Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Cell ; 76(1): 163-176.e8, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31492633

RESUMEN

Sensing nutrient availability is essential for appropriate cellular growth, and mTORC1 is a major regulator of this process. Mechanisms causing mTORC1 activation are, however, complex and diverse. We report here an additional important step in the activation of mTORC1, which regulates the efflux of amino acids from lysosomes into the cytoplasm. This process requires DRAM-1, which binds the membrane carrier protein SCAMP3 and the amino acid transporters SLC1A5 and LAT1, directing them to lysosomes and permitting efficient mTORC1 activation. Consequently, we show that loss of DRAM-1 also impacts pathways regulated by mTORC1, including insulin signaling, glycemic balance, and adipocyte differentiation. Interestingly, although DRAM-1 can promote autophagy, this effect on mTORC1 is autophagy independent, and autophagy only becomes important for mTORC1 activation when DRAM-1 is deleted. These findings provide important insights into mTORC1 activation and highlight the importance of DRAM-1 in growth control, metabolic homeostasis, and differentiation.


Asunto(s)
Aminoácidos/metabolismo , Proteína 7 Relacionada con la Autofagia/metabolismo , Metabolismo Energético , Lisosomas/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Membrana/metabolismo , Células 3T3-L1 , Adipocitos/enzimología , Adipogénesis , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistema de Transporte de Aminoácidos y+L/genética , Sistema de Transporte de Aminoácidos y+L/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/genética , Glucemia/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Activación Enzimática , Células HEK293 , Células HeLa , Humanos , Insulina/sangre , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Transporte de Proteínas
2.
Biochim Biophys Acta ; 1864(11): 1579-85, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27519163

RESUMEN

The majority of poorly differentiated hepatocellular carcinomas (HCCs) develop from well-differentiated tumors. Endocytosis is a cellular function which is likely to take part in this development due to its important role in regulating the abundances of vital signaling receptors. Here, we aimed to investigate the abundance of endocytosis-associated proteins in HCCs with various differentiation grades. Therefore, we analyzed 36 tissue specimens from HCC patients via LC-MS/MS-based label-free quantitative proteomics including 19 HCC tissue samples with different degrees of histological grades and corresponding non-tumorous tissue controls. As a result, 277 proteins were differentially regulated between well-differentiated tumors and controls. In moderately and poorly differentiated tumors, 278 and 1181 proteins, respectively, were significantly differentially regulated compared to non-tumorous tissue. We explored the regulated proteins based on their functions and identified thirty endocytosis-associated proteins, mostly overexpressed in poorly differentiated tumors. These included proteins that have been shown to be up-regulated in HCC like clathrin heavy chain-1 (CLTC) as well as unknown proteins, such as secretory carrier-associated membrane protein 3 (SCAMP3). The abundances of SCAMP3 and CLTC were immunohistochemically examined in tissue sections of 84 HCC patients. We demonstrate the novel association of several endocytosis-associated proteins, in particular, SCAMP3 with HCC progression.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas Portadoras/genética , Cadenas Pesadas de Clatrina/genética , Endocitosis/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Proteínas de la Membrana/genética , Adulto , Anciano , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/cirugía , Proteínas Portadoras/metabolismo , Cromatografía Liquida , Cadenas Pesadas de Clatrina/metabolismo , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Clasificación del Tumor , Proteoma/genética , Proteoma/metabolismo , Espectrometría de Masas en Tándem
3.
Dev Cell ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38870942

RESUMEN

Retrograde transport of WLS (Wntless) from endosomes to trans-Golgi network (TGN) is required for efficient Wnt secretion during development. However, the molecular players connecting endosomes to TGN during WLS trafficking are limited. Here, we identified a role for Eyes Absent (EYA) proteins during retrograde trafficking of WLS to TGN in human cell lines. By using worm, fly, and zebrafish models, we found that the EYA-secretory carrier-associated membrane protein 3 (SCAMP3) axis is evolved in vertebrates. EYAs form a complex and interact with retromer on early endosomes. Retromer-bound EYA complex recruits SCAMP3 to endosomes, which is necessary for the fusion of WLS-containing endosomes to TGN. Loss of EYA complex or SCAMP3 leads to defective transport of WLS to TGN and failed Wnt secretion. EYA mutations found in patients with hearing loss form a dysfunctional EYA-retromer complex that fails to activate Wnt signaling. These findings identify the EYA complex as a component of retrograde trafficking of WLS from the endosome to TGN.

4.
Cell Signal ; 104: 110591, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36627007

RESUMEN

The cellular trafficking protein secretory-carrier-membrane-protein 3 (SCAMP3) has been previously shown to promote hepatocellular carcinoma, melanoma, glioma and pancreatic adenocarcinoma. Moreover, previous work has shown that SCAMP3 regulates the epidermal growth factor receptor (EGFR) in triple negative breast cancer (TNBC). However, the oncogenic role of SCAMP3 in different molecular subtypes of breast cancer (BRCA) remains largely unknown. In this study, the role of SCAMP3 in different molecular subtypes of BRCA was investigated using in silico, in vitro and in vivo approaches. In silico analysis of BRCA patient samples showed that SCAMP3 is highly overexpressed in different BRCA molecular subtypes, advanced disease grades and lymph node metastatic stages. Depletion of SCAMP3 inhibited BRCA cell growth, stemness, clonogenic potential and migration and promoted autophagy and cellular senescence. The expression of stemness markers CD44 and OCT4A was reduced in SCAMP3-silenced MDA-MB-231 cells. SCAMP3 overexpression promoted cell proliferation, clonogenicity, tumor spheroid formation and migration in vitro and tumor growth in vivo. SCAMP3 promoted epithelial-mesenchymal-transition (EMT) by regulating E-cadherin expression. SCAMP3 enhanced in vivo tumor growth in MDA-MB-231 tumor xenograft mouse model. Mechanistically, SCAMP3 depletion inhibited ß-Catenin, c-MYC and SQSTM1 expression, while its overexpression increased the expression of the same oncogenic proteins. Increased SCAMP3 expression associated with increased chemoresistance in BRCA cells while its depletion associated with increased sensitivity to chemotherapy. BRCA patients with high SCAMP3 expression showed poor prognosis, decreased overall survival and relapse free survival relative to counterparts with reduced SCAMP3 expression. These findings suggest that SCAMP3 exerts a wide range of oncogenic effects in different molecular subtypes of BRCA by modulating the c-MYC-ß-Catenin-SQSTM1 axis that targets tumor growth, metastasis, stemness and chemoresistance.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , beta Catenina/metabolismo , Proteínas Portadoras/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Proteínas de la Membrana/metabolismo , Proteína Sequestosoma-1/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
5.
Cancers (Basel) ; 14(11)2022 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-35681787

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive, metastatic, and lethal breast cancer subtype. To improve the survival of TNBC patients, it is essential to explore new signaling pathways for the further development of effective drugs. This study aims to investigate the role of the secretory carrier membrane protein 3 (SCAMP3) in TNBC and its association with the epidermal growth factor receptor (EGFR). Through an internalization assay, we demonstrated that SCAMP3 colocalizes and redistributes EGFR from the cytoplasm to the perinucleus. Furthermore, SCAMP3 knockout decreased proliferation, colony and tumorsphere formation, cell migration, and invasion of TNBC cells. Immunoblots and degradation assays showed that SCAMP3 regulates EGFR through its degradation. In addition, SCAMP3 modulates AKT, ERK, and STAT3 signaling pathways. TNBC xenograft models showed that SCAMP3 depletion delayed tumor cell proliferation at the beginning of tumor development and modulated the expression of genes from the PDGF pathway. Additionally, analysis of TCGA data revealed elevated SCAMP3 expression in breast cancer tumors. Finally, patients with TNBC with high expression of SCAMP3 showed decreased RFS and DMFS. Our findings indicate that SCAMP3 could contribute to TNBC development through the regulation of multiple pathways and has the potential to be a target for breast cancer therapy.

6.
Microbiol Spectr ; 9(1): e0047521, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34378951

RESUMEN

Picornaviruses are a diverse and major cause of human disease, and their genomes replicate with intracellular membranes. The functionality of these replication organelles depends on the activities of both viral nonstructural proteins and co-opted host proteins. The mechanism by which viral-host interactions generate viral replication organelles and regulate viral RNA synthesis is unclear. To elucidate this mechanism, enterovirus A71 (EV-A71) was used here as a virus model to investigate how these replication organelles are formed and to identify the cellular components that are critical in this process. An immunoprecipitation assay was combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis to identify 172 cellular proteins and four viral proteins associating with viral 3A protein. Secretory carrier membrane protein 3 (SCAMP3) was one of the host proteins we selected for further investigation. Here, we demonstrate by immunoprecipitation assay that SCAMP3 associates with 3A protein and colocalizes with 3A protein during virus infection. SCAMP3 knockdown or knockout in infected cells decreases synthesis of EV-A71 viral RNA, viral proteins, and viral growth. Furthermore, the viral 3A protein associates with SCAMP3 and phosphatidylinositol-4-kinase type III ß (PI4KIIIß) as shown by immunoprecipitation assay and colocalizes to the replication complex. Upon infection of cells with a SCAMP3 knockout construct, PI4KIIIß and phosphatidylinositol-4-phosphate (PI4P) colocalization with EV-A71 3A protein decreases; viral RNA synthesis also decreases. SCAMP3 is also involved in the extracellular signal-regulated kinase (ERK) signaling pathway to regulate viral replication. The 3A and SCAMP3 interaction is also important for the replication of coxsackievirus B3 (CVB3). SCAMP3 also associates with 3A protein of CVB3 and enhances viral replication but does not regulate dengue virus 2 (DENV2) replication. Taken together, the results suggest that enterovirus 3A protein, SCAMP3, PI4KIIIß, and PI4P form a replication complex and positively regulate enterovirus replication. IMPORTANCE Virus-host interaction plays an important role in viral replication. 3A protein of enterovirus A71 (EV-A71) recruits other viral and host factors to form a replication complex, which is important for viral replication. In this investigation, we utilized immunoprecipitation combined with proteomics approaches to identify 3A-interacting factors. Our results demonstrate that secretory carrier membrane protein 3 (SCAMP3) is a novel host factor that associates with enterovirus 3A protein, phosphatidylinositol-4-kinase type III ß (PI4KIIIß), and phosphatidylinositol-4-phosphate (PI4P) to form a replication complex and positively regulates viral replication. SCAMP3 is also involved in the extracellular signal-regulated kinase (ERK) signaling pathway to regulate viral replication.


Asunto(s)
Proteínas Portadoras/metabolismo , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Proteínas Portadoras/genética , Enterovirus Humano A/genética , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Proteínas no Estructurales Virales/genética
7.
Onco Targets Ther ; 13: 3677-3687, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32431518

RESUMEN

INTRODUCTION: The secretory carrier-associated membrane protein 3 (SCAMP3) is a component of post-Golgi membranes, functions as a protein carrier and is critical for subcellular protein transportation. Limited studies revealed an elevated expression of SCAMP3 in breast cancer and hepatocellular carcinoma; however, its role in glioma remains unknown. The aim of our study is to investigate the expression pattern and functional mechanisms of SCAMP3 in glioma. METHODS: mRNA and protein levels of SCAMP3 were examined in glioma tissues together with nontumorous brain tissues by using quantitative real-time-PCR and immunohistochemistry staining. The prognostic role of SCAMP3 in glioma was evaluated through univariate and multivariate analyses. In vitro and in vivo assays were conducted to explore the underlying mechanisms of SCAMP3-induced glioma progression. RESULTS: The expression level of SCAMP3 was higher in glioma tissues than that in normal brain tissues. High protein level of SCAMP3 was correlated with larger tumor size and advanced WHO grade. Glioma patients with high-SCAMP3 level had worse overall survival. In addition, SCAMP3 was defined as an independent risk factor of glioma prognosis. Cellular and xenograft studies revealed that SCAMP3 promotes glioma proliferation possibly through enhancing EGFR and mTORC1 signaling. DISCUSSION: Our studies revealed that high-SCAMP3 expression level was closely related to the unfavorable clinical features and poor prognosis of glioma patients. SCAMP3 may serve as an invaluable prognostic indicator and novel therapeutic target for glioma treatment.

8.
Pharmgenomics Pers Med ; 13: 415-426, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33116758

RESUMEN

BACKGROUND: Secretory carrier membrane proteins 3 (SCAMP3) is an endocytosis-associated protein involved in regulating endosomal pathways and the trafficking of vital signaling receptors. This study aimed to comprehensively assess the role of SCAMP3 in hepatocellular carcinoma (HCC) by integrated bioinformatics analysis. METHODS: In this study, bioinformatics databases were used to explore the differential expression status and prognostic value of SCAMP3 gene in HCC, and bioinformatics analyses of survival data and interactors of SCAMP3 were conducted to predict the prognostic value of SCAMP3 in HCC. RESULTS: Using the TCGA data, our data shows that SCAMP3 mRNA expression is most significantly different between liver and hepatocellular carcinoma tissues and higher expression of SCAMP3 has unfavorable prognostic significance in HCC. Tumor grade, stage, and gender also showed a significant relevance with SCAMP3 expression. High SCAMP3 expression of males revealed significantly poorer survival and progression compared with low SCAMP3 expression of males. BioGRID statistics explores 79 unique interactions with SCAMP3 and multiple post translational modifications. Further analysis finds that SOCS2 may negatively correlate with SCAMP3, while GBA, MX1, and DDOST positively correlate with SCAMP3. Moreover, ncRNA analysis shows that SCAMP3 gene expression is positively associated with lncRNA SBF2-AS1 and negatively related with Has-miR-145. The expressions of SBF2-AS1 and Has-miR-145 are also significantly related with survival in HCC. DISCUSSION: SCAMP3 expression can be affected by multiple genes or ncRNAs expression that are associated with survival, thus suggesting that SCAMP3 can be used as a clinical diagnosis and prognostic biomarker in HCC.

9.
Onco Targets Ther ; 13: 1047-1057, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32099407

RESUMEN

BACKGROUND: Secretory Carrier Membrane Proteins 3 (SCAMP3) is a transmembrane protein that affects intracellular trafficking, protein sorting and vesicle formation. Overexpression of SCAMP3 correlates with poorly differentiated hepatocellular carcinoma (HCC). However, the expression and corresponding gene regulation of SCAMP3 in HCC remain unclear. METHODS: Bioinformatics analyses of clinical parameters and survival data were conducted to predict the prognostic value of SCAMP3 in HCC. RNA sequencing and real-time PCR were conducted to confirm the SCAMP3 expression in HCC tissue. Expression was analyzed using OncomineTM and UALCAN, while SCAMP3 alterations and survival analysis were identified by cBioPortal. Differential gene expression with SCAMP3 was analyzed by LinkedOmics and GEPIA. The target networks of enzymes and co-transcriptional factors were identified using Gene enrichment analysis. Expression of SCAMP3 in HCC tissue was detected by RNA-sequencing and Western-blotting. RESULTS: Based on bioinformatics analysis and detection of mRNA expression, SCAMP3 was over-expressed in numerous tumors, especially in HCC. SCAMP3 level was positively correlated with disease stages and tumor grades and negatively correlated with patient survival. Furthermore, functional network analysis indicated that SCAMP3 regulated metabolic process and DNA replication through oxidative phosphorylation and chromatin remodeling or Ribosome. SCAMP3 regulated a number of gene expressions including PPAP2B, SNRK, ARID4A, PRCC, VPS72 via protein binding and proteasome, which may affect cell adhesion, proliferation, transcription, cell cycle and metabolism. Further, Real-time PCR and Western-blotting showed that the SCAMP3 level was increased in HCC tissue. CONCLUSION: The present data analysis efficiently reveals information about SCAMP3 expression and correlated function in HCC, laying a foundation for further study of SCAMP3 in the tumor.

10.
Am J Transl Res ; 12(12): 7870-7884, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33437366

RESUMEN

PURPOSE: To explore the regulatory mechanism of secretory carrier membrane protein 3 (SCAMP3) and miR-128-3p in hepatocellular carcinoma (HCC). PATIENTS AND METHODS: Cancer tissues and adjacent tissues of 52 HCC patients treated in our hospital were collected to explore the prognostic factors affecting their 3-year survival. HCC cells were purchased, the gene expression of Huh-7 and MHCC97 were adjusted by transfection, and the levels of SCAMP3, miR-128-3p, EGFR, p-EGFR, MAPK p38, p-MAPK p38, N-cadherin, vimentin, E-cadherin, cell proliferation, migration, invasion, apoptosis and epithelial-mesenchymal transition (EMT) were detected. A nude mouse model of HCC was constructed to verify the effects of transfection of mimics. RESULTS: SCAMP3 was elevated in HCC patients and cancer tissues of HCC patients, while miR-128-3p showed opposite effects. High level SCAMP3 and low level miR-128-3p were related to poor prognosis of HCC. Both of them were correlated with excessive drinking history, N-stage, M-stage and pathological differentiation degree of HCC patients, as well as prognostic factors of HCC patients. SCAMP3 up-regulation or miR-128-3p down-regulation could promote HCC cell proliferation, migration, invasion, and transcription and protein levels of EGFR, p-EGFR, MAPK p38, p-MAPK p38, N-cadherin and vimentin, and inhibit HCC cell apoptosis and transcription and protein levels of E-cadherin. Dual luciferase reporter identified the targeting relationship between SCAMP3 and miR-128-3p. When both SCAMP3 and miR-128-3p were elevated or reduced, the biological manifestation of cells was not different from that of miR-NC transfected with unrelated sequences. Besides, miR-128-3p inhibited tumor growth in the HCC model in nude mice. CONCLUSION: SCAMP3 can be controlled by miR-128-3p and can mediate the EGFR-MAPK p38 signaling pathway to inhibit HCC cell metastasis, which is expected to become a promising therapeutic target for HCC.

11.
Am J Cancer Res ; 6(8): 1720-40, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27648361

RESUMEN

Inflammatory Breast Cancer (IBC) is the most lethal form of breast cancer with a 35% 5-year survival rate. The accurate and early diagnosis of IBC and the development of targeted therapy against this deadly disease remain a great medical challenge. Plasma membrane proteins (PMPs) such as E-cadherin and EGFR, play an important role in the progression of IBC. Because the critical role of PMPs in the oncogenic processes they are the perfect candidates as molecular markers and targets for cancer therapies. In the present study, Stable Isotope Labeling with Amino Acids in Cell Culture (SILAC) followed by mass spectrometry analysis was used to compare the relative expression levels of membrane proteins (MP) between non-cancerous mammary epithelial and IBC cells, MCF-10A and SUM-149, respectively. Six of the identified PMPs were validated by immunoblotting using the membrane fractions of non-IBC and IBC cell lines, compared with MCF-10A cells. Immunohistochemical analysis using IBC, invasive ductal carcinoma or normal mammary tissue samples was carried out to complete the validation method in nine of the PMPs. We identified and quantified 278 MPs, 76% of which classified as PMPs with 1.3-fold or higher change. We identified for the first time the overexpression of the novel plasminogen receptor, PLGRKT in IBC and of the carrier protein, SCAMP3. Furthermore, we describe the positive relationship between L1CAM expression and metastasis in IBC patients and the role of SCAMP3 as a tumor-related protein. Overall, the membrane proteomic signature of IBC reflects a global change in cellular organization and suggests additional strategies for cancer progression. Together, this study provides insight into the specialized IBC plasma membrane proteome with the potential to identify a number of novel therapeutic targets for IBC.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda