blood spot retention, and the types of disorders that should be on NBS panels. ScreenPlus has developed a stakeholder-based, collective funding model that includes federal support in addition to funding from 14 advocacy and industry sponsors, all of which have a particular interest in NBS for at least one of the ScreenPlus disorders. Taken together, ScreenPlus is a model, multi-sponsored pilot NBS program that will provide critical data about NBS for a broad panel of disorders, while gathering key stakeholder opinions to help guide ethically sensitive decision-making about NBS expansion.

9.
bioRxiv ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39071356

RESUMO

A general approach to design proteins that bind tightly and specifically to intrinsically disordered regions (IDRs) of proteins and flexible peptides would have wide application in biological research, therapeutics, and diagnosis. However, the lack of defined structures and the high variability in sequence and conformational preferences has complicated such efforts. We sought to develop a method combining biophysical principles with deep learning to readily generate binders for any disordered sequence. Instead of assuming a fixed regular structure for the target, general recognition is achieved by threading the query sequence through diverse extended binding modes in hundreds of templates with varying pocket depths and spacings, followed by RFdiffusion refinement to optimize the binder-target fit. We tested the method by designing binders to 39 highly diverse unstructured targets. Experimental testing of ∼36 designs per target yielded binders with affinities better than 100 nM in 34 cases, and in the pM range in four cases. The co-crystal structure of a designed binder in complex with dynorphin A is closely consistent with the design model. All by all binding experiments for 20 designs binding diverse targets show they are highly specific for the intended targets, with no crosstalk even for the closely related dynorphin A and dynorphin B. Our approach thus could provide a general solution to the intrinsically disordered protein and peptide recognition problem.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA