Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Shock ; 56(4): 629-638, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33534395

RESUMO

ABSTRACT: Sepsis is the leading cause of acute kidney injury (AKI) in critical care patients. A cornerstone of sepsis-associated AKI is dysregulated inflammation driven by excessive activation of Toll-like receptor 4 (TLR4) pathway. MUC1, a membrane-bound mucin expressed in both epithelial tubular cells and renal macrophages, has been shown to be involved in the regulation of TLRs. Therefore, we hypothesized that MUC1 could mitigate the renal inflammatory response to TLR4 activation. To test this hypothesis, we used a murine model of endotoxin-induced AKI by intraperitoneal injection of LPS. We showed that Muc1-/- mice have a more severe renal dysfunction, an increased activation of the tissular NF-kB pathway and secreted more pro inflammatory cytokines compare to Muc1+/+ mice. By flow cytometry, we observed that the proportion of M1 (pro-inflammatory) macrophages in the kidneys of Muc1-/- mice was significantly increased. In human and murine primary macrophages, we showed that MUC1 is only induced in M1 type macrophages and that macrophages derived from Muc1-/- mice secreted more pro-inflammatory cytokines. Eventually, in HEK293 cells, we showed that MUC1 cytosolic domain (CT) seems necessary for the negative regulation of TLR4 by proximity ligation assay, MUC1-CT is in close relationship with TLR4 and acts as a competitive inhibitor of the recruitment of MYD88. Overall our results support that in the context of endotoxin-induced AKI, MUC1 plays a significant role in controlling disease severity by regulating negatively the TLR4-MD2 axis.


Assuntos
Injúria Renal Aguda/etiologia , Antígeno 96 de Linfócito/fisiologia , Macrófagos/fisiologia , Mucina-1/fisiologia , Receptor 4 Toll-Like/fisiologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Endotoxinas , Feminino , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Tumour Biol ; 39(7): 1010428317707372, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28714373

RESUMO

Renal cell carcinoma, the most common neoplasm of adult kidney, accounts for about 3% of adult malignancies and is usually highly resistant to conventional therapy. MicroRNAs are a class of small non-coding RNAs, which have been previously shown to promote malignant initiation and progression. In this study, we focused our attention on miR-21, a well described oncomiR commonly upregulated in cancer. Using a cohort of 99 primary renal cell carcinoma samples, we showed that miR-21 expression in cancer tissues was higher than in adjacent non-tumor tissues whereas no significant difference was observed with stages, grades, and metastatic outcome. In vitro, miR-21 was also overexpressed in renal carcinoma cell lines compared to HK-2 human proximal tubule epithelial cell line. Moreover, using Boyden chambers and western blot techniques, we also showed that miR-21 overexpression increased migratory, invasive, proliferative, and anti-apoptotic signaling pathways whereas opposite results were observed using an anti-miR-21-based silencing strategy. Finally, we assessed the role of miR-21 in mediating renal cell carcinoma chemoresistance and further showed that miR-21 silencing significantly (1) increased chemosensitivity of paclitaxel, 5-fluorouracil, oxaliplatin, and dovitinib; (2) decreased expression of multi-drug resistance genes; and (4) increased SLC22A1/OCT1, SLC22A2/OCT2, and SLC31A1/CTR1 platinum influx transporter expression. In conclusion, our results showed that miR-21 is a key actor of renal cancer progression and plays an important role in the resistance to chemotherapeutic drugs. In renal cell carcinoma, targeting miR-21 is a potential new therapeutic strategy to improve chemotherapy efficacy and consequently patient outcome.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Proteínas de Transporte de Cátions/biossíntese , MicroRNAs/genética , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Transportador 1 de Cátions Orgânicos/biossíntese , Antagomirs/genética , Apoptose/efeitos dos fármacos , Benzimidazóis/administração & dosagem , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Transportador de Cobre 1 , Resistencia a Medicamentos Antineoplásicos/genética , Fluoruracila/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Transportador 2 de Cátion Orgânico , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Paclitaxel/administração & dosagem , Quinolonas/administração & dosagem , Transdução de Sinais
3.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1336-1349, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28366875

RESUMO

Acute kidney injury (AKI) is characterized by acute tubular necrosis (ATN) which involves mainly proximal tubules. Past AKI is associated with higher risk of chronic kidney disease (CKD). The MUC1 mucin is a large glycoprotein responsible for epithelial protection and locates to convoluted distal tubules and collecting ducts. Since MUC1 activates the epithelial-mesenchymal transition (EMT) in carcinoma cells, we hypothesized that MUC1 could be involved in epithelial tubular cell plasticity, a process that not only accompanies epithelial repair, but also participates into kidney fibrosis, histological substratum of CKD. In cultured human proximal cells and in human kidney allograft biopsies, we observed MUC1 induction in proximal tubules displaying ATN. Transient MUC1 induction localized with mesenchymal and stem-cell markers and was associated in vitro with reduced anoikis. In a mouse ischemia-reperfusion (IR) model, Muc1 expression mitigates severe tubular injury, as WT displayed less ATN than Muc1 KO mice. But, WT mice displayed more severe kidney fibrosis than Muc1 KO 28days after ischemia. Besides, sustained Muc1 expression in WT was associated with less kidney M2 macrophages. Human kidney biopsies performed within the first week (W1) of transplantation in the context of IR showed MUC1 W1 induction associated with EMT markers. Protocol biopsies performed 3months after demonstrated sustained abnormal MUC1 induction in atrophic tubules within kidney fibrosis. Altogether these data showed that sustained abnormal MUC1 induction accompanies failing epithelial repair, chronic inflammation and kidney fibrosis. In conclusion, MUC1 exerts opposite effects during kidney response to IR: first protective and then harmful.


Assuntos
Nefropatias/metabolismo , Túbulos Renais/metabolismo , Mucina-1/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Fibrose , Células HEK293 , Humanos , Nefropatias/genética , Nefropatias/patologia , Túbulos Renais/patologia , Camundongos , Camundongos Knockout , Mucina-1/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
4.
Oncotarget ; 5(3): 754-63, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24504508

RESUMO

MUC1 is a membrane-anchored mucin and its cytoplasmic tail (CT) can interact with many signaling pathways and act as a co-transcription factor to activate genes involved in tumor progression and metastasis. MUC1 is overexpressed in renal cell carcinoma with correlation to prognosis and has been implicated in the hypoxic pathway, the main renal carcinogenetic pathway. In this context, we assessed the effects of MUC1 overexpression on renal cancer cells properties. Using shRNA strategy and/or different MUC1 constructs, we found that MUC1-extracellular domain and MUC1-CT are involved in increase of migration, cell viability, resistance to anoikis and in decrease of cell aggregation in cancer cells. Invasiveness depends only on MUC1-CT. Then, by using siRNA strategy and/or pharmacological inhibitors or peptides, we showed that sheddases ADAM10, ADAM17 and gamma-secretase are necessary for MUC1 C-terminal subunit (MUC1-C) nuclear location and in increase of invasion property. Finally, MUC1 overexpression increases ADAM10/17 protein expression suggesting a positive regulatory loop. In conclusion, we report that MUC1 acts in renal cancer progression and MUC1-C nuclear localization drives invasiveness of cancer cells through a sheddase/gamma secretase dependent pathway. MUC1 appears as a therapeutic target by blocking MUC1 cleavage or nuclear translocation by using pharmacological approach and peptide strategies.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Neoplasias Renais/metabolismo , Mucina-1/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Progressão da Doença , Xenoenxertos , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Camundongos SCID , Mucina-1/genética , Subunidades Proteicas , Transdução de Sinais , Transfecção
5.
Cancer Lett ; 346(2): 225-36, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24384091

RESUMO

MUC1 is overexpressed in human carcinomas. The transcription factor SNAIL can activate epithelial-mesenchymal transition (EMT) in cancer cells. In this study, in renal carcinoma, we demonstrate that (i) MUC1 and SNAIL were overexpressed in human sarcomatoid carcinomas, (ii) SNAIL increased indirectly MUC1 expression, (iii) MUC1 overexpression induced EMT, (iv) MUC1 C-terminal domain (MUC1-C) and ß-catenin increased SNAIL transcriptional activity by interaction with its promoter and (v) blocking MUC1-C nuclear localization decreased Wnt/ß-catenin signaling pathway activation and SNAIL expression. Altogether, our findings demonstrate that MUC1 is an actor in EMT and appears as a new therapeutic target.


Assuntos
Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Mucina-1/biossíntese , Fatores de Transcrição/biossíntese , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Transição Epitelial-Mesenquimal , Células HEK293 , Humanos , Neoplasias Renais/genética , Mucina-1/genética , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Ativação Transcricional , Regulação para Cima , Via de Sinalização Wnt
6.
Cancer Res ; 69(14): 5707-15, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19549898

RESUMO

The hypoxia inducible factor (HIF) signaling pathway is known as the main renal carcinogenetic pathway. MUC1, an O-glycoprotein membrane-bound mucin, is overexpressed in clear renal cell carcinomas (cRCC) with correlation to two major prognostic factors: tumor-node-metastasis stage and nuclear Fürhman grade. We questioned whether there is a direct link between the HIF pathway and MUC1 overexpression in renal tumors. Interestingly, we observed concomitant increase of HIF-1alpha and MUC1 in metastatic cRCC group versus nonmetastatic cRCC group. Using different renal cell models and small interfering RNA assays targeting either HIF-1alpha or YC-1, a HIF-1 pharmacologic inhibitor, we showed induction of MUC1 expression under hypoxia by a HIF-dependent mechanism. Chromatin immunoprecipitation assay showed a direct binding of HIF-1alpha at the MUC1 promoter. In addition, combined site-directed mutagenesis and gel shift assay allowed the identification of two functional putative hypoxia responsive elements at -1488/-1485 and at -1510/-1507 in the promoter. Using a rat kidney model of ischemia/reperfusion, we confirmed in vivo that clamping renal pedicle for 1 hour followed by 2 hours of reperfusion induced increased MUC1 expression. Furthermore, MUC1 knockdown induced significant reduction of invasive and migration properties of renal cancer cells under hypoxia. Altogether, these results show that MUC1 is directly regulated by HIF-1alpha and affects the invasive and migration properties of renal cancer cells. Thus, MUC1 could serve as a potential therapeutic target in cRCC.


Assuntos
Carcinoma de Células Renais/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/patologia , Mucina-1/metabolismo , Animais , Western Blotting , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Hipóxia Celular , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Imunoprecipitação da Cromatina , Progressão da Doença , Humanos , Imuno-Histoquímica , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Rim/irrigação sanguínea , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Mucina-1/genética , Estadiamento de Neoplasias , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Ratos , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
7.
Surgery ; 143(1): 58-71, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18154934

RESUMO

BACKGROUND: In esophageal adenocarcinoma, MUC1 mucin expression increases in early stages of the carcinogenetic sequence, during which bile reflux has been identified as a major carcinogen. However, no link between MUC1 overexpression and the presence of bile acids in the reflux has been established so far, and molecular mechanisms regulating MUC1 expression during esophageal carcinogenetic sequence are unknown. Our aim was to identify (1) the bile acids able to upregulate MUC1 expression in esophageal cancer cells and mucosal samples, (2) the regulatory regions in MUC1 promoter responsive to bile acids, and (3) the signaling pathway(s) involved in this regulation. METHODS: MUC1 mRNA and mucin expression were studied by the means of real-time reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry, both in the human esophageal OE33 adenocarcinoma cell line and in an ex vivo explant model. MUC1 promoter was cloned and transcription regulation was studied by transient cell transfection to identify the bile acid-responsive regions. Signaling pathways involved were identified using specific pharmacologic inhibitors and siRNA approach. RESULTS: Taurocholic, taurodeoxycholic, taurochenodeoxycholic, glycocholic, sodium glycocholate, and deoxycholic bile acids upregulated MUC1 mRNA and protein expression. The highest induction was obtained with deoxycholic and taurocholic acids in both cellular and explant models. The bile acid-mediated upregulation of MUC1 transcription occurs at the promoter level, with responsive elements located in the -1472/-234 region of the promoter, and involves the phosphatidylinositol 3-kinase signaling pathway. CONCLUSIONS: Bile acids induce MUC1 mucin overexpression in human esophageal adenocarcinoma cells and tissues by activating its transcription through a process involving phosphatidylinositol 3-kinase.


Assuntos
Adenocarcinoma/metabolismo , Ácidos e Sais Biliares/farmacologia , Neoplasias Esofágicas/metabolismo , Mucina-1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Ácido Desoxicólico/farmacologia , Neoplasias Esofágicas/patologia , Esôfago/metabolismo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Mucina-5AC , Mucina-1/genética , Mucinas/metabolismo , Mucosa/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Ácido Taurocólico/farmacologia , Transfecção , Regulação para Cima
8.
Carcinogenesis ; 28(11): 2305-12, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17621592

RESUMO

MUC4 is a transmembrane mucin expressed in pancreatic ductal adenocarcinoma (DAC) in contrast to normal pancreas, and is an independent predictor of poor prognosis in patients with invasive DAC. Our aim was therefore to investigate the mechanisms that control MUC4 expression in pancreatic cancer cells. We focused our study on activator protein (AP)-2alpha transcription factor that acts as a tumour suppressor gene in several cancers. In a series of 18 human DAC, using immunohistochemistry, we confirmed that MUC4 was exclusively expressed in cancerous or preneoplastic lesions in 83% of the samples. On the contrary, AP-2 was mainly expressed by non-tumoural ductal cells (61%) or endocrine cells (67%). Moreover, MUC4 and AP-2 were never found co-expressed suggesting an inhibitory role of AP-2alpha in normal ductal cells. In CAPAN-1 and CAPAN-2 cells, transient AP-2alpha over-expression decreased both MUC4 mRNA and apomucin levels by 20-40% by a mechanism involving inhibition of MUC4 promoter. By chromatin immunoprecipitation and gel-shift assays, we demonstrated that this inhibition involved two AP-2 cis-elements located in the -475/-238 region of the promoter. CAPAN-1 clones, which stably over-expressed AP-2alpha, displayed a strong MUC4 down-regulation (-38 to -100%), a significant decrease of both cell proliferation and invasion concomitant to the up-regulation of p27 cyclin-dependent kinase inhibitor. In conclusion, our data provide evidence that AP-2alpha is an important in vivo negative regulator of MUC4 expression in human pancreatic tissue and that AP-2alpha may play a tumour-suppressive role in pancreatic DAC.


Assuntos
Proliferação de Células , Mucinas/metabolismo , Neoplasias Pancreáticas/patologia , Fator de Transcrição AP-2/fisiologia , Imunoprecipitação da Cromatina , Regulação para Baixo , Humanos , Imuno-Histoquímica , Mucina-4 , Mucinas/genética , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas
9.
Biochem J ; 402(1): 81-91, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17037983

RESUMO

MUC4 (mucin 4) is a membrane-bound mucin overexpressed in the early steps of oesophageal carcinogenesis and implicated in tumour progression. We previously showed that bile acids, main components of gastro-oesophageal reflux and tumour promoters, up-regulate MUC4 expression [Mariette, Perrais, Leteurtre, Jonckheere, Hemon, Pigny, Batra, Aubert, Triboulet and Van Seuningen (2004) Biochem. J. 377, 701-708]. HNF (hepatocyte nuclear factor) 1alpha and HNF4alpha transcription factors are known to mediate bile acid effects, and we previously identified cis-elements for these factors in MUC4 distal promoter. Our aim was to demonstrate that these two transcription factors were directly involved in MUC4 activation by bile acids. MUC4, HNF1alpha and HNF4alpha expressions were evaluated by immunohistochemistry in human oesophageal tissues. Our results indicate that MUC4, HNF1alpha and HNF4alpha were co-expressed in oesophageal metaplastic and adenocarcinomatous tissues. Studies at the mRNA, promoter and protein levels indicated that HNF1alpha regulates endogenous MUC4 expression by binding to two cognate cis-elements respectively located at -3332/-3327 and -3040/-3028 in the distal promoter. We also showed by siRNA (small interfering RNA) approach, co-transfection and site-directed mutagenesis that HNF1alpha mediates taurodeoxycholic and taurochenodeoxycholic bile acid activation of endogenous MUC4 expression and transcription in a dose-dependent manner. In conclusion, these results describe a new mechanism of regulation of MUC4 expression by bile acids, in which HNF1alpha is a key mediator. These results bring new insights into MUC4 up-regulation in oesophageal carcinoma associated with bile reflux.


Assuntos
Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Mucinas/genética , Ácido Tauroquenodesoxicólico/farmacologia , Ácido Taurodesoxicólico/farmacologia , Refluxo Biliar/metabolismo , Linhagem Celular Tumoral , Neoplasias Esofágicas/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Imuno-Histoquímica , Mucina-4 , Mucinas/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Transcrição Gênica , Transfecção , Regulação para Cima
10.
Biochem J ; 377(Pt 3): 701-8, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14583090

RESUMO

Abnormal gastro-oesophageal reflux and bile acids have been linked to the presence of Barrett's oesophageal premalignant lesion associated with an increase in mucin-producing goblet cells and MUC4 mucin gene overexpression. However, the molecular mechanisms underlying the regulation of MUC4 by bile acids are unknown. Since total bile is a complex mixture, we undertook to identify which bile acids are responsible for MUC4 up-regulation by using a wide panel of bile acids and their conjugates. MUC4 apomucin expression was studied by immunohistochemistry both in patient biopsies and OE33 oesophageal cancer cell line. MUC4 mRNA levels and promoter regulation were studied by reverse transcriptase-PCR and transient transfection assays respectively. We show that among the bile acids tested, taurocholic, taurodeoxycholic, taurochenodeoxycholic and glycocholic acids and sodium glycocholate are strong activators of MUC4 expression and that this regulation occurs at the transcriptional level. By using specific pharmacological inhibitors of mitogen-activated protein kinase, phosphatidylinositol 3-kinase, protein kinase A and protein kinase C, we demonstrate that bile acid-mediated up-regulation of MUC4 is promoter-specific and mainly involves activation of phosphatidylinositol 3-kinase. This new mechanism of regulation of MUC4 mucin gene points out an important role for bile acids as key molecules in targeting MUC4 overexpression in early stages of oesophageal carcinogenesis.


Assuntos
Adenocarcinoma/genética , Ácidos e Sais Biliares/fisiologia , Neoplasias Esofágicas/genética , Mucinas/genética , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas/fisiologia , Transdução de Sinais/fisiologia , Neoplasias Gástricas/genética , Transcrição Gênica/fisiologia , Adenocarcinoma/enzimologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Ácidos e Sais Biliares/farmacologia , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Ativação Enzimática/fisiologia , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Mucinas Gástricas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Mucina-1/biossíntese , Mucina-4 , Mucinas/biossíntese , Mucosa/química , Mucosa/metabolismo , Fragmentos de Peptídeos/biossíntese , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
11.
J Histochem Cytochem ; 51(3): 349-61, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12588963

RESUMO

Our previous studies on an inhibitor of O-glycosylation of glycoproteins, GalNAcalpha-O-bn, in the model of enterocytic HT-29 cells, have shown at the cellular level an alteration of the normal localization of apical glycoproteins, and at the biochemical level an in situ synthesis and storage of sialylated GalNAcalpha-O-bn oligosaccharides. The purpose of this study was to examine if a relation existed between these two events, using different cell lines. Intracellular storage of GalNAcalpha-O-bn metabolites occurred in HT-29 and CAPAN-1 cells but not in Caco-2 cells. On the other hand, an accumulation of endosomal/lysosomal compartments was observed in HT-29 and CAPAN-1 cells but not in Caco-2 cells. These data focused on a GalNAcalpha-O-bn-derived storage phenotype in HT-29 and CAPAN-1 cells. The apical membrane glycoproteins MUC1 and CEA showed an abnormal localization inside intracytoplasmic vesicles in HT-29 cells, whereas they kept their normal localization in Caco-2 and CAPAN-1 cells. Studies on the glycosylation of these apical glycoproteins showed that GalNAcalpha-O-bn inhibited the glycosylation in a cell-specific manner. The alteration in the apical targeting of glycoproteins, and the appearance of a GalNAcalpha-O-bn-derived storage phenotype are two independent and cell type-specific events. The former depends on the inhibition pattern of the glycosylation of endogenous glycoproteins, whereas the latter is connected to the intracellular accumulation of GalNAcalpha-O-bn metabolites.


Assuntos
Galactose/metabolismo , Glicoproteínas/metabolismo , Antígenos/metabolismo , Antígenos de Neoplasias , Antígeno Carcinoembrionário/metabolismo , Linhagem Celular , Polaridade Celular , Endossomos/metabolismo , Galactose/análogos & derivados , Glicosilação , Humanos , Imuno-Histoquímica , Cinética , Lisossomos/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Mucina-1 , Mucinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA