Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Glycobiology ; 33(3): 215-224, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-36651496

RESUMO

The monocyte adhesion to endothelial cells is an early step in chronic inflammation. Interferon-γ (IFN-γ) is regarded as a master regulator of inflammation development. However, the significance and mechanisms of IFN-γ in the monocyte adhesion to endothelial cells remains largely unknown. IFN-γ up-regulates PD-L1 on various types of cells. Here, we performed flow cytometry to examine the contribution of IFN-γ-induced PD-L1 expression on monocyte adhesion to endothelial cells. Up-regulation of PD-L1 by IFN-γ enhanced the adhesion of monocytes to endothelial cells. By immunoprecipitation and lectin blot, PD-L1 in endothelial cells interacted with CD169/Siglec 1 in monocytes depending on the α2,3-sialylation of PD-L1. ST3Gal family (ST3ß-galactoside α-2,3-sialyltransferase) was the major glycosyltransferase responsible for the α2,3-sialylation of membrane proteins. Down-regulation of ST3Gal4 by RNAinterference partially reduced the α2,3-sialylation of PD-L1 and the PD-L1-CD169 interaction. Finally, purified PD-L1 protein with α2,3-sialylation, but not PD-L1 protein without α2,3-sialylation, partially reduced IFN-γ-induced monocyte adhesion to endothelial cells. These findings provide evidence that the interaction between PD-L1 and CD169 promoted monocyte adhesion to endothelial cells and might elucidate a new mechanism of monocyte adhesion to endothelial cells.


Assuntos
Células Endoteliais , Monócitos , Humanos , Células Endoteliais/metabolismo , Inflamação , Interferon gama/farmacologia , Interferon gama/metabolismo , Monócitos/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Antígeno B7-H1/metabolismo
2.
Glycobiology ; 33(6): 464-475, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37129515

RESUMO

PURPOSE: Sialic acid-bound immunoglobulin-like lectin 15 (Siglec15) has emerged as a novel therapeutic target in tumor immunotherapy. This study is designed to investigate the function and mechanism of Siglec15 in thyroid carcinoma (THCA). MATERIALS AND METHODS: The information on patients with THCA from TGCA and GEO database were used to analyze the expression of Siglec15 in THCA. THCA cells were treated with Siglec15-mFc, a recombinant fusion protein consisting of the extracellular domain of human Siglec15 and murine IgG Fc. THP-1 cells expressing human Siglec15 and its mutant were co-cultured with THCA cells to mimic the contact between Siglec15-expressing tumor-associated macrophages and THCA cells. Wound-healing assay and transwell migration assay were used to examine the migration abilities of BCPAP and C643 cells. Pull-down assay was performed to examine the interaction between Siglec15 and epidermal growth factor receptor (EGFR) on the cancer cells. Cycloheximide (CHX) assay was used to evaluate the stability of the protein. RESULTS: The expression of Siglec15 in thyroid carcinoma tissues is higher than in normal tissues. Siglec15 promotes the migration of THCA cells by binding to EGFR in a sialic acid-dependent manner and increases EGFR protein expression. Inhibition of the EGFR pathway blocks the effect of Siglec15 on the migration of THCA cells. CONCLUSIONS: Our findings reveals that Siglec15 promotes the migration of thyroid carcinoma cells by enhancing the EGFR protein stability.


Assuntos
Ácido N-Acetilneuramínico , Neoplasias da Glândula Tireoide , Humanos , Animais , Camundongos , Movimento Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/patologia , Estabilidade Proteica
3.
J Cell Mol Med ; 23(4): 2384-2398, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30677218

RESUMO

O-GlcNAcylation catalysed by O-GlcNAc transferase (OGT) is a reversible post-translational modification. O-GlcNAcylation participates in transcription, epigenetic regulation, and intracellular signalling. Dysregulation of O-GlcNAcylation in response to high glucose or OGT expression has been implicated in metabolic diseases and cancer. However, the underlying mechanisms by which OGT regulates hepatoma development remain largely unknown. Here, we employed the lentiviral shRNA-based system to knockdown OGT to analyse the contribution of OGT in hepatoma cell proliferation and stem-like cell potential. The sphere-forming assay and western blot analysis of stem-related gene expression were used to evaluate stem-like cell potential of hepatoma cell. We found that the level of total O-GlcNAcylation or OGT protein was increased in hepatocellular carcinoma. OGT activated stem-like cell potential in hepatoma through eukaryotic initiation factor 4E (eIF4E) which bound to stem-related gene Sox2 5'-untranslated region. O-GlcNAcylation of eIF4E at threonine 168 and threonine 177 protected it from degradation through proteasome pathway. Expression of eIF4E in hepatoma was determined by immunostaining in 232 HCC patients, and Kaplan-Meier survival analysis was used to determine the correlation of eIF4E expression with prognosis. High glucose promoted stem-like cell potential of hepatoma cell through OGT-eIF4E axis. Collectively, our findings indicate that OGT promotes the stem-like cell potential of hepatoma cell through O-GlcNAcylation of eIF4E. These results provide a mechanism of HCC development and a cue between the pathogenesis of HCC and high glucose condition.


Assuntos
Carcinoma Hepatocelular/genética , Fator de Iniciação 4E em Eucariotos/genética , Neoplasias Hepáticas/genética , N-Acetilglucosaminiltransferases/genética , Acilação/genética , Idoso , Apoptose/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células/genética , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Glucose/metabolismo , Humanos , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Prognóstico , Processamento de Proteína Pós-Traducional/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/genética
4.
J Biol Chem ; 293(6): 2219-2230, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29269413

RESUMO

Malignant glioblastoma multiforme is one of the most aggressive human cancers, with very low survival rates. Recent studies have reported that glioma stem-like cells transdifferentiate into endothelial cells, indicating a new mechanism for tumor angiogenesis and potentially providing new therapeutic options for glioblastoma treatment. Glioma malignancy is strongly associated with altered expression of N-linked oligosaccharide structures on the cell surface. We have previously reported that ß1,4-galactosyltransferase V (ß1,4GalTV), which galactosylates the GlcNAcß1-6Man arm of the branched N-glycans, is highly expressed in glioma and promotes glioma cell growth in vitro and in vivo However, the mechanism by which ß1,4GalTV stimulates glioma growth is unknown. Here we demonstrate that short hairpin RNA-mediated ß1,4GalTV knockdown inhibits the tumorigenesis of glioma stem-like cells and reduces their transdifferentiation into endothelial cells. We also found that ß1,4GalTV overexpression increased glioma stem-like cell transdifferentiation into endothelial cells and that this effect required ß1,4GalTV galactosylation activity. Moreover, ß1,4GalTV promoted ß1,4-galactosylation of Notch1 and increased Notch1 protein levels. Of note, ectopic expression of activated Notch1 rescued the inhibitory effect of ß1,4GalTV depletion on glioma stem-like cell transdifferentiation. In summary, our findings indicate that ß1,4GalTV stimulates transdifferentiation of glioma stem-like cells into endothelial cells by activating Notch1 signaling. These detailed insights shed important light on the mechanisms regulating glioma angiogenesis.


Assuntos
Transdiferenciação Celular , Glioma/patologia , N-Acetil-Lactosamina Sintase/fisiologia , Receptor Notch1/metabolismo , Transdução de Sinais , Células Endoteliais/patologia , Humanos , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica
5.
J Hepatol ; 71(6): 1206-1215, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31499129

RESUMO

BACKGROUND & AIMS: The microenvironment regulates hepatoma stem cell behavior. However, the contributions of lymphatic endothelial cells to the hepatoma stem cell niche remain largely unknown; we aimed to analyze this contribution and elucidate the mechanisms behind it. METHODS: Associations between lymphatic endothelial cells and CD133+ hepatoma stem cells were analyzed by immunofluorescence and adhesion assays; with the effects of their association on IL-17A expression examined using western blot, quantitative reverse transcription PCR and luciferase reporter assay. The effects of IL-17A on the self-renewal and tumorigenesis of hepatoma stem cells were examined using sphere and tumor formation assays. The role of IL-17A in immune escape by hepatoma stem cells was examined using flow cytometry. The expression of IL-17A in hepatoma tissues was examined using immunohistochemistry. RESULTS: CD133+ hepatoma stem cells preferentially interact with lymphatic endothelial cells. The interaction between the mannose receptor and high-mannose type N-glycans mediates the interaction between CD133+ hepatoma stem cells and lymphatic endothelial cells. This interaction activates cytokine IL-17A expression in lymphatic endothelial cells. IL-17A promotes the self-renewal of hepatoma stem cells. It also promotes their immune escape, partly through upregulation of PD-L1. CONCLUSION: Interactions between lymphatic endothelial cells and hepatoma stem cells promote the self-renewal and immune escape of hepatoma stem cells, by activating IL-17A signaling. Thus, inhibiting IL-17A signaling may be a promising approach for hepatoma treatment. LAY SUMMARY: The microenvironment is crucial for the self-renewal and development of hepatoma stem cells, which lead to the development of liver cancer. Lymphatic endothelial cells are an important component of this niche microenvironment, helping hepatoma stem cells to self-renew and escape immune attack, by upregulating IL-17A signaling. Thus, targeting IL-17A signaling is a potential strategy for the treatment of hepatoma.


Assuntos
Antígeno AC133/imunologia , Antígeno B7-H1/imunologia , Carcinoma Hepatocelular , Células Endoteliais , Interleucina-17/imunologia , Neoplasias Hepáticas , Células-Tronco Neoplásicas/metabolismo , Carcinogênese/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Transdução de Sinais , Evasão Tumoral , Microambiente Tumoral , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Biol Chem ; 291(30): 15540-50, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27226554

RESUMO

CD133, a widely known cancer stem cell marker, has been proved to promote tumor metastasis. However, the mechanism by which CD133 regulates metastasis remains largely unknown. Here, we report that CD133 knockdown inhibits cancer cell migration, and CD133 overexpression promotes cell migration. CD133 expression is beneficial to activate the Src-focal adhesion kinase (FAK) signaling pathway. Further studies show that CD133 could interact with Src, and the region between amino acids 845 and 857 in the CD133 C-terminal domain is indispensable for its interaction with Src. The interaction activates Src to phosphorylate its substrate FAK and to promote cell migration. Likewise, a Src binding-deficient CD133 mutant loses the abilities to increase Src and FAK phosphorylation and to promote cell migration. Inhibition of Src activity by PP2, a known Src activity inhibitor, could block the activation of FAK phosphorylation and cell migration induced by CD133. In summary, our data suggest that activation of FAK by the interaction between CD133 and Src promotes cell migration, providing clues to understand the migratory mechanism of CD133(+) tumor cells.


Assuntos
Antígeno AC133/metabolismo , Movimento Celular , Quinase 1 de Adesão Focal/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Antígeno AC133/genética , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/genética , Humanos , Células-Tronco Neoplásicas/patologia , Fosforilação , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/genética
7.
Proc Natl Acad Sci U S A ; 110(17): 6829-34, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23569237

RESUMO

The biological significance of a known normal and cancer stem cell marker CD133 remains elusive. We now demonstrate that the phosphorylation of tyrosine-828 residue in CD133 C-terminal cytoplasmic domain mediates direct interaction between CD133 and phosphoinositide 3-kinase (PI3K) 85 kDa regulatory subunit (p85), resulting in preferential activation of PI3K/protein kinase B (Akt) pathway in glioma stem cell (GSC) relative to matched nonstem cell. CD133 knockdown potently inhibits the activity of PI3K/Akt pathway with an accompanying reduction in the self-renewal and tumorigenicity of GSC. The inhibitory effects of CD133 knockdown could be completely rescued by expression of WT CD133, but not its p85-binding deficient Y828F mutant. Analysis of glioma samples reveals that CD133 Y828 phosphorylation level is correlated with histopathological grade and overlaps with Akt activation. Our results identify the CD133/PI3K/Akt signaling axis, exploring the fundamental role of CD133 in glioma stem cell behavior.


Assuntos
Antígenos CD/metabolismo , Transformação Celular Neoplásica/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Glioma/fisiopatologia , Glicoproteínas/metabolismo , Células-Tronco Neoplásicas/fisiologia , Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Antígeno AC133 , Animais , Antígenos CD/genética , Western Blotting , Glioma/metabolismo , Glicoproteínas/genética , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação de Sentido Incorreto/genética , Células-Tronco Neoplásicas/metabolismo , Peptídeos/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Adv Sci (Weinh) ; 11(3): e2306715, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37997289

RESUMO

Targeting the niche components surrounding glioblastoma stem cells (GSCs) helps to develop more effective glioblastoma treatments. However, the mechanisms underlying the crosstalk between GSCs and microenvironment remain largely unknown. Clarifying the extracellular molecules binding to GSCs marker CD133 helps to elucidate the mechanism of the communication between GSCs and the microenvironment. Here, it is found that the extracellular domain of high mannose type CD133 physically interacts with Collagen 1 (COL1) in GSCs. COL1, mainly secreted by cancer-associated fibroblasts, is a niche component for GSCs. COL1 enhances the interaction between CD133 and p85 and activates Akt phosphorylation. Activation of Akt pathway increases transcription factor ATF4 protein level, subsequently enhances SLC1A5-dependent glutamine uptake and glutathione synthesis. The inhibition of CD133-COL1 interaction or down-regulation of SLC1A5 reduces COL1-accelerated GSCs self-renewal and tumorigenesis. Analysis of glioma samples reveals that the level of COL1 is correlated with histopathological grade of glioma and the expression of SLC1A5. Collectively, COL1, a niche component for GSCs, enhances the tumorigenesis of GSCs partially through CD133-Akt-SLC1A5 signaling axis, providing a new mechanism underlying the cross-talk between GSCs and extracellular matrix (ECM) microenvironment.


Assuntos
Glioblastoma , Glioma , Humanos , Glioblastoma/metabolismo , Glutamina/metabolismo , Manose/metabolismo , Manose/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células-Tronco Neoplásicas/metabolismo , Carcinogênese/metabolismo , Transformação Celular Neoplásica , Glioma/metabolismo , Colágeno/metabolismo , Microambiente Tumoral , Antígenos de Histocompatibilidade Menor/metabolismo , Antígenos de Histocompatibilidade Menor/farmacologia , Sistema ASC de Transporte de Aminoácidos/metabolismo
9.
Cell Rep ; 42(12): 113588, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38117655

RESUMO

CD133 is widely used as a marker to isolate tumor-initiating cells in many types of cancers. The structure of N-glycan on CD133 is altered during the differentiation of tumor-initiating cells. However, the relationship between CD133 N-glycosylation and stem cell characteristics remains elusive. Here, we found that the level of α-1,2-mannosylated CD133 was associated with the level of stemness genes in intrahepatic cholangiocarcinoma (iCCA) tissues. α-1,2-mannosylated CD133+ cells possessed the characteristics of tumor-initiating cells. The loss of the Golgi α-mannosidase I coding gene MAN1C1 resulted in the formation of α-1,2-mannosylated CD133 in iCCA-initiating cells. Mechanistically, α-1,2-mannosylation promoted the cytoplasmic distribution of CD133 and enhanced the interaction between CD133 and the autophagy gene FIP200, subsequently promoting the tumorigenesis of α-1,2-mannosylated CD133+ cells. Analysis of iCCA samples showed that the level of cytoplasmic CD133 was associated with poor iCCA prognosis. Collectively, α-1,2-mannosylated CD133 is a functional marker of iCCA-initiating cells.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Humanos , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Transformação Celular Neoplásica/patologia , Carcinogênese/patologia , Proteínas de Ciclo Celular , Ductos Biliares Intra-Hepáticos/patologia
10.
Adv Sci (Weinh) ; 9(26): e2202216, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35798319

RESUMO

The quiescent/slow-cycling state preserves the self-renewal capacity of cancer stem cells (CSCs) and leads to the therapy resistance of CSCs. The mechanisms maintaining CSCs quiescence remain largely unknown. Here, it is demonstrated that lower expression of MAN1A1 in glioma stem cell (GSC) resulted in the formation of high-mannose type N-glycan on CD133. Furthermore, the high-mannose type N-glycan of CD133 is necessary for its interaction with DNMT1. Activation of p21 and p27 by the CD133-DNMT1 interaction maintains the slow-cycling state of GSC, and promotes chemotherapy resistance and tumorigenesis of GSCs. Elimination of the CD133-DNMT1 interaction by a cell-penetrating peptide or MAN1A1 overexpression inhibits the tumorigenesis of GSCs and increases the sensitivity of GSCs to temozolomide. Analysis of glioma samples reveals that the levels of high-mannose type N-glycan are correlated with glioma recurrence. Collectively, the high mannose CD133-DNMT1 interaction maintains the slow-cycling state and tumorigenic potential of GSC, providing a potential strategy to eliminate quiescent GSCs.


Assuntos
Glioma , Manose , Antígeno AC133/metabolismo , Antígeno AC133/uso terapêutico , Carcinogênese , Glioma/tratamento farmacológico , Glioma/metabolismo , Humanos , Manose/metabolismo , Manose/uso terapêutico , Células-Tronco Neoplásicas/metabolismo
11.
Hepatology ; 52(1): 142-54, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20578140

RESUMO

UNLABELLED: One of the serious sequelae of chronic hepatitis B virus (HBV) infection is hepatocellular carcinoma (HCC). Among all the proteins encoded by the HBV genome, hepatitis B virus X protein (HBx) is highly associated with the development of HCC. Although Notch1 signaling has been found to exert a tumor-suppressive function during HCC development, the mechanism of interaction between HBx expression and Notch1 signaling needs to be explored. In this study, we report that HBx expression in hepatic and hepatoma cells resulted in decreased endogenous protein levels of Notch1 intracellular domain (ICN1) and messenger RNA levels of its downstream target genes. These effects were due to a reduction of Notch1 cleavage by HBx through the suppression of presenilin1 (Psen1) transcription rather than inhibition of Notch1 transcription or its ligands' expression. Through transient HBx expression, decreased ICN1 resulted in enhanced cell proliferation, induced G1-S cell cycle progression, and blunted cellular senescence in vitro. Furthermore, the effect of blunted senescence-like growth arrest by stable HBx expression through suppression of ICN1 was shown in a nude mouse xenograft transplantation model. The correlation of inhibited Psen1-dependent Notch1 signaling and blunted senescence-like growth arrest was also observed in HBV-associated HCC patient tumor samples. CONCLUSION: Our results reveal a novel function of HBx in blunting senescence-like growth arrest by decreasing Notch1 signaling, which could be a putative molecular mechanism mediating HBV-associated hepatocarcinogenesis.


Assuntos
Carcinoma Hepatocelular/virologia , Transformação Celular Viral , Vírus da Hepatite B/metabolismo , Neoplasias Hepáticas/virologia , Receptor Notch1/metabolismo , Transativadores/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/metabolismo , Presenilina-1/genética , Transcrição Gênica , Proteínas Virais Reguladoras e Acessórias
12.
Cancer Lett ; 513: 90-100, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-33984420

RESUMO

The tumor-initiating cell (TIC) marker CD133 promotes TIC self-renewal and tumorigenesis through the tyrosine phosphorylation of its c-terminal domain. Therefore, finding compounds that target the phosphorylation of CD133 will provide an effective method for inhibiting TICs characteristics. Here, through small molecule microarray screening, compound LDN193189 was found to bind to the c-terminus of CD133 and influenced its tyrosine phosphorylation. LDN193189 inhibited the interaction between CD133 and p85, accompanied by a reduction in the self-renewal and tumorigenicity of liver TIC. In addition, LDN193189 inhibited the expression and transcription of Galectin-3 by reducing the tyrosine phosphorylation of CD133. Galectin-3 secreted by liver TICs inhibited the proliferation of activated CD8+ T cells by binding to PD-1. LDN193189 suppressed the immune escape ability of liver TICs by downregulating Galectin-3. Taken together, LDN193189 suppressed the tumorigenesis and immune escape of liver CSCs by targeting the CD133-Galectin-3 axis.


Assuntos
Antígeno AC133/metabolismo , Células-Tronco Neoplásicas/metabolismo , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Evasão Tumoral/fisiologia , Carcinogênese , Linhagem Celular Tumoral , Humanos , Pirazóis/farmacologia , Pirimidinas/farmacologia
13.
FEBS Lett ; 595(17): 2290-2302, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34328657

RESUMO

Sialic acid-binding immunoglobulin-like lectin-15 (Siglec-15) has been identified as a novel potential target for cancer immunotherapy. Here, we explored the role of Siglec-15 in human hepatoma cells. In this study, we found that the expression of Siglec-15 is substantially upregulated in liver cancer tissues in comparison with the nontumor tissues. Functionally, in vitro experiments show that Siglec-15 promotes the migration of hepatoma cells. Furthermore, the data demonstrated an interaction between Siglec-15 and CD44, a transmembrane glycoprotein that mediates tumor progression and metastasis. In addition, we show that CD44 is modified by α2,6-linked sialic acids on N-glycans in hepatoma cells and that CD44 sialylation affects its interaction with Siglec-15. Removal of the sialic acid residues from CD44 resulted in suppressed interaction between Siglec-15 and CD44. We further demonstrate that Siglec-15 interacts and promotes the stability of CD44 by preventing its lysosomal-mediated degradation. Taken together, our findings demonstrate that Siglec-15 promotes the migration of hepatoma cells by regulating the CD44 protein stability.


Assuntos
Carcinoma Hepatocelular/patologia , Receptores de Hialuronatos/metabolismo , Imunoglobulinas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/genética , Imunoglobulinas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/genética , Estabilidade Proteica , Microambiente Tumoral
14.
J Cell Biochem ; 109(5): 1013-24, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20069563

RESUMO

The androgen-signaling pathway plays critical roles in normal prostate development, benign prostatic hyperplasia, established prostate cancer, and in prostate carcinogenesis. In this study, we report that trihydrophobin 1 (TH1) is a potent negative regulator to attenuate the androgen signal-transduction cascade through promoting androgen receptor (AR) degradation. TH1 interacts with AR both in vitro and in vivo, decreases the stability of AR, and promotes AR ubiquitination in a ligand-independent manner. TH1 also associates with AR at the active androgen-responsive prostate-specific antigen (PSA) promoter in the nucleus of LNCaP cells. Decrease of endogenous AR protein by TH1 interferes with androgen-induced luciferase reporter expression and reduces endogenous PSA expression. Taken together, these results indicate that TH1 is a novel regulator to control the duration and magnitude of androgen signal transduction and might be directly involved in androgen-related developmental, physiological, and pathological processes.


Assuntos
Androgênios/metabolismo , Proteínas de Transporte/metabolismo , Processamento de Proteína Pós-Traducional , Receptores Androgênicos/metabolismo , Transdução de Sinais , Animais , Células COS , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Regulação para Baixo , Elementos Facilitadores Genéticos/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Antígeno Prostático Específico/genética , Ligação Proteica , Estabilidade Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição , Ativação Transcricional/genética , Ubiquitinação
15.
Biochem Biophys Res Commun ; 396(3): 602-7, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20417617

RESUMO

Glioma results from unregulated expansion of a self-renewing glioma-initiating cell population. The regulatory pathways which are essential for sustaining the self-renewal of glioma-initiating cells remain largely unknown. Cell surface N-linked oligosaccharides play functional roles in determining cell fate and are associated with glioma malignancy. Previously, we have reported that beta1,4-galactosyltransferase V (beta1,4GalT V) effectively galactosylates the GlcNAcbeta1-->6Man arm of the highly branched N-glycans and positively regulates glioma cell growth. Here, we show that decreasing the expression of beta1,4GalT V by RNA interference in glioma cells attenuated the formation of polylactosamine and inhibited the ability of tumor formation in vivo. Down-regulation of beta1,4GalT V depleted CD133-positive cells in glioma xenograft, and inhibited the self-renewal capacity and the tumorigenic potential of glioma-initiating cells. These data reveal a critical role of beta1,4GalT V in the self-renewal and tumorigenicity of glioma-initiating cells, and indicate that manipulating beta1,4GalT V expression may have therapeutic potential for the treatment of malignant glioma.


Assuntos
Galactosiltransferases/fisiologia , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Amino Açúcares/metabolismo , Animais , Linhagem Celular Tumoral , Galactosiltransferases/genética , Técnicas de Silenciamento de Genes , Glioma/enzimologia , Humanos , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/enzimologia , Polissacarídeos/metabolismo , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Biochem Biophys Res Commun ; 397(4): 711-7, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20537983

RESUMO

Sox2, a master transcription factor, contributes to the generation of induced pluripotent stem cells and plays significant roles in sustaining the self-renewal of neural stem cells and glioma-initiating cells. Understanding the functional differences of Sox2 between glioma-initiating cells and normal neural stem cells would contribute to therapeutic approach for treatment of brain tumors. Here, we first demonstrated that Sox2 could contribute to the self-renewal and proliferation of glioma-initiating cells. The following experiments showed that Sox2 was activated at translational level in a subset of human glioma-initiating cells compared with the normal neural stem cells. Further investigation revealed there was a positive correlation between Sox2 and eukaryotic initiation factor 4E (eIF4E) in glioma tissues. Down-regulation of eIF4E decreased Sox2 protein level without altering its mRNA level in glioma-initiating cells, indicating that Sox2 was activated by eIF4E at translational level. Furthermore, eIF4E was presumed to regulate the expression of Sox2 by its 5' untranslated region (5' UTR) sequence. Our results suggest that the eIF4E-Sox2 axis is a novel mechanism of unregulated self-renewal of glioma-initiating cells, providing a potential therapeutic target for glioma.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Glioma/metabolismo , Biossíntese de Proteínas , Fatores de Transcrição SOXB1/biossíntese , Linhagem Celular Tumoral , Proliferação de Células , Glioma/patologia , Humanos , Fatores de Transcrição SOXB1/genética
17.
Mol Cell Biol ; 27(20): 7125-42, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17698582

RESUMO

Androgen receptor (AR) is essential for the maintenance of the male reproductive systems and is critical for the carcinogenesis of human prostate cancers (PCas). D-type cyclins are closely related to the repression of AR function. It has been well documented that cyclin D1 inhibits AR function through multiple mechanisms, but the mechanism of how cyclin D3 exerts its repressive role in the AR signaling pathway remains to be identified. In the present investigation, we demonstrate that cyclin D3 and the 58-kDa isoform of cyclin-dependent kinase 11 (CDK11p58) repressed AR transcriptional activity as measured by reporter assays of transformed cells and prostate-specific antigen expression in PCa cells. AR, cyclin D3, and CDK11p58 formed a ternary complex in cells and were colocalized in the luminal epithelial layer of the prostate. AR activity is controlled by phosphorylation at specific sites. We found that AR was phosphorylated at Ser-308 by cyclin D3/CDK11p58 in vitro and in vivo, leading to the repressed activity of AR transcriptional activation unit 1 (TAU1). Furthermore, androgen-dependent proliferation of PCa cells was inhibited by cyclin D3/CDK11p58 through AR repression. These data suggest that cyclin D3/CDK11p58 signaling is involved in the negative regulation of AR function.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Regulação da Expressão Gênica , Isoformas de Proteínas/metabolismo , Receptores Androgênicos/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Ciclina D3 , Quinases Ciclina-Dependentes/genética , Ciclinas/genética , Genes Reporter , Humanos , Masculino , Camundongos , Complexos Multiproteicos/metabolismo , Fosforilação , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Interferência de RNA , Receptores Androgênicos/genética , Serina/metabolismo , Transdução de Sinais/fisiologia , Testículo/citologia , Testículo/metabolismo , Transcrição Gênica
18.
Zhonghua Yan Ke Za Zhi ; 46(7): 580-4, 2010 Jul.
Artigo em Zh | MEDLINE | ID: mdl-21054963

RESUMO

OBJECTIVE: To investigate whether hyperglycemia affect the expression of glycosyltransferases and Enzyme-catalyzed glycosylation in the retina of diabetic rat. METHOD: It was an experimental study. RT-PCR was used to analyze the mRNA level of six glycosyltransferases in the retina of streptozocin diabetic rats; Lectin blot assay with RCA-I was performed to investigate the level of Galß 1→4GlcNAc or N-glycans on total retinal glycoproteins. RESULTS: mRNA level of six glycosyltransferases in the retina of streptomycin diabetic rats and of normal rat is: O-linked N-acetylglucosamine transferase (0.1650 ± 0.1160 versus 0.1160 ± 0.036); UDP-Gal: betaGlcNAc beta1,3-galactosyltransferase (0.0186 ± 0.0122 versus 0.0152 ± 0.0047); alpha 1,4-galactosyltransferase (0.0040 ± 0.0040 versus 0.0054 ± 0.0022); mannoside acetylglucosaminyltransferase 1 (0.0228 ± 0.0166 versus 0.0187 ± 0.0050); UDP-glucose ceramide glucosyltransferase (0.0129 ± 0.0096 versus 0.0116 ± 0.0040); UDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase 1 (0.0157 ± 0.0010 versus 0.0081 ± 0.0016). The mRNA level of UDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase 1 was up-regulated in the retina of streptomycin diabetic rats (t = 6.847, P = 0.002). Consistent with this, the level of Galß 1→4GlcNAc of glycoproteins in streptomycin diabetic rat's retina was strengthened compared with that in control rats. CONCLUSIONS: The results from this study showed that Hyperglycemia could up-regulate the expression of UDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase 1 and strengthen the level of Galß 1→4GlcNAc of glycoproteins in the retina of streptomycin diabetic rats. Our initial results will contribute to the research for the relation between the Enzyme-catalyzed Glycosylation and the etiopathogenesis of diabetic retinopathy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/metabolismo , Glicosiltransferases/metabolismo , Retina/metabolismo , Animais , Produtos Finais de Glicação Avançada , Glicosilação , Masculino , Ratos , Ratos Sprague-Dawley
19.
ACS Biomater Sci Eng ; 6(6): 3550-3562, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-33463170

RESUMO

Radiotherapy (RT) is a major treatment method for non-small-cell lung cancer (NSCLC), and development of new treatment modality is now critical to amplify the negative effects of RT on tumors. In this study, we demonstrated a nanoparticle-loaded block copolymer micellar system for cancer hyperthermia treatment (HT) that can be used for synergistic therapy under alternating magnetic field (AMF) and radiation field. Block copolymer micelles (polyethylene glycol-block-polycaprolactone, or PEG-PCL) containing hyaluronic acid (HA) and Mn-Zn ferrite magnetic nanoparticles (MZF) were fabricated via a two-step preparation. HA-modified Mn-Zn ferrite magnetic nanoparticles (MZF-HA) can be enriched in CD44 highly expressing tumor cells, such as A549 (human lung adenocarcinoma cell line), through an active targeting mechanism via receptor-ligand binding of HA and CD44 (HA receptor). MZF can generate thermal energy under an AMF, leading to a local temperature increase to approximately 43 °C at tumor sites for mild HT, and the increased tumor oxygenation can enhance the therapeutic effect of RT. In vitro experiments show that MZF-HA is able to achieve excellent specific targeting performance toward A549 cells with excellent biocompatibility as well as enhanced therapy performance under HT and RT in vitro by apoptosis flow cytometry. In the A549 subcutaneous tumor xenografts model, MRI confirms the enrichment of MZF-HA in tumor, and hypoxia immunohistochemistry analysis (IHC) proved the increased tumor oxygenation after HT. Furthermore, the tumor volume decreases to 49.6% through the combination of HT and RT in comparison with the 58.8% increase of the untreated group. These results suggest that the application of MZF-HA is able to increase the therapeutic effect of RT on A549 and can be used for further clinical NSCLC treatment evaluation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Hipertermia Induzida , Neoplasias Pulmonares , Nanopartículas de Magnetita , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Compostos Férricos , Humanos , Hipertermia , Neoplasias Pulmonares/radioterapia , Zinco
20.
FEBS Lett ; 594(2): 358-366, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31494931

RESUMO

Abnormal alteration of N-glycosylation structure contributes to glioma progression. N-acetylglucosaminyltransferase I (MGAT1) plays an essential role in the conversion of processed high-mannose cores into complex or hybrid N-linked oligosaccharide structures. The function of MGAT1 in glioma development remains largely unknown. Here, we found that the expression of MGAT1 is higher in glioblastoma compared to normal brain tissues. Inhibition of EGFR signalling pathway or serum starvation reduces MGAT1 expression. Knockdown of MGAT1 inhibits glioma cell proliferation and migration. Furthermore, MGAT1 promotes complex N-glycosylation of glucose transporter 1 (Glut1) and increases Glut1 protein levels. In summary, our findings indicate that MGAT1 is highly expressed in glioblastoma and promotes glioma cells at least partly through upregulation of Glut1 protein.


Assuntos
Glioma/genética , Transportador de Glucose Tipo 1/genética , N-Acetilglucosaminiltransferases/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glioma/patologia , Glucose/metabolismo , Glicosilação , Humanos , Oligossacarídeos/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA