Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Proteome Res ; 18(7): 2903-2914, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31117637

RESUMEN

Small cell lung cancer (SCLC) accounts for ∼14% of total lung cancer, which is the worldwide leading cause of morbidity and mortality in cancer. Although SCLC can be treated with chemotherapy and radiotherapy, its 5 year survival rate is still below 7%. Therefore, it is essential to discover new molecules and elucidate the underlying mechanisms modulating the tumorigenesis and metastasis of SCLC for the unmet medical needs. The secreted form of A Disintegrin And Metalloproteinase 12 (ADAM12S) is highly expressed in SCLC and promotes the proliferation, migration, and invasion of SCLC cells. However, the underlying molecular mechanism is still elusive. Using stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics, we identify 82 ADAM12S-regulated proteins in an SCLC cell line. Our proteomics and biochemical analyses discover that ADAM12S overexpression elevates while ADAM12 knockdown reduces the rate-limiting enzyme hexokinase 1 (HK1) in glycolysis. Through bioinformatics analyses, genetic manipulation, and in vitro assays, we further reveal that ADAM12S promotes the proliferation, colony formation, migration, and invasion of SCLC cells through upregulating HK1. This work links ADAM12S to glucose metabolic pathways in its attribution to the tumorigenesis and metastasis of SCLC cells and might provide valuable information for the exploration of therapeutic intervention for SCLC.


Asunto(s)
Proteína ADAM12/fisiología , Hexoquinasa/metabolismo , Proteómica/métodos , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Marcaje Isotópico , Invasividad Neoplásica , Metástasis de la Neoplasia , Regulación hacia Arriba
2.
J Proteomics ; 182: 34-44, 2018 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-29729432

RESUMEN

A Disintegrin And Metalloproteinase 12 (ADAM12) is highly expressed in multiple cancers such as breast and cervical cancers and its high expression reduces the overall patient survival rate. ADAM12 has two major splicing variants, the long membrane-anchored form ADAM12L and the short secreted form ADAM12S. However, how they are regulated and whether they are modulated similarly or differently in cells are not clear. Here, we use affinity purification and mass spectrometry to identify the ADAM12S-interacting proteins. Spectral counting and MaxQuant label-free quantification reveal that ADAM12S but not ADAM12L specifically interacts with a subset of endoplasmic reticulum proteins, such as endoplasmin (GRP94), 78 kDa glucose-regulated protein (GRP78), and UDP-glucose:glycoprotein glucosyltransferase I (UGGT1), that regulate the folding and processing of secreted proteins. Further biochemical experiments validate the interaction between ADAM12S and several of its interacting proteins. Computational docking analysis demonstrates that GRP94 preferentially interacts with ADAM12S over ADAM12L. The data also suggest that both the protein expression level and the secretion of ADAM12S are regulated by GRP94 expression and knockdown. Our results reveal a link between these two proteins that are highly expressed in cancer cells. Furthermore, our studies define a new ADAM12S-specific regulator that may contribute to the cancer development. SIGNIFICANCE: A Disintegrin And Metalloproteinase 12 (ADAM12) is highly expressed in many cancers such as lung, breast, and cervical cancers. ADAM12 has two major splicing variants, the long membrane-anchored form ADAM12L and the short secreted form ADAM12S. However, how they are regulated and whether they are modulated similarly or differently are not completely understood. We use affinity purification and label-free quantitative proteomics to identify the ADAM12S-interacting proteins. Our results reveal that ADAM12S specifically interacts with a subset of endoplasmic reticulum proteins, including endoplasmin (GRP94), UDP-glucose:glycoprotein glucosyltransferase I (UGGT1), and neutral α-glucosidase AB (GANAB). Computer modeling reveals that ADAM12S interacts with the surface amino acids of GRP94 more strongly than ADAM12L. Biochemical experiments further reveal that GRP94 regulates both the protein level and the secretion of ADAM12S. Database mining finds that both GRP94 and ADAM12 are highly expressed in multiple cancers and their high expression is correlated with poor patient survival rate. Taken together, our work discovers a new upstream regulator for ADAM12S, which may contribute to its distinct functions in the regulation of the migration and invasion of cancer cells.


Asunto(s)
Proteína ADAM12/metabolismo , Glicoproteínas de Membrana/fisiología , Proteómica/métodos , Línea Celular Tumoral , Cromatografía de Afinidad , Retículo Endoplásmico/química , Chaperón BiP del Retículo Endoplásmico , Proteínas HSP70 de Choque Térmico , Humanos , Espectrometría de Masas , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana , Simulación del Acoplamiento Molecular , Neoplasias/etiología , Unión Proteica , Isoformas de Proteínas
3.
Nanoscale ; 9(24): 8410-8417, 2017 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-28604895

RESUMEN

The research field in enzyme-based biotechnology urgently requires the discovery of new materials and methods with high-performance. Here we report that highly crystalline graphite dots (GDs) can modulate enzyme activities, and simultaneously allow for real-time measurements on enzyme kinetics in combination with mass spectrometry (MS). A well-defined modulation of lipolytic activities from inhibition to enhancement can be realized by selectively coupling lipase enzymes with GDs containing specific functional groups on the surface. As a unique feature of our approach, GDs in the enzyme reaction can simultaneously serve as a versatile matrix for rapid and sensitive detection of the residual enzyme substrate, the intermediate or final product of lipolytic digestion using MS technology. Therefore, enzyme kinetic data can be collected in a real-time, high-throughput format. This work provides a new platform for enzymological research in hybrid bio-catalytic processes with advanced nanotechnology.

4.
J Proteomics ; 148: 94-104, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27432471

RESUMEN

UNLABELLED: A Disintegrin and Metalloproteinase 12 (ADAM12) is expressed significantly higher in multiple tumors than in normal tissues and has been used as a prognostic marker for the evaluation of cancer progression. Although several ADAM12 substrates have been identified biochemically and its proteolytic function has been explored, the upstream regulators and the interacting proteins have not been systematically investigated. Here, we use immunoprecipitation and mass spectrometry (MS)-based quantitative proteomic approaches to identify 28 interacting partners for the long form of ADAM12 (ADAM12-L) in HeLa cells. Proteins that regulate cell proliferation, invasion, and epithelial to mesenchymal transition are among the identified ADAM12-interacting proteins. Further biochemical experiments discover that the protein level and the stability of ADAM12 are upregulated by one of its interacting proteins, myoferlin. In addition, myoferlin also increases the proteolytic activity of ADAM12, leading to the reduction of an ADAM12 substrate, E-cadherin. This result implies that ADAM12 and its interacting proteins might converge to certain signaling pathways in the regulation of cancer cell progression. The information obtained here might be useful in the development of new strategies for modulating cell proliferation and invasion involved in the regulation between ADAM12 and its interacting partners. MS data are available via ProteomeXchange with identifier PXD003560. BIOLOGICAL SIGNIFICANCE: Regulation of the proliferation and invasion of cancer cells is important in cancer treatment. ADAM12 has been found to play important roles in regulating these processes and identification of its interacting partners will improve our understanding of its biological functions and provide basis for functional modulation. Through mass spectrometry-based quantitative proteomic approaches, we identify the interacting partners for ADAM12 in a human cancer cell line and find many proteins that are involved in the proliferation and invasion of cancer cells. A novel regulator, myoferlin, of ADAM12 is discovered and this protein increases ADAM12 expression level, stability, and its enzymatic activity, leading to the reduction of its substrate, E-cadherin, which plays important roles in the regulation of cell adhesion and tumor metastasis. This result provides a connection for two highly expressed proteins in cancer cells and may shed light on the regulation of their biological functions in cancer progression.


Asunto(s)
Proteína ADAM12/metabolismo , Cadherinas/metabolismo , Proteínas de Unión al Calcio/fisiología , Proteínas de la Membrana/fisiología , Proteínas Musculares/fisiología , Mapas de Interacción de Proteínas , Proteómica/métodos , Proteína ADAM12/análisis , Cadherinas/análisis , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/aislamiento & purificación , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/aislamiento & purificación , Proteínas Musculares/análisis , Proteínas Musculares/aislamiento & purificación , Invasividad Neoplásica , Neoplasias/metabolismo , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA