Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Invest Ophthalmol Vis Sci ; 63(2): 31, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35212722

RESUMO

Purpose: To determine the role of transmembrane mucins in blocking fluorescein ingress to the corneal epithelium and its deficiency in contributing to corneal fluorescein punctate staining. Methods: A dry eye model was established by extirpating lacrimal and Harderian glands in rabbits to correlate the expression of mucins with fluorescein-stained areas on the corneal button using immunofluorescence. Expression of transmembrane mucins was promoted in human corneal epithelial cells (HCECs) by culturing with the mucin-promoting medium (MPM) or diquafosol treatment. Conversely, the expression of mucins was downregulated by knockdown with short hairpin RNA. The role of mucin1 extracellular domain in fluorescein ingress was further verified by overexpression of N-terminally truncated mucin1 in HCECs. Results: In the rabbit dry eye model, the expression level of mucin1 was significantly decreased in superficial corneal epithelial cells where fluorescein punctate staining was observed. Upregulation of mucin1 and mucin16 in HCECs promoted by MPM or by diquafosol treatment impeded intracellular fluorescein ingress. Downregulation of mucin1 and mucin16 enhanced fluorescence ingress in HCECs after fluorescein staining. Overexpression of truncated mucin1 did not alter the fluorescein intensity of fluorescein-stained HCECs, supporting the notion that the ability of mucin1 to block fluorescein ingress was primarily mediated by its extracellular domain. Minimal inherent expression of mucin16 in the rabbit cornea limited the validation of its role in blocking fluorescein ingress in vivo. Conclusion: Transmembrane mucin1 blocks fluorescein ingress in the corneal epithelium, explaining how fluorescein staining is positive when the level of transmembrane mucins is disturbed in dry eyes.


Assuntos
Síndromes do Olho Seco/metabolismo , Fluoresceína/metabolismo , Corantes Fluorescentes/metabolismo , Mucina-1/fisiologia , Animais , Transporte Biológico/fisiologia , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Epitélio Corneano/citologia , Epitélio Corneano/metabolismo , Citometria de Fluxo , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , RNA Interferente Pequeno/genética , Coelhos , Reação em Cadeia da Polimerase em Tempo Real
2.
J Pharmacol Sci ; 147(4): 331-339, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34663515

RESUMO

Mucin 1 (MUC1) is a transmembrane glycoprotein that contributes to the cellular response in hypoxic conditions in different carcinomas. We investigated the gene expression pattern of MUCs (1, 2, 4, 5AC, 5B, 6, 15, 16, and 19) in isogenic primary (HN4 and HN30) and metastatic (HN12 and HN31) head and neck squamous cell carcinoma (HNSCC) cell lines. MUC1 was significantly up-regulated at the mRNA and protein levels in HN12 and HN31 cells, whereas, other MUCs exhibited diverse expression patterns between HNSCC cell lines. Immunohistochemistry demonstrated that MUC1 was exclusively expressed in cancer cells; however, there was no significant correlation between MUC1 expression and malignancy grading. Inducing hypoxia with CoCl2 significantly increased cell viability, MUC1, hypoxia-inducible factor alpha (HIF-1α), and vascular endothelial growth factor A (VEGF-A) expression in HN12 cells, but not HN31 cells. Interestingly, in hypoxia, cell viability, HIF-1α and VEGF-A expression were significantly reduced in MUC1-knockdown HN12 cells. The current report is the first to demonstrate that MUC1 is required in the regulation of hypoxia-related genes in HNSCC cells. Thus, our results suggest that MUC1 modulates the hypoxic effects in HNSCC cells through HIF-1α regulation.


Assuntos
Regulação da Expressão Gênica/genética , Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/genética , Mucina-1/genética , Mucina-1/fisiologia , Transdução de Sinais/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Humanos , Mucina-1/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
BMB Rep ; 54(8): 425-430, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33832550

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces coronavirus disease 2019 (COVID-19) and may increase the risk of adverse outcomes in lung cancer patients. In this study, we investigated the expression and function of mucin 1 (MUC1) after SARS-CoV-2 infection in the lung epithelial cancer cell line Calu-3. MUC1 is a major constituent of the mucus layer in the respiratory tract and contributes to pathogen defense. SARS-CoV-2 infection induced MUC1 C-terminal subunit (MUC1-C) expression in a STAT3 activation-dependent manner. Inhibition of MUC1-C signaling increased apoptosis-related protein levels and reduced proliferation-related protein levels; however, SARS-CoV-2 replication was not affected. Together, these results suggest that increased MUC1-C expression in response to SARS-CoV-2 infection may trigger the growth of lung cancer cells, and COVID-19 may be a risk factor for lung cancer patients. [BMB Reports 2021; 54(8): 425-430].


Assuntos
Adenocarcinoma/patologia , COVID-19/complicações , Neoplasias Pulmonares/patologia , Mucina-1/fisiologia , Adenocarcinoma/complicações , COVID-19/virologia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/complicações , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/fisiologia , Replicação Viral/fisiologia
4.
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
5.
Cancer Res ; 81(4): 1111-1122, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33323379

RESUMO

The Brg/Brahma-associated factor (BAF, mSWI/SNF) chromatin remodeling complex is of importance in development and has been linked to prostate oncogenesis. The oncogenic MUC1-C protein promotes lineage plasticity in the progression of neuroendocrine prostate cancer (NEPC), however, there is no known association between MUC1-C and BAF. We report here that MUC1-C binds directly to the E2F1 transcription factor and that the MUC1-C→E2F1 pathway induces expression of embryonic stem cell-specific BAF (esBAF) components BRG1, ARID1A, BAF60a, BAF155, and BAF170 in castrate-resistant prostate cancer (CRPC) and NEPC cells. In concert with this previously unrecognized pathway, MUC1 was associated with increased expression of E2F1 and esBAF components in NEPC tumors as compared with CRPC, supporting involvement of MUC1-C in activating the E2F1→esBAF pathway with progression to NEPC. MUC1-C formed a nuclear complex with BAF and activated cancer stem cell (CSC) gene signatures and the core pluripotency factor gene network. The MUC1-C→E2F1→BAF pathway was necessary for induction of both the NOTCH1 effector of CSC function and the NANOG pluripotency factor, and collectively, this network drove CSC self-renewal. These findings indicate that MUC1-C promotes NEPC progression by integrating activation of E2F1 and esBAF with induction of NOTCH1, NANOG, and stemness. SIGNIFICANCE: These findings show that MUC1-C, which promotes prostate cancer progression, activates a novel pathway that drives the BAF remodeling complex, induces NOTCH1 and NANOG, and promotes self-renewal of prostate cancer stem cells.


Assuntos
Carcinoma Neuroendócrino , Mucina-1/fisiologia , Complexos Multiproteicos/genética , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/metabolismo , Carcinoma Neuroendócrino/patologia , Autorrenovação Celular/genética , Montagem e Desmontagem da Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Masculino , Complexos Multiproteicos/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
6.
PLoS Pathog ; 15(2): e1007566, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30716138

RESUMO

The cellular invasion machinery of the enteric pathogen Salmonella consists of a type III secretion system (T3SS) with injectable virulence factors that induce uptake by macropinocytosis. Salmonella invasion at the apical surface of intestinal epithelial cells is inefficient, presumably because of a glycosylated barrier formed by transmembrane mucins that prevents T3SS contact with host cells. We observed that Salmonella is capable of apical invasion of intestinal epithelial cells that express the transmembrane mucin MUC1. Knockout of MUC1 in HT29-MTX cells or removal of MUC1 sialic acids by neuraminidase treatment reduced Salmonella apical invasion but did not affect lateral invasion that is not hampered by a defensive barrier. A Salmonella deletion strain lacking the SiiE giant adhesin was unable to invade intestinal epithelial cells through MUC1. SiiE-positive Salmonella closely associated with the MUC1 layer at the apical surface, but invaded Salmonella were negative for the adhesin. Our findings uncover that the transmembrane mucin MUC1 is required for Salmonella SiiE-mediated entry of enterocytes via the apical route.


Assuntos
Adesinas Bacterianas/metabolismo , Mucina-1/fisiologia , Infecções por Salmonella/metabolismo , Proteínas de Bactérias , Linhagem Celular , Elonguina/metabolismo , Enterócitos , Células Epiteliais , Humanos , Mucina-1/genética , Mucina-1/metabolismo , Salmonella enterica/patogenicidade , Salmonella typhimurium/patogenicidade , Fatores de Virulência
7.
Curr Med Chem ; 26(3): 554-563, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28820070

RESUMO

In the course of studying human mucin MUC1, the attitude towards this molecule has been changing time and again. Initially, the list of presumable functions of MUC1 was restricted to protecting and lubricating epithelium. To date, it is assumed to play an important role in cell signaling as well as in all stages of oncogenesis, from malignant cell transformation to tumor dissemination. The story of MUC1 is full of hopes and disappointments. However, the scientific interest to MUC1 has never waned, and the more profoundly it has been investigated, the clearer its hidden potential turned to be disclosed. The therapeutic potential of mucin MUC1 has already been noted by various scientific groups at the early stages of research. Over forty years ago, the first insights into MUC1 functions became a strong ground for considering this molecule as potential target for anticancer therapy. Therefore, this direction of research has always been of particular interest and practical importance. More than 200 papers on MUC1 were published in 2016; the majority of them are dedicated to MUC1-related anticancer diagnostics and therapeutics. Here we review the history of MUC1 studies from the very first attempts to reveal its functions to the ongoing renaissance.


Assuntos
Mucina-1/fisiologia , Animais , Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Humanos , Mucina-1/efeitos dos fármacos
8.
Invest Ophthalmol Vis Sci ; 59(14): DES157-DES162, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30481821

RESUMO

Many studies have revealed that transmembrane mucins, large glycoproteins with heavily glycosylated glycans, are essential for maintaining ocular surface epithelium lubrication and wettability. Recent reports indicate that transmembrane mucins and galectin-3, a chimera type of galectin that binds ß-galactoside in the glycan, play a crucial role in maintaining the epithelial glycocalyx barrier. This review summarizes current evidence regarding the role of galectin-3, the role of the three major transmembrane mucins (i.e., MUC1, MUC4, and MUC16), in the maintenance of ocular surface wettability and transcellular barrier. Pathological mechanisms of glycocalyx barrier disruption and epithelial surface wettability decreases in dry eye disease are also summarized. Lastly, new ophthalmic drugs that target transmembrane mucin are described.


Assuntos
Síndromes do Olho Seco/metabolismo , Glicocálix/metabolismo , Proteínas Sanguíneas , Antígeno Ca-125/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Células Epiteliais/metabolismo , Galectina 3/fisiologia , Galectinas , Humanos , Proteínas de Membrana/fisiologia , Mucina-1/fisiologia , Mucina-4/fisiologia , Molhabilidade
9.
Biochem Soc Trans ; 46(3): 659-668, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29784646

RESUMO

Currently, there is renewed interest in attempting to recruit the host immune system to eliminate cancers, and within this renewed activity, MUC1 continues to arouse interest. MUC1 has been considered a possible therapeutic target for the past 30 years as it is up-regulated, aberrantly glycosylated and its polarization is lost in many adenocarcinomas. Moreover, MUC1 is expressed by some haematopoietic cancers, including acute myeloid leukaemia and myeloma. Although multiple clinical trials have been initiated and immune responses have been documented, effective clinical benefit worthy of approval for general application has not as yet been achieved. However, this does not appear to have quelled the interest in MUC1 as a therapeutic target, as shown by the increase in the number of MUC1-based clinical trials initiated in 2017 ( Figure 1). As with all translational studies, incorporating new relevant research findings into therapeutic strategy is difficult. Decisions are made to commit to a specific strategy based on the information and data available when the trial is initiated. However, the time required for preclinical studies and early trials can render the founding concept not always appropriate for proceeding to a larger definitive trial. Here, we summarize the attempts made, to date, to bring MUC1 into the world of cancer immunotherapy and discuss how research findings regarding MUC1 structure and function together with expanded knowledge of its interactions with the tumour environment and immune effector cells could lead to improved therapeutic approaches. ppbiost;46/3/659/BST20170400CF1F1BST-2017-0400CF1Figure 1.Number of MUC1-targeted trials initiated each year.


Assuntos
Imunoterapia , Mucina-1/imunologia , Neoplasias/terapia , Antimetabólitos Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto , Terapia Combinada , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Glicosilação , Humanos , Mucina-1/química , Mucina-1/fisiologia , Neoplasias/tratamento farmacológico , Microambiente Tumoral , Gencitabina
10.
Oncogene ; 37(16): 2079-2088, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29379165

RESUMO

The PRC2 and PRC1 complexes are aberrantly expressed in human cancers and have been linked to decreases in patient survival. MUC1-C is an oncoprotein that is also overexpressed in diverse human cancers and is associated with a poor prognosis. Recent studies have supported a previously unreported function for MUC1-C in activating PRC2 and PRC1 in cancer cells. In the regulation of PRC2, MUC1-C (i) drives transcription of the EZH2 gene, (ii) binds directly to EZH2, and (iii) enhances occupancy of EZH2 on target gene promoters with an increase in H3K27 trimethylation. Regarding PRC1, which is recruited to PRC2 sites in the hierarchical model, MUC1-C induces BMI1 transcription, forms a complex with BMI1, and promotes H2A ubiquitylation. MUC1-C thereby contributes to the integration of PRC2 and PRC1-mediated repression of tumor suppressor genes, such as CDH1, CDKN2A, PTEN and BRCA1. Like PRC2 and PRC1, MUC1-C is associated with the epithelial-mesenchymal transition (EMT) program, cancer stem cell (CSC) state, and acquisition of anticancer drug resistance. In concert with these observations, targeting MUC1-C downregulates EZH2 and BMI1, inhibits EMT and the CSC state, and reverses drug resistance. These findings emphasize the significance of MUC1-C as a therapeutic target for inhibiting aberrant PRC function and reprogramming the epigenome in human cancers.


Assuntos
Genes Supressores de Tumor , Mucina-1/fisiologia , Neoplasias/genética , Proteínas Oncogênicas/genética , Proteínas do Grupo Polycomb/genética , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 2/genética , Isoformas de Proteínas/fisiologia
11.
Theriogenology ; 108: 339-347, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29288978

RESUMO

Mucin 1 (MUC1), a cell surface glycoprotein, is expressed mainly in the endometrial luminal epithelium (LE) and glandular epithelium (GE) of the endometrium in many mammalian species including mice, rats, pigs, sheep, horses and humans during various stages of a menstrual or oestrous cycle, where it plays an important role in embryo implantation and placentation. However, the expression and function of MUC1 in uterine tissues during early pregnancy in sheep after artificially-induced oestrous is not known. Therefore, we investigated the expression and function of MUC1 in the early pregnant and non-pregnant uterine tissues of sheep with natural oestrous or artificially-induced oestrous on days 10, 12, 14, 16 and 18 of the cycle by in situ hybridization, quantitative real-time polymerase chain reaction, immunohistochemical staining and western blotting methods. According to our results, MUC1 mRNA and protein expression increased initially but then decreased from days 10-18, peaking on day 14 in the uterine tissues of non-pregnant ewes after both natural and artificially-induced oestrous. MUC1 protein localisation was observed in the LE on days 10, 12 and 14 and in the GE on days 16 and 18. In contrast, MUC1 mRNA and protein expression increased on days 10 and 12, decreased on day 14, but increased again on days 16 and 18 in the uterine tissues of pregnant ewes both in natural oestrous and in artificially-induced oestrous. Additionally, the MUC1 mRNA and protein expression levels in the uterine tissues of the early pregnant sheep were significantly lower than those in the non-pregnant sheep on days 10, 14, and 18, except on day 16(P < 0.01). Enhancing MUC1 protein expression with oestrogen or/and progesterone decreased the blastocyst adhesion rate when blastocysts were co-cultured with endometrial epithelial cells (EECs), while inhibiting MUC1 protein expression with IFN-τ increased the blastocyst adhesion rate when the blastocysts were co-cultured with EECs. Compared with the ewes undergoing natural oestrus, the expression trend and regulation of MUC1 did not change, and the MUC1 expression levels only increased under artificial oestrus conditions. Our data provide important information for improving the conception rate in sheep undergoing artificially-induced oestrus and offer some reference points relating to embryo transfer, oestrus synchronisation and superovulation.


Assuntos
Mucina-1/fisiologia , Ovinos/metabolismo , Útero/metabolismo , Animais , Blastocisto/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Transferência Embrionária/veterinária , Estrogênios/farmacologia , Ciclo Estral/genética , Ciclo Estral/metabolismo , Sincronização do Estro , Feminino , Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Mucina-1/genética , Mucina-1/metabolismo , Progesterona/farmacologia , RNA Mensageiro/metabolismo , Ovinos/genética
12.
Carcinogenesis ; 38(6): 604-614, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28472347

RESUMO

Mucin 1 (MUC1) is a tumor antigen that is aberrantly overexpressed in various cancers, including lung cancer. Our previous in vitro studies showed that MUC1 facilitates carcinogen-induced EGFR activation and transformation in human lung bronchial epithelial cells (HBECs), which along with other reports suggests an oncogenic property for MUC1 in lung cancer. However, direct evidence for the role of MUC1 in lung carcinogenesis is lacking. In this study, we used the 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced A/J mouse lung tumor model to investigate the effect of whole-body Muc1 knockout (KO) on carcinogen-induced lung carcinogenesis. Surprisingly, lung tumor multiplicity was significantly increased in Muc1 KO compared to wild-type (WT) mice. The EGFR/AKT pathway was unexpectedly activated, and expression of the EGFR ligand epiregulin (EREG) was increased in the lung tissues of the Muc1 KO compared to the WT mice. EREG stimulated proliferation and protected against cigarette smoke extract (CSE)-induced cytotoxicity in in vitro cultured human bronchial epithelial cells. Additionally, we determined that MUC1 was expressed in human fibroblast cell lines where it suppressed CSE-induced EREG production. Further, suppression of MUC1 cellular activity with GO-201 enhanced EREG production in lung cancer cells, which in turn protected cancer cells from GO-201-induced cell death. Moreover, an inverse association between MUC1 and EREG was detected in human lung cancer, and EREG expression was inversely associated with patient survival. Together, these results support a promiscuous role of MUC1 in lung cancer development that may be related to cell-type specific functions of MUC1 in the tumor microenvironment, and MUC1 deficiency in fibroblasts and malignant cells results in increased EREG production that activates the EGFR pathway for lung carcinogenesis.


Assuntos
Transformação Celular Neoplásica/patologia , Epirregulina/metabolismo , Receptores ErbB/metabolismo , Neoplasias Pulmonares/patologia , Mucina-1/fisiologia , Animais , Carcinógenos/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Epirregulina/genética , Receptores ErbB/genética , Retroalimentação Fisiológica , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrosaminas/toxicidade , Fumar/efeitos adversos
13.
Oncogene ; 36(28): 4037-4046, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28288138

RESUMO

Immunotherapeutic approaches, particularly programmed death 1/programmed death ligand 1 (PD-1/PD-L1) blockade, have improved the treatment of non-small-cell lung cancer (NSCLC), supporting the premise that evasion of immune destruction is of importance for NSCLC progression. However, the signals responsible for upregulation of PD-L1 in NSCLC cells and whether they are integrated with the regulation of other immune-related genes are not known. Mucin 1 (MUC1) is aberrantly overexpressed in NSCLC, activates the nuclear factor-κB (NF-κB) p65→︀ZEB1 pathway and confers a poor prognosis. The present studies demonstrate that MUC1-C activates PD-L1 expression in NSCLC cells. We show that MUC1-C increases NF-κB p65 occupancy on the CD274/PD-L1 promoter and thereby drives CD274 transcription. Moreover, we demonstrate that MUC1-C-induced activation of NF-κB→︀ZEB1 signaling represses the TLR9 (toll-like receptor 9), IFNG, MCP-1 (monocyte chemoattractant protein-1) and GM-CSF genes, and that this signature is associated with decreases in overall survival. In concert with these results, targeting MUC1-C in NSCLC tumors suppresses PD-L1 and induces these effectors of innate and adaptive immunity. These findings support a previously unrecognized central role for MUC1-C in integrating PD-L1 activation with suppression of immune effectors and poor clinical outcome.


Assuntos
Antígeno B7-H1/genética , Carcinoma Pulmonar de Células não Pequenas , Imunidade Celular/genética , Neoplasias Pulmonares , Mucina-1/fisiologia , Evasão Tumoral/genética , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transdução de Sinais/genética , Transdução de Sinais/imunologia
14.
Blood ; 129(13): 1791-1801, 2017 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-28126925

RESUMO

Myeloid-derived suppressor cells (MDSCs) play a critical role in promoting immune tolerance and disease growth. The mechanism by which tumor cells evoke the expansion of MDSCs in acute myeloid leukemia (AML) has not been well described. We have demonstrated that patients with AML exhibit increased presence of MDSCs in their peripheral blood, in comparison with normal controls. Cytogenetic studies demonstrated that MDSCs in patients with AML may be derived from leukemic or apparently normal progenitors. Engraftment of C57BL/6 mice with TIB-49 AML led to an expansion of CD11b+ Gr1+ MDSCs in bone marrow and spleen. Coculture of the AML cell lines MOLM-4, THP-1 or primary AML cells with donor peripheral blood mononuclear cells elicited a cell contact-dependent expansion of MDSCs. MDSCs were suppressive of autologous T-cell responses as evidenced by reduced T-cell proliferation and a switch from a Th1 to a Th2 phenotype. We hypothesized that the expansion of MDSCs in AML is accomplished by tumor-derived extracellular vesicles (EVs). Using tracking studies, we demonstrated that AML EVs are taken-up myeloid progenitor cells, resulting in the selective proliferation of MDSCs in comparison with functionally competent antigen-presenting cells. The MUC1 oncoprotein was subsequently identified as the critical driver of EV-mediated MDSC expansion. MUC1 induces increased expression of c-myc in EVs that induces proliferation in the target MDSC population via downstream effects on cell cycle proteins. Moreover, we demonstrate that the microRNA miR34a acts as the regulatory mechanism by which MUC1 drives c-myc expression in AML cells and EVs.


Assuntos
Proliferação de Células , Leucemia Mieloide Aguda/patologia , Mucina-1/fisiologia , Células Supressoras Mieloides/patologia , Animais , Comunicação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Vesículas Extracelulares/patologia , Xenoenxertos , Humanos , Leucócitos Mononucleares , Camundongos , MicroRNAs/fisiologia , Proteínas Proto-Oncogênicas c-myc/biossíntese
15.
Oncogene ; 35(50): 6439-6445, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27212035

RESUMO

Aberrant expression of the DNA methyltransferases (DNMTs) and disruption of DNA methylation patterns are associated with carcinogenesis and cancer cell survival. The oncogenic MUC1-C protein is aberrantly overexpressed in diverse carcinomas; however, there is no known link between MUC1-C and DNA methylation. Our results demonstrate that MUC1-C induces the expression of DNMT1 and DNMT3b, but not DNMT3a, in breast and other carcinoma cell types. We show that MUC1-C occupies the DNMT1 and DNMT3b promoters in complexes with NF-κB p65 and drives DNMT1 and DNMT3b transcription. In this way, MUC1-C controls global DNA methylation as determined by analysis of LINE-1 repeat elements. The results further demonstrate that targeting MUC1-C downregulates DNA methylation of the CDH1 tumor suppressor gene in association with induction of E-cadherin expression. These findings provide compelling evidence that MUC1-C is of functional importance to induction of DNMT1 and DNMT3b and, in turn, changes in DNA methylation patterns in cancer cells.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA , Mucina-1/fisiologia , Neoplasias/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Transição Epitelial-Mesenquimal , Humanos , Células MCF-7 , Fator de Transcrição RelA/fisiologia , DNA Metiltransferase 3B
16.
J Am Soc Nephrol ; 27(11): 3447-3458, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27036738

RESUMO

Hypercalciuria is a major risk factor for nephrolithiasis. We previously reported that Uromodulin (UMOD) protects against nephrolithiasis by upregulating the renal calcium channel TRPV5. This channel is crucial for calcium reabsorption in the distal convoluted tubule (DCT). Recently, mutations in the gene encoding Mucin-1 (MUC1) were found to cause autosomal dominant tubulointerstitial kidney disease, the same disease caused by UMOD mutations. Because of the similarities between UMOD and MUC1 regarding associated disease phenotype, protein structure, and function as a cellular barrier, we examined whether urinary MUC1 also enhances TRPV5 channel activity and protects against nephrolithiasis. We established a semiquantitative assay for detecting MUC1 in human urine and found that, compared with controls (n=12), patients (n=12) with hypercalciuric nephrolithiasis had significantly decreased levels of urinary MUC1. Immunofluorescence showed MUC1 in the thick ascending limb, DCT, and collecting duct. Applying whole-cell patch-clamp recording of HEK cells, we found that wild-type but not disease mutant MUC1 increased TRPV5 activity by impairing dynamin-2- and caveolin-1-mediated endocytosis of TRPV5. Coimmunoprecipitation confirmed a physical interaction between TRPV5 and MUC1. However, MUC1 did not increase the activity of N-glycan-deficient TRPV5. MUC1 is characterized by variable number tandem repeats (VNTRs) that bind the lectin galectin-3; galectin-3 siRNA but not galectin-1 siRNA prevented MUC1-induced upregulation of TRPV5 activity. Additionally, MUC1 lacking VNTRs did not increase TRPV5 activity. Our results suggest that MUC1 forms a lattice with the N-glycan of TRPV5 via galectin-3, which impairs TRPV5 endocytosis and increases urinary calcium reabsorption.


Assuntos
Mucina-1/fisiologia , Mucina-1/urina , Nefrolitíase/etiologia , Nefrolitíase/urina , Canais de Cátion TRPV/fisiologia , Cálcio/análise , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Regulação para Cima
17.
Am J Respir Cell Mol Biol ; 54(4): 515-23, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26393683

RESUMO

MUC1 (MUC in human; Muc in animals) is a transmembrane mucin glycoprotein expressed in mucosal epithelial cells and hematopoietic cells. MUC1 is involved in the resolution of inflammation during airway Pseudomonas aeruginosa (Pa) infection by suppressing Toll-like receptor signaling in airway epithelial cells. Although alveolar macrophages are recognized as critical mediators of cell-mediated immunity against microorganisms inhaled into the airways, the role of MUC1 in regulating their response is unknown. The aims of this study were to determine whether macrophages express MUC1, and, if so, whether MUC1 expression might be associated with macrophage M0/M1/M2 differentiation or phagocytic activity. Human and mouse MUC1/Muc1 expression was drastically up-regulated in classically activated (M1) macrophages compared with nonactivated (M0) and alternatively activated (M2) macrophages. M1 polarization and Pa stimulation each increased MUC1 ectodomain shedding from the macrophage surface in a TNF-α-converting enzyme-dependent manner. MUC1/Muc1 deficiency in M0 macrophages increased adhesion and phagocytosis of Pa and Escherichia coli compared with MUC1/Muc1-expressing cells, and attenuation of phagocytosis by MUC1 was augmented after polarization into M1 macrophages compared with M0 macrophages. Finally, MUC1/Muc1 deficiency in macrophages increased reactive oxygen species production and TNF-α release in response to Pa compared with MUC1/Muc1-sufficient cells. These results indicate that MUC1/Muc1 expression by macrophages is predominantly in the M1 subtype, and that MUC1/Muc1 expression in these cells decreases their phagocytic activity in an antiinflammatory manner.


Assuntos
Macrófagos/imunologia , Mucina-1/fisiologia , Fagocitose/fisiologia , Animais , Aderência Bacteriana , Ensaio de Imunoadsorção Enzimática , Humanos , Macrófagos/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
18.
Cell Mol Life Sci ; 72(23): 4475-500, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26294353

RESUMO

Mucin 1 (MUC1 [CD227]) is a high-molecular weight (>400 kDa), type I membrane-tethered glycoprotein that is expressed on epithelial cells and extends far above the glycocalyx. MUC1 is overexpressed and aberrantly glycosylated in adenocarcinomas and in hematological malignancies. As a result, MUC1 has been a target for tumor immunotherapeutic studies in mice and in humans. MUC1 has been shown to have anti-adhesive and immunosuppressive properties, protects against infections, and is involved in the oncogenic process as well as in cell signaling. In addition, MUC1 plays a key role in the reproductive tract, in the immune system (affecting dendritic cells, monocytes, T cells, and B cells), and in chronic inflammatory diseases. Evidence for all of these roles for MUC1 is discussed herein and demonstrates that MUC1 is truly a multitasked molecule.


Assuntos
Mucina-1/química , Mucina-1/fisiologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Comunicação Celular , Células Epiteliais/fisiologia , Feminino , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunoterapia/métodos , Leucócitos/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Gravidez , Isoformas de Proteínas
19.
Int J Biol Markers ; 30(3): e294-300, 2015 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25982681

RESUMO

BACKGROUND: The glycoprotein MUC1 is overexpressed and underglycosylated in cancer cells. MUC1 is translated as a single polypeptide that undergoes autocleavage into 2 subunits (the extracellular domain and the cytoplasmic tail), and forms a stable heterodimer at the apical membrane of normal epithelial cells. The MUC1 cytoplasmic tail localizes to the cytoplasm of transformed cells and is targeted to the nucleus. AIMS: To study the expression of the MUC1 extracellular subunit in cell nuclei of neoplastic breast, head and neck, and colon samples. MATERIALS AND METHODS: 330 primary tumor samples were analyzed: 166 invasive breast carcinomas, 127 head and neck tumors, and 47 colon tumors; 10 benign breast disease (BBD) and 40 normal specimens were also included. A standard immunohistochemical method with antigen retrieval was performed. Nuclear fractions from tissue homogenates and breast cancer cell lines (ZR-75, MDA-MB-231, MCF7, and T47D) were obtained and analyzed by Western blotting (WB). The anti-MUC1 extracellular subunit monoclonal antibody HMFG1 was used for immunohistochemistry. RESULTS: 37/166 breast cancer specimens, 5/127 head and neck cancer specimens, 2/47 colon cancer samples, and 3/10 BBD samples showed immunohistochemical staining at the nuclear level. No nuclear reaction was detected in normal samples. By WB, breast and colon cancer purified nuclear fractions showed reactivity at 200 kDa in 3/30 breast and 3/20 colon cancer samples as well as purified nuclear fractions obtained from breast cancer cell lines. CONCLUSIONS: This study shows that the MUC1 extracellular domain might be translocated to the cell nucleus in breast, head and neck, and colon cancer as well as BBD.


Assuntos
Neoplasias da Mama/química , Carcinoma/química , Núcleo Celular/química , Neoplasias do Colo/química , Neoplasias de Cabeça e Pescoço/química , Mucina-1/análise , Proteínas de Neoplasias/análise , Adenocarcinoma/química , Adenocarcinoma/ultraestrutura , Neoplasias da Mama/ultraestrutura , Carcinoma/ultraestrutura , Linhagem Celular Tumoral , Neoplasias do Colo/ultraestrutura , Feminino , Fibroadenoma/química , Fibroadenoma/ultraestrutura , Doença da Mama Fibrocística/metabolismo , Doença da Mama Fibrocística/patologia , Neoplasias de Cabeça e Pescoço/ultraestrutura , Humanos , Hiperplasia , Mucina-1/fisiologia , Proteínas de Neoplasias/fisiologia , Estrutura Terciária de Proteína , Frações Subcelulares/química
20.
Fertil Steril ; 103(4): 1059-1064.e2, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25747132

RESUMO

OBJECTIVE: To evaluate and compare the levels of Mucin 1 (MUC-1) and Glycodelin A (GdA) in precisely timed endometrial biopsies and blood samples taken from women with recurrent implantation failure, and women with proven fertility, in a control group. DESIGN: Molecular studies in human blood and tissue. SETTING: University hospital. PATIENT(S): Women with recurrent implantation failure and women with proven fertility. INTERVENTION(S): Primary endometrial cells and blood samples during the implantation "window" (between day 7 and day 9 after the surge in luteinizing hormone). MAIN OUTCOME MEASURE(S): Expression of MUC-1 and GdA in the human endometrium and in blood during the implantation window were analyzed by enzyme-linked immunosorbent assay. Additionally, MUC-1 and GdA levels in tissue were analyzed by western blot during the same period. RESULT(S): Both blood and tissue measurements of MUC-1 and GdA were significantly lower in women with recurrent implantation failure than in fertile women during the implantation window. In addition, we found a highly significant correlation between blood vs. tissue measurements of both MUC-1 and GdA. CONCLUSION(S): The present study reveals that blood and tissue levels of MUC-1 and GdA are much lower in women with RIF, compared with those in fertile women. Receptivity can be evaluated with noninvasive blood sampling, rather than more-invasive endometrium sampling, as the blood and tissue measurements of MUC-1 and GdA are correlated.


Assuntos
Aborto Habitual/metabolismo , Implantação do Embrião , Glicoproteínas/fisiologia , Infertilidade Feminina/metabolismo , Mucina-1/fisiologia , Aborto Habitual/sangue , Aborto Habitual/patologia , Adulto , Estudos de Casos e Controles , Endométrio/metabolismo , Endométrio/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Glicodelina , Glicoproteínas/sangue , Glicoproteínas/metabolismo , Humanos , Infertilidade Feminina/sangue , Infertilidade Feminina/patologia , Ciclo Menstrual/sangue , Ciclo Menstrual/metabolismo , Mucina-1/sangue , Mucina-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...