Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 310
Filtrar
1.
Matrix Biol ; 131: 1-16, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38750698

RESUMEN

Extracellular matrix remodeling mechanisms are understudied in cardiac development and congenital heart defects. We show that matrix-degrading metalloproteases ADAMTS1 and ADAMTS5, are extensively co-expressed during mouse cardiac development. The mouse mutants of each gene have mild cardiac anomalies, however, their combined genetic inactivation to elicit cooperative roles is precluded by tight gene linkage. Therefore, we coupled Adamts1 inactivation with pharmacologic ADAMTS5 blockade to uncover stage-specific cooperative roles and investigated their potential substrates in mouse cardiac development. ADAMTS5 blockade was achieved in Adamts1 null mouse embryos using an activity-blocking monoclonal antibody during distinct developmental windows spanning myocardial compaction or cardiac septation and outflow tract rotation. Synchrotron imaging, RNA in situ hybridization, immunofluorescence microscopy and electron microscopy were used to determine the impact on cardiac development and compared to Gpc6 and ADAMTS-cleavage resistant versican mutants. Mass spectrometry-based N-terminomics was used to seek relevant substrates. Combined inactivation of ADAMTS1 and ADAMTS5 prior to 12.5 days of gestation led to dramatic accumulation of versican-rich cardiac jelly and inhibited formation of compact and trabecular myocardium, which was also observed in mice with ADAMTS cleavage-resistant versican. Combined inactivation after 12.5 days impaired outflow tract development and ventricular septal closure, generating a tetralogy of Fallot-like defect. N-terminomics of combined ADAMTS knockout and control hearts identified a cleaved glypican-6 peptide only in the controls. ADAMTS1 and ADAMTS5 expression in cells was associated with specific glypican-6 cleavages. Paradoxically, combined ADAMTS1 and ADAMTS5 inactivation reduced cardiac glypican-6 and outflow tract Gpc6 transcription. Notably, Gpc6-/- hearts demonstrated similar rotational defects as combined ADAMTS inactivated hearts and both had reduced hedgehog signaling. Thus, versican proteolysis in cardiac jelly at the canonical Glu441-Ala442 site is cooperatively mediated by ADAMTS1 and ADAMTS5 and required for proper ventricular cardiomyogenesis, whereas, reduced glypican-6 after combined ADAMTS inactivation impairs hedgehog signaling, leading to outflow tract malrotation.


Asunto(s)
Proteína ADAMTS1 , Proteína ADAMTS5 , Glipicanos , Corazón , Proteolisis , Versicanos , Animales , Ratones , Versicanos/metabolismo , Versicanos/genética , Proteína ADAMTS5/metabolismo , Proteína ADAMTS5/genética , Proteína ADAMTS1/metabolismo , Proteína ADAMTS1/genética , Glipicanos/metabolismo , Glipicanos/genética , Corazón/crecimiento & desarrollo , Ratones Noqueados , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología
2.
Cell Death Dis ; 15(1): 82, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38263290

RESUMEN

The matrix metalloprotease A disintegrin and metalloprotease with thrombospondin motifs 1 (ADAMTS1) was reported to be involved in tumor progression in several cancer types, but its contributions appear discrepant. At present, the role of ADAMTS1 in oral squamous cell carcinoma (SCC; OSCC) remains unclear. Herein, The Cancer Genome Atlas (TCGA) database showed that ADAMTS1 transcripts were downregulated in head and neck SCC (HNSCC) tissues compared to normal tissues, but ADAMTS1 levels were correlated with poorer prognoses of HNSCC patients. In vitro, we observed that ADAMTS1 expression levels were correlated with the invasive abilities of four OSCC cell lines, HSC-3, SCC9, HSC-3M, and SAS. Knockdown of ADAMTS1 in OSCC cells led to a decrease and its overexpression led to an increase in cell-invasive abilities in vitro as well as tumor growth and lymph node (LN) metastasis in OSCC xenografts. Mechanistic investigations showed that the cyclic increase in ADAMTS1-L1 cell adhesion molecule (L1CAM) axis-mediated epidermal growth factor receptor (EGFR) activation led to exacerbation of the invasive abilities of OSCC cells via inducing epithelial-mesenchymal transition (EMT) progression. Clinical analyses revealed that ADAMTS1, L1CAM, and EGFR levels were all correlated with worse prognoses of HNSCC patients, and patients with ADAMTS1high/L1CAMhigh or EGFRhigh tumors had the shortest overall and disease-specific survival times. As to therapeutic aspects, we discovered that an edible plant-derived flavonoid, apigenin (API), drastically inhibited expression of the ADAMTS1-L1CAM-EGFR axis and reduced the ADAMTS1-triggered invasion and LN metastasis of OSCC cells in vitro and in vivo. Most importantly, API treatment significantly prolonged survival rates of xenograft mice with OSCC. In summary, ADAMTS1 may be a useful biomarker for predicting OSCC progression, and API potentially retarded OSCC progression by targeting the ADAMTS1-L1CAM-EGFR signaling pathway.


Asunto(s)
Proteína ADAMTS1 , Receptores ErbB , Neoplasias de la Boca , Molécula L1 de Adhesión de Célula Nerviosa , Carcinoma de Células Escamosas de Cabeza y Cuello , Animales , Humanos , Ratones , Apigenina , Transición Epitelial-Mesenquimal , Metástasis Linfática
3.
Exp Cell Res ; 433(2): 113852, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37951335

RESUMEN

In the study of tumorigenesis, the involvement of molecules within the extracellular matrix (ECM) is crucial. ADAMTSs (A Disintegrin and Metalloproteinase with Thrombospondin motifs), a group of secreted proteases known for their role in ECM remodeling, were primarily considered to be extracellular proteases. However, our research specifically detected ADAMTS-1, a member of this family, predominantly within the nucleus of mammary cells. Our main objective was to understand the mechanism of ADAMTS-1 translocation to the nucleus and its functional significance in this cellular compartment. Our investigation uncovered that nuclear ADAMTS-1 was present in cells exhibiting an epithelial phenotype, while cells of mesenchymal origin contained the protease in the cytoplasm. Moreover, disruption of ADAMTS-1 secretion, induced by Monensin treatment, resulted in its accumulation in the cytoplasm. Notably, our research indicated that alterations in the secretory pathways could influence the protease's compartmentalization. Additionally, experiments with conditioned medium from cells containing nuclear ADAMTS-1 demonstrated its internalization into the nucleus by HT-1080 cells and fibroblasts. Furthermore, heightened levels of ADAMTS-1 within the ECM reduced the migratory potential of mesenchymal cells. This highlights the potential significance of nuclear ADAMTS-1 as a critical component within the tumor microenvironment due to its functional activity in this specific cellular compartment.


Asunto(s)
Proteína ADAMTS1 , Movimiento Celular , Núcleo Celular , Matriz Extracelular , Trombospondinas , Humanos , Proteína ADAMTS1/genética , Proteína ADAMTS1/metabolismo , Carcinogénesis/metabolismo , Endopeptidasas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Trombospondinas/metabolismo , Microambiente Tumoral , Núcleo Celular/metabolismo
4.
Circ Res ; 133(1): 48-67, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37254794

RESUMEN

BACKGROUND: Left ventricular noncompaction (LVNC) is a prevalent cardiomyopathy associated with excessive trabeculation and thin compact myocardium. Patients with LVNC are vulnerable to cardiac dysfunction and at high risk of sudden death. Although sporadic and inherited mutations in cardiac genes are implicated in LVNC, understanding of the mechanisms responsible for human LVNC is limited. METHODS: We screened the complete exome sequence database of the Pediatrics Cardiac Genomics Consortium and identified a cohort with a de novo CHD4 (chromodomain helicase DNA-binding protein 4) proband, CHD4M202I, with congenital heart defects. We engineered a humanized mouse model of CHD4M202I (mouse CHD4M195I). Histological analysis, immunohistochemistry, flow cytometry, transmission electron microscopy, and echocardiography were used to analyze cardiac anatomy and function. Ex vivo culture, immunopurification coupled with mass spectrometry, transcriptional profiling, and chromatin immunoprecipitation were performed to deduce the mechanism of CHD4M195I-mediated ventricular wall defects. RESULTS: CHD4M195I/M195I mice developed biventricular hypertrabeculation and noncompaction and died at birth. Proliferation of cardiomyocytes was significantly increased in CHD4M195I hearts, and the excessive trabeculation was associated with accumulation of ECM (extracellular matrix) proteins and a reduction of ADAMTS1 (ADAM metallopeptidase with thrombospondin type 1 motif 1), an ECM protease. We rescued the hyperproliferation and hypertrabeculation defects in CHD4M195I hearts by administration of ADAMTS1. Mechanistically, the CHD4M195I protein showed augmented affinity to endocardial BRG1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4). This enhanced affinity resulted in the failure of derepression of Adamts1 transcription such that ADAMTS1-mediated trabeculation termination was impaired. CONCLUSIONS: Our study reveals how a single mutation in the chromatin remodeler CHD4, in mice or humans, modulates ventricular chamber maturation and that cardiac defects associated with the missense mutation CHD4M195I can be attenuated by the administration of ADAMTS1.


Asunto(s)
No Compactación Aislada del Miocardio Ventricular , Mutación Missense , Humanos , Animales , Niño , Ratones , Ventrículos Cardíacos , Causalidad , Mutación , Miocitos Cardíacos , Cromatina , No Compactación Aislada del Miocardio Ventricular/genética , Proteína ADAMTS1/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética
5.
Aging (Albany NY) ; 15(6): 2097-2114, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36947712

RESUMEN

Non-small cell lung cancer (NSCLC) accounts for approximately 80% of all lung cancers. Identifying key molecular targets related to the initiation, development, and metastasis of lung cancer is important for its diagnosis and target therapy. The ADAMTS families of multidomain extracellular protease enzymes have been reported to be involved in many physiological processes. In this study, we found that ADAMTS1 was highly expressed in NSCLC tissues, which promoted cell proliferation, migration, invasion, and epithelial to mesenchymal transition (EMT) of NSCLC cells. In the NSCLC tumor metastasis model involving nude mice, overexpression of ADAMTS1 promoted EMT and lung metastasis of tumor cells. Moreover, ADAMTS1 positively regulated TGF-ß expression, and TGF-ß was highly expressed in NSCLC tumor tissues. si-TGF-ß or inhibition of TGF-ß expression through the short peptide KTFR on ADAMTS1 protein could reverse the oncogenic effects of ADAMTS1 on lung cancer cells. Taken together, ADAMTS1 functioned as an oncogene in NSCLC cells by promoting TGF-ß expression, indicating that ADAMTS1 has important regulatory roles in the progression of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Animales , Ratones , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Transición Epitelial-Mesenquimal/genética , Factor de Crecimiento Transformador beta/metabolismo , Ratones Desnudos , Proteína ADAMTS1/genética , Proteína ADAMTS1/metabolismo , MicroARNs/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica
6.
J Biol Chem ; 299(4): 103048, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36813235

RESUMEN

A disintegrin-like and metalloproteinase with thrombospondin type 1 motifs (ADAMTS1) is a protease involved in fertilization, cancer, cardiovascular development, and thoracic aneurysms. Proteoglycans such as versican and aggrecan have been identified as ADAMTS1 substrates, and Adamts1 ablation in mice typically results in versican accumulation; however, previous qualitative studies have suggested that ADAMTS1 proteoglycanase activity is weaker than that of other family members such as ADAMTS4 and ADAMTS5. Here, we investigated the functional determinants of ADAMTS1 proteoglycanase activity. We found that ADAMTS1 versicanase activity is approximately 1000-fold lower than ADAMTS5 and 50-fold lower than ADAMTS4 with a kinetic constant (kcat/Km) of 3.6 × 103 M-1 s-1 against full-length versican. Studies on domain-deletion variants identified the spacer and cysteine-rich domains as major determinants of ADAMTS1 versicanase activity. Additionally, we confirmed that these C-terminal domains are involved in the proteolysis of aggrecan as well as biglycan, a small leucine-rich proteoglycan. Glutamine scanning mutagenesis of exposed positively charged residues on the spacer domain loops and loop substitution with ADAMTS4 identified clusters of substrate-binding residues (exosites) in ß3-ß4 (R756Q/R759Q/R762Q), ß9-ß10 (residues 828-835), and ß6-ß7 (K795Q) loops. This study provides a mechanistic foundation for understanding the interactions between ADAMTS1 and its proteoglycan substrates and paves the way for development of selective exosite modulators of ADAMTS1 proteoglycanase activity.


Asunto(s)
Proteína ADAMTS1 , Animales , Ratones , Proteína ADAMTS1/química , Proteína ADAMTS1/metabolismo , Proteína ADAMTS4/metabolismo , Proteína ADAMTS5/metabolismo , Agrecanos/metabolismo , Versicanos/metabolismo
7.
Int J Clin Oncol ; 28(1): 52-68, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36371587

RESUMEN

BACKGROUND: Glioma is the most common intracranial malignancy in adults with a high degree of malignancy and poor prognosis, which is largely attributed to the existence of glioma stem cells (GSCs). Previous evidence indicated that the matrix metalloproteinase ADAMTS1 was implicated in the process of tumor invasion, but the involvement of ADAMTS1 in glioma malignant invasion remains poorly understood. METHODS: The expression and prognosis values of ADAMTS1 were investigated in patients with glioma based on ONCOMINE and GEPIA databases. ADAMTS1 expression of different malignancy grade tissues was determined by immunohistochemistry. The effects of ADAMTS1 on cell proliferation and invasion were determined by clone formation assay and Transwell migration assay. The animal experiment was performed in an intracranial orthotopic xenograft model by knockout of ADAMTS1. Stemness properties and Notch1-SOX2 pathway were examined in stable ADAMTS1 knockdown GSCs. RESULTS: The expression levels of ADAMTS1 were significantly higher in glioma tissues and significantly correlated with the grade of malignancy and prognosis of glioma. Elevated ADAMTS1 expression was associated with SOX2, N-cadherin and the resistance of chemoradiotherapy of glioma patients. ADAMTS1 knockout suppressed the intracranial orthotopic xenograft growth and prolonged the survival of xenograft mice in vivo. Mechanistically, we found a blockade of the migration and invasiveness of GSCs and the expression levels of Notch1 and SOX2 in absence of ADAMTS1. CONCLUSION: As a biomarker for prediction of prognosis, ADAMTS1 may affect the invasive phenotype of GSCs by regulating Notch1-SOX2 signaling pathway, thereby promoting the invasive growth of glioma.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Ratones , Animales , Pronóstico , Línea Celular Tumoral , Glioma/patología , Neoplasias Encefálicas/patología , Transducción de Señal , Proliferación Celular/genética , Células Madre Neoplásicas/patología , Regulación Neoplásica de la Expresión Génica , Proteína ADAMTS1/genética , Proteína ADAMTS1/metabolismo
8.
BMC Pregnancy Childbirth ; 22(1): 499, 2022 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-35725415

RESUMEN

BACKGROUND: We aimed to investigate the levels of ADAMTS-1, which is secreted from the extracellular matrix during trophoblastic invasion in hyperemesis gravidarum (HEG). METHODS: In this cross-sectional study, we compared 45 HEG patients aged between 21 and 34 in terms of ADAMTS-1 levels with a control group consisting of 44 healthy pregnant women. The demographic characteristics and several laboratory parameters of the patients were recorded. Both groups were also compared in terms of ketonuria. We evaluated the correlation between ADAMTS-1 levels and ketonuria. RESULTS: The 2 groups were matched in terms of age, gestational age, gravidity, parity, and body mass index. Some inflammatory markers, such as neutrophil count, MPV, PDW, and PCT levels, were significantly higher in the HEG groups compared to the control group (all p < 0.05). However, mean MCV and serum TSH levels were statistically significantly lower in this group (both p < 0.001). ADAMTS-1 levels were 12.6 ± 1.4 ng/ml in the HEG group and 6.2 ± 1.6 ng/ml in the control group (p < 0.001). It was significantly and positively correlated with urine ketone, neutrophil count, and PDW, whereas negatively correlated with MCV and TSH value in the HEG group. ROC analysis showed that a threshold value of 11.275 ng/ml for ADAMTS-1 predicted HEG patients with a sensitivity of 60% and specificity of 95.5%. CONCLUSION: ADAMTS-1 serum levels are increased in HEG patients, and there is a positive correlation between ADAMTS-1 levels and ketonuria.


Asunto(s)
Proteína ADAMTS1 , Hiperemesis Gravídica , Cetosis , Proteína ADAMTS1/sangre , Adulto , Estudios Transversales , Femenino , Humanos , Recuento de Leucocitos , Embarazo , Tirotropina , Adulto Joven
9.
Eur J Hum Genet ; 30(1): 22-33, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34135477

RESUMEN

Hearing impairment (HI) is a common disorder of sensorineural function with a highly heterogeneous genetic background. Although substantial progress has been made in the understanding of the genetic etiology of hereditary HI, many genes implicated in HI remain undiscovered. Via exome and Sanger sequencing of DNA samples obtained from consanguineous Pakistani families that segregate profound prelingual sensorineural HI, we identified rare homozygous missense variants in four genes (ADAMTS1, MPDZ, MVD, and SEZ6) that are likely the underlying cause of HI. Linkage analysis provided statistical evidence that these variants are associated with autosomal recessive nonsyndromic HI. In silico analysis of the mutant proteins encoded by these genes predicted structural, conformational or interaction changes. RNAseq data analysis revealed expression of these genes in the sensory epithelium of the mouse inner ear during embryonic, postnatal, and adult stages. Immunohistochemistry of the mouse cochlear tissue, further confirmed the expression of ADAMTS1, SEZ6, and MPDZ in the neurosensory hair cells of the organ of Corti, while MVD expression was more prominent in the spiral ganglion cells. Overall, supported by in silico mutant protein analysis, animal models, linkage analysis, and spatiotemporal expression profiling in the mouse inner ear, we propose four new candidate genes for HI and expand our understanding of the etiology of HI.


Asunto(s)
Proteína ADAMTS1/genética , Carboxiliasas/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Proteína ADAMTS1/química , Proteína ADAMTS1/metabolismo , Animales , Carboxiliasas/química , Carboxiliasas/metabolismo , Femenino , Genes Recesivos , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Linaje , Dominios Proteicos
10.
Eur J Pharmacol ; 914: 174681, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34871556

RESUMEN

Secreted protein acidic and rich in cysteine (SPARC), an extracellular matrix (ECM) protein, was recently shown to induce collagen deposition through the production of a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS1) in the aging heart. ADAMTS1 regulates ECM turnover by degrading ECM components, and its excessive activation contributes to various pathological states, including fibrosis. The present study investigated the pathophysiological regulation and role of SPARC and ADAMTS1 in renal fibrosis using uninephrectomized rats treated with deoxycorticosterone acetate (DOCA, 40 mg/kg/week, subcutaneously) and salt (1% in drinking water). The administration of DOCA and salt gradually and significantly elevated systolic blood pressure during the 3-week treatment period, induced proteinuria, decreased creatinine clearance, and increased NADPH oxidase-derived superoxide production, malondialdehyde concentrations, and monocyte chemoattractant protein-1 and osteopontin expression in the kidneys. Glomerulosclerosis, fibrillar collagen deposition, and transforming growth factor-ß expression increased in a time-dependent manner, and SPARC and ADAMTS1 expression showed a similar pattern to these changes. The angiotensin II type-1 receptor blocker losartan suppressed the overexpression of SPARC and ADAMTS1, and an in vitro exposure to angiotensin II induced the production of both SPARC and ADAMTS1 in renal fibroblast NRK-49F cells. Knockdown of the SPARC gene with small interfering RNA reduced all forms (the 110-kDa latent and 87- and 65-kDa bioactive forms) of ADAMTS1 expression as well as collagen production. These results suggest that SPARC is induced by the renin-angiotensin system and may be a fibrogenic factor, at least in part, by producing ADAMTS1 in hypertensive renal disease.


Asunto(s)
Proteína ADAMTS1/metabolismo , Colágenos Fibrilares , Riñón , Losartán/farmacología , Osteonectina/metabolismo , Sistema Renina-Angiotensina , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Acetato de Desoxicorticosterona/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Colágenos Fibrilares/biosíntesis , Colágenos Fibrilares/metabolismo , Fibrosis/metabolismo , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Riñón/metabolismo , Riñón/patología , Mineralocorticoides/farmacología , Ratas , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Cloruro de Sodio Dietético/administración & dosificación
11.
Pancreas ; 50(9): 1267-1273, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34860810

RESUMEN

OBJECTIVES: The potential of DNA methylation alterations in early pancreatic cancer (PC) detection among pancreatic tissue cell-free DNA seems promising. This study investigates the diagnostic capacity of the 4-gene methylation biomarker panel, which included ADAMTS1, BNC1, LRFN5, and PXDN genes, in a case-control study. METHODS: A genome-wide pharmacoepigenetic approach identified ADAMTS1, BNC1, LRFN5, and PXDN genes as putative targets. Tissue samples including stage I-IV PC (n = 44), pancreatic intraepithelial neoplasia (n = 15), intraductal papillary mucinous neoplasms (n = 24), and normal pancreas (n = 8), and cell-free DNA, which was acquired through methylation on beads technology from PC (n = 22) and control patients (n = 10), were included. The 2-∆ct was the outcome of interest and underwent receiver operating characteristic analysis to determine the diagnostic accuracy of the panel. RESULTS: Receiver operating characteristic analysis revealed an area under the curve of 0.93 among ADAMTS1, 0.76 among BNC1, 0.75 among PXDN, and 0.69 among LRFN5 gene. The combination gene methylation panel (ADAMTS1, BNC1, LRFN5, and PXDN) had an area under the curve of 0.94, with a sensitivity of 100% and specificity of 90%. CONCLUSIONS: This methylation-based biomarker panel had promising accuracy for PC detection and warranted further validation in prospective PC surveillance trials.


Asunto(s)
Biomarcadores de Tumor/genética , Ácidos Nucleicos Libres de Células/genética , Metilación de ADN , Detección Precoz del Cáncer/métodos , Neoplasias Pancreáticas/genética , Proteína ADAMTS1/genética , Anciano , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de Unión al ADN/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/diagnóstico , Peroxidasas/genética , Curva ROC , Factores de Transcripción/genética
12.
J Proteomics ; 249: 104358, 2021 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-34450332

RESUMEN

The chondroitin sulfate proteoglycan versican is important for embryonic development and several human disorders. The versican V1 splice isoform is widely expressed and cleaved by ADAMTS proteases at a well-characterized site, Glu441-Ala442. Since ADAMTS proteases cleave the homologous proteoglycan aggrecan at multiple sites, we hypothesized that additional cleavage sites existed within versican. We report a quantitative label-free approach that ranks abundance of liquid chromatography-tandem mass spectrometry (LC-MS/MS)-identified semi-tryptic peptides after versican digestion by ADAMTS1, ADAMTS4 and ADAMTS5 to identify site-specific cleavages. Recombinant purified versican V1 constructs were digested with the recombinant full-length proteases, using catalytically inactive mutant proteases in control digests. Semi-tryptic peptide abundance ratios determined by LC-MS/MS in ADAMTS:control digests were compared to the mean of all identified peptides to obtain a z-score by which outlier peptides were ranked, using semi-tryptic peptides identifying Glu441 -Ala442 cleavage as the benchmark. Tryptic peptides with higher abundance in control digests supported cleavage site identification. We identified several novel cleavage sites supporting the ADAMTS1/4/5 cleavage site preference for a P1-Glu residue in proteoglycan substrates. Digestion of proteins in vitro and application of this z-score approach is potentially widely applicable for mapping protease cleavage sites using label-free proteomics. SIGNIFICANCE: Versican abundance and turnover are relevant to the pathogenesis of several human disorders. Versican is cleaved by A Disintegrin-like And Metalloprotease with Thrombospondin type 1 motifs (ADAMTS) family members at Glu441-Ala442, generating a bioactive proteoform called versikine, but additional cleavage sites and the site-specificity of individual ADAMTS proteases is unexplored. Here, we used a label-free proteomics strategy to identify versican cleavage sites for 3 ADAMTS proteases, applying a novel z-score-based statistical approach to compare the protease digests of versican to controls (digests with inactive protease) using the known protease cleavage site as a benchmark. We identified 21 novel cleavage sites that had a comparable z-score to the benchmark. Given the functional significance of versikine, they represent potentially significant cleavages and helped to refine a substrate site preference for each protease.The z-score approach is potentially widely applicable for discovery of site-specific cleavages within an purified protein or small ensemble of proteins using any protease.


Asunto(s)
Proteómica , Versicanos , Proteínas ADAM , Proteína ADAMTS1 , Proteína ADAMTS4 , Proteína ADAMTS5 , Cromatografía Liquida , Humanos , Espectrometría de Masas en Tándem , Versicanos/química
13.
Turk Neurosurg ; 31(5): 731-739, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34169995

RESUMEN

AIM: To analyze the expression of ADAMTS-1, NF-?B, and STAT3 in human pleomorphic xanthoastrocytoma specimens, and their correlation with glioma advancement. MATERIAL AND METHODS: Pleomorphic xanthoastrocytoma tumor cell lines were treated with low and high doses of cytokines at 24 and 48 hours (h) to replicate the inflammatory environment. The effects of IL-1 were assessed with the scratch wound-healing assay, and the expression levels of ADAMTS-1, NF-?B, and STAT3 of the groups were determined by western blot analysis. RESULTS: Cytokine treatment significantly increased the migration of PXA glioma cells after scratching at 24h and 48h time points. Similarly, 10 and 30 ng/mL IL-1 induced 1.86 and 1.94 fold increases, respectively, in ADAMTS-1 expression after 24h, and 3 and 3.27 fold increases, respectively, after 48h, compared with the non-treatment control group.10 and 30 ng/mL IL-1 doses caused 2.5 and 2.6 fold increase, in NF-?B protein levels after 24h, and 3.16 and 3.41 fold increases after 48h, compared with the non-treatment group. The protein levels of STAT3 after 24h were 2.62 and 2.43 fold higher, and 3.78 and 3.84 fold higher after 48 hours, with 10 and 30 ng/mL IL-1, compared with the non-treatment group. CONCLUSION: The proliferation and progression of glioma cells were proportional to the increased expression levels of ADAMTS-1, NF-?B, and STAT3. Our findings indicate that the proteolytic function of ADAMTS-1 may be associated with the malignant transformation of low-grade gliomas.


Asunto(s)
Proteína ADAMTS1/metabolismo , Astrocitoma , Glioma , Transformación Celular Neoplásica , Citocinas , Humanos
14.
Life Sci ; 281: 119756, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34175316

RESUMEN

Duchenne Muscular Dystrophy (DMD) is caused by mutations in the dystrophin gene, accompanied by aberrant extracellular matrix synthesis and muscle damage. ADAMTS1 metalloproteinase was reported increased in dystrophin-deficient mdx mouse. The aim of this study was to explore the role of ADAMTS1 in muscle function, fibrosis and damage, and respiratory function of mdx mice. 102 DMD patients and their mothers were included in this study. Multiplex ligation dependent probe amplification (MLPA) assay and Next-generation sequencing (NGS) were adopted to do genetic diagnosis. Dystrophin-deficient mdx mice were treated with anti-ADAMTS1 antibody (anti-ADAMTS1) for three weeks. The results showed that ADAMTS1 was increased in gastrocnemius muscle of mdx mice and serum of DMD patients. Anti-ADAMTS1 treatment increased Versican transcription but suppressed versican protein expression. Besides, we found anti-ADAMTS1 improved muscle strength, diaphragm and extensor digitorum longus muscles functions in mdx mice. Meanwhile, muscle fibrosis and damage were attenuated in anti-ADAMTS1 treated dystrophic mice. In summary, anti-ADAMTS1 antibody relieved muscle dysfunction and fibrosis in dystrophic mice. It is suggested that ADAMTS1 is a potential target for developing new biological therapies for DMD.


Asunto(s)
Proteína ADAMTS1/antagonistas & inhibidores , Anticuerpos Monoclonales/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/terapia , Proteína ADAMTS1/genética , Proteína ADAMTS1/inmunología , Proteína ADAMTS1/metabolismo , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Fibrosis/terapia , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Proteínas Musculares/metabolismo , Fuerza Muscular/inmunología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , ARN Mensajero/metabolismo , Versicanos/inmunología
15.
Gen Comp Endocrinol ; 311: 113835, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34181931

RESUMEN

Prostaglandins (PGs) are a class of fatty-acid derived hormones that are essential in ovulation of teleosts, but their exact role remains unknown. One putative target of PGs in ovulation is regulation of the expression of members of the A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) family, which are implicated in follicular rupture. This study investigated the regulation of ADAMTS, other proteases, and their inhibitors in response to treatment with PGE2 or PGF2α. Four members of the ADAMTS family, ADAMTS1, ADAMTS5, ADAMTS9, and ADAMTS16 were shown to be expressed in the ovary of zebrafish, but only adamts1 was upregulated in full-grown follicles following treatment with PGE2. Inhibitors of the PG receptors EP1 and EP2 had no effect on PGE2-stimulated adamts1 expression, while treatment of full-grown follicles with both PGE2 and GW627368x, an inhibitor of EP4 function, prevented the PGE2-induced increase in adamts1 expression. Treatment of full-grown follicles with the maturation-inducing hormone 17α,20ß-dihydroxy-4-pregnen-3-one (17,20ß-P) in vitro had no effect on the expression of adamts1 mRNA. These findings suggest that expression of ADAMTS1 in zebrafish ovarian follicles is regulated by the prostaglandin PGE2 via the EP4 series prostaglandin receptor.


Asunto(s)
Ovario , Pez Cebra , Proteína ADAMTS1/metabolismo , Animales , Femenino , Folículo Ovárico/metabolismo , Ovulación/fisiología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Pez Cebra/genética
16.
JCI Insight ; 6(9)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33986190

RESUMEN

White adipose tissue not only serves as a reservoir for energy storage but also secretes a variety of hormonal signals and modulates systemic metabolism. A substantial amount of adipose tissue develops in early postnatal life, providing exceptional access to the formation of this important tissue. Although a number of factors have been identified that can modulate the differentiation of progenitor cells into mature adipocytes in cell-autonomous assays, it remains unclear which are connected to physiological extracellular inputs and are most relevant to tissue formation in vivo. Here, we elucidate that mature adipocytes themselves signal to adipose depot-resident progenitor cells to direct depot formation in early postnatal life and gate adipogenesis when the tissue matures. Our studies revealed that as the adipose depot matures, a signal generated in mature adipocytes is produced, converges on progenitor cells to regulate the cytoskeletal protein MYH9, and attenuates the rate of adipogenesis in vivo.


Asunto(s)
Proteína ADAMTS1/genética , Adipocitos/metabolismo , Adipogénesis/genética , Tejido Adiposo Blanco/metabolismo , Homeostasis/genética , Cadenas Pesadas de Miosina/genética , Células Madre/metabolismo , Proteína ADAMTS1/metabolismo , Tejido Adiposo/metabolismo , Animales , Masculino , Ratones , Ratones Transgénicos , Cadenas Pesadas de Miosina/metabolismo
17.
Ann Clin Lab Sci ; 51(2): 198-205, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33941559

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease caused by both genetic and environmental factors. This study aimed to explore the underlying molecular mechanism of AD by bioinformatic gene analysis. The gene expression profiles of GSE16759 and GSE28146 were downloaded, and co-differentially expressed genes (co-DEGs) were identified by R software. Subsequently, the data were analyzed using a combined bioinformatics approach and predicted the microRNAs (miRNAs) targeting the key gene using miRNA databases. Based on the results of these analyses, ADAMTS1, CITED2, and GABRA2 were identified as co-DEGs. They were all associated with learning disorders (inference Score: 103.22, 140.41, and 96.26, respectively) and memory disorders (inference Score: 102.77, 132.68, and 81.80, respectively). The hub-genes of ADAMTS1, CITED2, and GABRA2 may be associated with AD. Additionally, miR-548c- 3p is probably a common target for ADAMTS1, CITED2, and GABRA2.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Predisposición Genética a la Enfermedad/genética , Proteína ADAMTS1/genética , Biomarcadores , China , Biología Computacional/métodos , Bases de Datos Genéticas , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes/genética , Pruebas Genéticas/métodos , Humanos , MicroARNs/genética , Enfermedades Neurodegenerativas/genética , Receptores de GABA-A/genética , Proteínas Represoras/genética , Transactivadores/genética , Transcriptoma/genética
18.
J Mol Neurosci ; 71(1): 28-41, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32567007

RESUMEN

This study was designed to test whether the Cronobacter sakazakii infection-impaired contextual learning and memory are mediated by the activation of the complement system; subsequent activation of inflammatory signals leads to alternations in serotonin transporter (SERT). To test this, rat pups (postnatal day, PND 15) were treated with either C. sakazakii (107 CFU) or Escherichia coli OP50 (107 CFU) or Luria bertani broth (100 µL) through oral gavage and allowed to stay with their mothers until PND 24. Experimental groups' rats were allowed to explore (PNDs 31-35) and then trained in contextual learning task (PNDs 36-43). Five days after training, individuals were tested for memory retention (PNDs 49-56). Observed behavioural data showed that C. sakazakii infection impaired contextual-associative learning and memory. Furthermore, our analysis showed that C. sakazakii infection activates complement system complement anaphylatoxin (C5a) (a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS1)) and mitogen-activated protein kinase kinase1 (MEKK1). Subsequently, MEKK1 induces pro-inflammatory signals possibly through apoptosis signal-regulating kinase-1 (ASK-1), c-Jun N-terminal kinase (JNK1/3) and protein kinase B gamma (AKT-3). In parallel, activated nuclear factor kappa-light-chain-enhancer B cells (NF-κB) induces interleukin-6 (IL-6) and IFNα-1, which may alter the level of serotonin transporter (SERT). Observed results suggest that impaired contextual learning and memory could be correlated with C5a-mediated NF-κß and ASK1 pathways.


Asunto(s)
Aprendizaje por Asociación/fisiología , Activación de Complemento , Complemento C5a/fisiología , Cronobacter sakazakii/patogenicidad , Infecciones por Enterobacteriaceae/complicaciones , Discapacidades para el Aprendizaje/etiología , MAP Quinasa Quinasa Quinasa 5/fisiología , Trastornos de la Memoria/etiología , FN-kappa B/fisiología , Proteínas del Tejido Nervioso/fisiología , Serotonina/metabolismo , Transducción de Señal/fisiología , Proteína ADAMTS1/metabolismo , Animales , Animales Lactantes , Corteza Cerebral/metabolismo , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/inmunología , Regulación de la Expresión Génica/inmunología , Inflamación , Interferón-alfa/metabolismo , Interleucina-6/metabolismo , Quinasas Janus/metabolismo , Discapacidades para el Aprendizaje/inmunología , Discapacidades para el Aprendizaje/microbiología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Trastornos de la Memoria/inmunología , Trastornos de la Memoria/microbiología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
19.
Cell Signal ; 77: 109827, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33161094

RESUMEN

ADAMTSs (A Disintegrin And Metalloproteinase with ThromboSpondin motifs) are secreted proteases dependent on Zn2+/Ca2+, involved in physiological and pathological processes and are part of the extracellular matrix (ECM). Here, we investigated if ADAMTS-1 is required for invasion and migration of cells and the possible mechanism involved. In order to test ADAMTS-1's role in ovarian cancer cells (CHO, NIH-OVCAR-3 and ES2) and NIH-3 T3 fibroblasts, we modified the levels of ADAMTS-1 and compared those to parental. Cells exposed to ADAMTS-1-enriched medium exhibited a decline in cell migration and invasion when compared to controls with or without a functional metalloproteinase domain. The opposite was observed in cells when ADAMTS-1 was deleted via the CRISPR/Cas9 approach. The decline in ADAMTS-1 levels enhanced the phosphorylated form of Src and FAK. We also evaluated the activities of cellular Rho GTPases from cell lysates using the GLISA® kit. The Cdc42-GTP signal was significantly increased in the CRISPR ADAMTS-1 ES-2 cells. By a Förster resonance energy transfer (FRET) biosensor for Cdc42 activity in ES-2 cells we demonstrated that Cdc42 activity was strongly polarized at the leading edge of migrating cells with ADAMTS-1 deletion, compared to the wild type cells. As conclusion, ADAMTS-1 inhibits proliferation, polarization and migration.


Asunto(s)
Proteína ADAMTS1/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína ADAMTS1/deficiencia , Proteína ADAMTS1/genética , Sistemas CRISPR-Cas/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Fosforilación , ARN Guía de Kinetoplastida/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
20.
Elife ; 92020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32909945

RESUMEN

The umbilical artery lumen closes rapidly at birth, preventing neonatal blood loss, whereas the umbilical vein remains patent longer. Here, analysis of umbilical cords from humans and other mammals identified differential arterial-venous proteoglycan dynamics as a determinant of these contrasting vascular responses. The umbilical artery, but not the vein, has an inner layer enriched in the hydrated proteoglycan aggrecan, external to which lie contraction-primed smooth muscle cells (SMC). At birth, SMC contraction drives inner layer buckling and centripetal displacement to occlude the arterial lumen, a mechanism revealed by biomechanical observations and confirmed by computational analyses. This vascular dimorphism arises from spatially regulated proteoglycan expression and breakdown. Mice lacking aggrecan or the metalloprotease ADAMTS1, which degrades proteoglycans, demonstrate their opposing roles in umbilical vascular dimorphism, including effects on SMC differentiation. Umbilical vessel dimorphism is conserved in mammals, suggesting that differential proteoglycan dynamics and inner layer buckling were positively selected during evolution.


Asunto(s)
Agrecanos/metabolismo , Miocitos del Músculo Liso , Arterias Umbilicales , Proteína ADAMTS1/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Humanos , Ratones Transgénicos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Parto/fisiología , Embarazo , Arterias Umbilicales/citología , Arterias Umbilicales/metabolismo , Arterias Umbilicales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA