hes1-protein--human and Adenocarcinoma

hes1-protein--human has been researched along with Adenocarcinoma* in 20 studies

Other Studies

20 other study(ies) available for hes1-protein--human and Adenocarcinoma

ArticleYear
Loss of HES1 expression is associated with extracellular matrix remodeling and tumor immune suppression in KRAS mutant colon adenocarcinomas.
    Scientific reports, 2023, 09-25, Volume: 13, Issue:1

    The loss of HES1, a canonical Notch signaling target, may cooperate with KRAS mutations to remodel the extracellular matrix and to suppress the anti-tumor immune response. While HES1 expression is normal in benign hyperplastic polyps and normal colon tissue, HES1 expression is often lost in sessile serrated adenomas/polyps (SSAs/SSPs) and colorectal cancers (CRCs) such as those right-sided CRCs that commonly harbor BRAF or KRAS mutations. To develop a deeper understanding of interaction between KRAS and HES1 in colorectal carcinogenesis, we selected microsatellite stable (MSS) and KRAS mutant or KRAS wild type CRCs that show aberrant expression of HES1 by immunohistochemistry. By comparing the transcriptional landscapes of microsatellite stable (MSS) CRCs with or without nuclear HES1 expression, we investigated differentially expressed genes and activated pathways. We identified pathways and markers in the extracellular matrix and immune microenvironment that are associated with mutations in KRAS. We found that loss of HES1 expression positively correlated with matrix remodeling and epithelial-mesenchymal transition but negatively correlated with tumor cell proliferation. Furthermore, loss of HES1 expression in KRAS mutant CRCs correlates with a higher M2 macrophage polarization and activation of IL6 and IL10 immunosuppressive signature. Identifying these HES1-related markers may be useful for prognosis stratification and developing treatment for KRAS-mutant CRCs.

    Topics: Adenocarcinoma; Colonic Neoplasms; Extracellular Matrix; Humans; Immunosuppression Therapy; Proto-Oncogene Proteins p21(ras); Transcription Factor HES-1; Tumor Microenvironment

2023
Notch and mTOR Signaling Pathways Promote Human Gastric Cancer Cell Proliferation.
    Neoplasia (New York, N.Y.), 2019, Volume: 21, Issue:7

    Notch pathway signaling is known to promote gastric stem cell proliferation, and constitutive pathway activation induces gastric tumors via mTORC1 activation in mouse genetic models. The purpose of this study was to determine whether human gastric adenocarcinomas are similarly dependent on Notch and mTORC1 signaling for growth. Gene expression profiling of 415 human gastric adenocarcinomas in The Cancer Genome Atlas, and a small set of locally obtained gastric cancers showed enhanced expression of Notch pathway components, including Notch ligands, receptors and downstream target genes. Human gastric adenocarcinoma tissues and chemically induced mouse gastric tumors both exhibited heightened Notch and mTORC1 pathway signaling activity, as evidenced by increased expression of the NOTCH1 receptor signaling fragment NICD, the Notch target HES1, and the mTORC1 target phosphorylated S6 ribosomal protein. Pharmacologic inhibition of either Notch or mTORC1 signaling reduced growth of human gastric cancer cell lines, with combined pathway inhibition causing a further reduction in growth, suggesting that both pathways are activated to promote gastric cancer cell proliferation. Further, mTORC1 signaling was reduced after Notch inhibition suggesting that mTOR is downstream of Notch in gastric cancer cells. Analysis of human gastric organoids derived from paired control and gastric cancer tissues also exhibited reduced growth in culture after Notch or mTOR inhibition. Thus, our studies demonstrate that Notch and mTOR signaling pathways are commonly activated in human gastric cancer to promote cellular proliferation. Targeting these pathways in combination might be an effective therapeutic strategy for gastric cancer treatment.

    Topics: Adenocarcinoma; Animals; Cell Line, Tumor; Cell Proliferation; Female; Gene Expression Regulation, Neoplastic; Humans; Male; Mechanistic Target of Rapamycin Complex 1; Mice; Receptor, Notch1; Receptors, Notch; Signal Transduction; Stomach Neoplasms; TOR Serine-Threonine Kinases; Transcription Factor HES-1

2019
Modification of α2,6-sialylation mediates the invasiveness and tumorigenicity of non-small cell lung cancer cells in vitro and in vivo via Notch1/Hes1/MMPs pathway.
    International journal of cancer, 2018, 11-01, Volume: 143, Issue:9

    The alterations of sialylation on cell surface N-glycans due to overexpression of different sialyltransferases play a vital role in tumorigenesis and tumor progression. The β-galactoside α2-6-sialyltransferase 1 (ST6Gal-I) has been reported to be highly expressed in several cancers, including breast cancer, hepatocellular cancer and colon carcinoma. However, the roles and underlying mechanisms of ST6Gal-I in non-small cell lung cancer (NSCLC) still need to be elucidated. In this study, we determined that mRNA levels of ST3GAL1, ST6GALNAC3 and ST8SIA6 were remarkably reduced in lung cancer tissues and cells, whereas ST6GAL1 level significantly increased. The mRNA, protein and glycan levels of ST6Gal-I were higher in lung cancer tissues and cells. Moreover, down-regulation of ST6Gal-I decreased protein levels of Jagged1, DLL-1, Notch1, Hes1, Hey1, matrix-metalloproteinases (MMPs) and VEGF, and suppressed proliferation, migration and invasion capabilities of A549 and H1299 cells in vitro. In vivo, ST6Gal-I silencing suppressed tumorigenicity of NSCLC cells in athymic nude mice via the Notch1/Hes1/MMPs pathway. In addition, overexpression of Notch1 rescued the reduced growth and metastasis of A549 and H1299 cells resulted by ST6Gal-I silencing. Modification of α2,6-sialylation positively associates with lung cancer progression, thereby indicating that ST6Gal-I may mediate the invasiveness and tumorigenicity of NSCLC cells via the Notch1/Hes1/MMPs pathway both in vitro and in vivo. Thus, our results provide a novel therapeutic approach for blocking metastasis in lung cancer patients.

    Topics: Adenocarcinoma; Adult; Aged; Aged, 80 and over; Animals; Antigens, CD; Apoptosis; Carcinoma, Non-Small-Cell Lung; Cell Movement; Cell Proliferation; Female; Humans; In Vitro Techniques; Lung Neoplasms; Male; Matrix Metalloproteinases; Mice; Mice, Nude; Middle Aged; N-Acetylneuraminic Acid; Neoplasm Invasiveness; Prognosis; Protein Processing, Post-Translational; Receptor, Notch1; Sialyltransferases; Transcription Factor HES-1; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2018
Characterization of Notch Signalling Pathway Members in Normal Prostate, Prostatic Intraepithelial Neoplasia (PIN) and Prostatic Adenocarcinoma.
    Pathology oncology research : POR, 2016, Volume: 22, Issue:1

    Prostate Cancer (PCa) holds the second place in terms of cancer-related mortality rate among men. The Notch signalling pathway regulates the proliferation and differentiation in embryonic and adult tissues and determines the cell fate. The body of knowledge in the present literature is currently controversial about the effect of the Notch pathway on prostatic cancer. Therefore, the present study aimed to examine the immunolocalization and expression levels of Notch1-4, Jagged1-2, Delta, HES1 and HES5 from among the members of the Notch signalling pathway in tissues of normal, prostatic intraepithelial neoplasia (PIN) and malignant prostate. The current study included a sample of 20 patients with localised prostatic adenocarcinoma, 18 patients with high grade PIN (H-PIN) and 18 normal prostatic tissue. Immunolocalisations of Notch1, 2, 3, 4, Jagged1, 2, Delta, HES1 and HES5 were identified through the immunohistochemical method. The findings of the present study showed that all in-scope members of the Notch signalling pathway were localised in PIN structures to a greater extent than in other tissues and from amongst these members, specifically Notch1, Notch4, Jagged1 and HES1 were at more significant levels. Consequently, the findings of the present study may indicate that the Notch signalling pathway can play a role especially in the formation of PIN structures.

    Topics: Adenocarcinoma; Adult; Basic Helix-Loop-Helix Transcription Factors; Biomarkers, Tumor; Calcium-Binding Proteins; Case-Control Studies; Follow-Up Studies; Homeodomain Proteins; Humans; Immunoenzyme Techniques; Intercellular Signaling Peptides and Proteins; Jagged-1 Protein; Male; Membrane Proteins; Middle Aged; Neoplasm Grading; Neoplasm Staging; Prognosis; Prostate; Prostatic Intraepithelial Neoplasia; Prostatic Neoplasms; Receptors, Notch; Serrate-Jagged Proteins; Signal Transduction; Transcription Factor HES-1

2016
Loss of Hes1 expression is associated with poor prognosis in colorectal adenocarcinoma.
    Human pathology, 2016, Volume: 57

    Alterations in the Notch signaling pathway play a role in colorectal cancer (CRC). Hes1, a Notch-induced transcription factor, has recently been reported to show decreased expression by immunohistochemistry in sessile serrated adenomas. Variable staining patterns have been reported in tubular adenomas, and existing data on Hes1 expression in CRC are limited and inconsistent. We therefore sought to investigate the expression of Hes1 by immunohistochemistry in a large and well-characterized cohort of CRC patients to determine clinicopathological associations and prognostic significance. Immunohistochemistry for Hes1 was performed on 2775 consecutive CRCs in tissue microarray format. Hes1 expression was classified into 3 categories: absent, 1302 cases (46.9%); cytoplasmic staining only with loss of nuclear staining, 1002 cases (36.1%); and nuclear with or without cytoplasmic staining, 471 cases (17%). In univariate analysis, loss of nuclear expression of HES1 was significantly associated with older age, female sex, right-sided location, mucinous or medullary histology, higher histological grade, microsatellite instability, BRAFV600E mutation, and larger tumor size. Strong and statistically significant associations with female sex, right-sided location, BRAFV600E mutation, microsatellite instability, and larger size remained in multivariate analysis. Patients with loss of nuclear expression of Hes1 had a significantly worse all-cause 5-year survival in both univariate (P = .002) and multivariate (P = .009) analysis. We conclude that loss of nuclear expression of Hes1 occurs in 83% of CRCs when studied in tissue microarray format and is associated with female sex, right-sided location, BRAFV600E mutation, microsatellite instability, larger tumor size, and significantly worse survival.

    Topics: Adenocarcinoma; Adolescent; Adult; Age Factors; Aged; Aged, 80 and over; Biomarkers, Tumor; Cell Nucleus; Colorectal Neoplasms; Cytoplasm; Down-Regulation; Female; Humans; Immunohistochemistry; Kaplan-Meier Estimate; Male; Microsatellite Instability; Microsatellite Repeats; Middle Aged; Multivariate Analysis; Mutation; Odds Ratio; Predictive Value of Tests; Proportional Hazards Models; Proto-Oncogene Proteins B-raf; Risk Factors; Tissue Array Analysis; Transcription Factor HES-1; Treatment Outcome; Tumor Burden; Young Adult

2016
HES 1 is essential for chemoresistance induced by stellate cells and is associated with poor prognosis in pancreatic cancer.
    Oncology reports, 2015, Volume: 33, Issue:4

    The role of pancreatic stellate cells (PSCs) has been established in many studies. However, the potential mechanism for the chemoresistance induced by PSCs has not been fully elucidated. In the present study, human pancreatic cancer cell lines were directly or indirectly co-cultured with PSCs. The inhibition rate and IC50 values were assessed to determine the ability of chemoresistance. RT-PCR and western blot analysis were used to evaluate Hes 1 and Jagged 1 expression before and after co-culture with PSCs. To determine the relationship between Hes 1 expression and survival in pancreatic cancer patients, Kaplan-Meier survival analysis was performed. PSCs promoted the expression of Hes 1 in both PANC-1 and BxPC-3 cell lines and induced chemoresistance to gemcitabine. A Notch signaling pathway inhibitor (L1790) and Hes 1 siRNA reversed the chemoresistance induced by PSCs. In 72 resected pancreatic cancer patients, high Hes 1 expression was observed in 34 patients with shorter overall and progression-free survival times. In conclusion, Hes 1 is essential for chemoresistance induced by PSCs and is associated with poor prognosis in pancreatic cancer patients. Therapy targeting the Notch signaling pathway may reverse chemoresistance and improve survival in patients with pancreatic cancer.

    Topics: Adenocarcinoma; Aged; Animals; Antimetabolites, Antineoplastic; Basic Helix-Loop-Helix Transcription Factors; Cell Line, Tumor; Coculture Techniques; Culture Media, Conditioned; Deoxycytidine; Disease Progression; Disease-Free Survival; Drug Resistance, Neoplasm; Female; Gemcitabine; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Kaplan-Meier Estimate; Male; Middle Aged; Neoplasm Proteins; Pancreatic Neoplasms; Pancreatic Stellate Cells; Prognosis; Proportional Hazards Models; Rats; Receptors, Notch; RNA Interference; RNA, Small Interfering; Signal Transduction; Transcription Factor HES-1

2015
Insulinoma-Associated Protein 1 Is a Crucial Regulator of Neuroendocrine Differentiation in Lung Cancer.
    The American journal of pathology, 2015, Volume: 185, Issue:12

    Insulinoma-associated protein 1 (INSM1) is expressed exclusively in embryonic developing neuroendocrine (NE) tissues. INSM1 gene expression is specific for small-cell lung cancer (SCLC), along with achaete-scute homolog-like 1 (ASCL1) and several NE molecules, such as chromogranin A, synaptophysin, and neural cell adhesion molecule 1. However, the underlying biological role of INSM1 in lung cancer remains largely unknown. We first showed that surgically resected SCLC samples specifically expressed INSM1. Forced expression of the INSM1 gene in adenocarcinoma cell lines (H358 and H1975) induced the expression of ASCL1, brain-2 (BRN2), chromogranin A, synaptophysin, and neural cell adhesion molecule 1; in contrast, knockdown of the INSM1 gene by siRNA in SCLC (H69 and H889) decreased their expression. However, forced/knockdown expression of ASCL1 and BRN2 did not affect INSM1 expression. A chromatin immunoprecipitation study revealed that INSM1 bound to the promoter region of the ASCL1 gene. A xenotransplantation assay using tet-on INSM1 gene-transfected adenocarcinoma cell lines demonstrated that INSM1 induced NE differentiation and growth inhibition. Furthermore, we found that INSM1 was not expressed in non-small-cell lung cancer and some SCLC cell lines expressing Notch1-Hes1. By forced/knockdown expression of Notch1 or Hes1 genes, we revealed that Notch1-Hes1 signaling suppressed INSM1, as well as ASCL1 and BRN2. INSM1, expressed exclusively in SCLC, is a crucial regulator of NE differentiation in SCLCs, and is regulated by the Notch1-Hes1 signaling pathway.

    Topics: Adenocarcinoma; Animals; Apoptosis; Basic Helix-Loop-Helix Transcription Factors; Cell Differentiation; Cell Line, Tumor; Cell Proliferation; Gene Knockdown Techniques; Heterografts; Homeodomain Proteins; Humans; Lung Neoplasms; Mice, Inbred Strains; Neoplasm Proteins; Neoplasm Transplantation; Neuroendocrine Cells; POU Domain Factors; Receptor, Notch1; Repressor Proteins; Signal Transduction; Small Cell Lung Carcinoma; Transcription Factor HES-1

2015
Hes1 is involved in the self-renewal and tumourigenicity of stem-like cancer cells in colon cancer.
    Scientific reports, 2014, Feb-04, Volume: 4

    A small subpopulation of cancer cells with stem cell-like features might be responsible for tumour generation, progression, and chemoresistance. Hes1 influences the maintenance of certain stem cells and progenitor cells and the digestive systems. We found upregulated Hes1 in poorly differentiated cancer samples compared with well-differentiated tumour samples, and most of the adenocarcinomas exhibited significantly higher levels of Hes1 mRNA compared with that observed in matched normal colon samples. Moreover, Hes1 expression was found to be correlated with the expression of stem cell markers in colon cancer samples, and Hes1 upregulates the expression of stemness-related genes in colon cancer cells. In addition, Hes1 enhances the self-renewal properties of the stem-like cells by increasing the sizes of CD133+ cells and SP cells and the ability of tumour sphere formation. Additionally, the Hes1-overexpressing cells formed significantly larger and higher number of colonies, as determined through the colony and the soft agar assays. More importantly, Hes1 enhances the tumourigenicity of colon cancer cell lines in nude mice and exhibits a strong tumour-formation ability at a cell density of 1 × 10(3). Taken together, our data indicate that Hes1 induces stem-like cell self-renewal and increases the number of tumour-initiating cells in colon cancer.

    Topics: AC133 Antigen; Adenocarcinoma; Animals; Antigens, CD; Basic Helix-Loop-Helix Transcription Factors; Cell Transformation, Neoplastic; Colonic Neoplasms; Flow Cytometry; Gene Expression Regulation, Neoplastic; Glycoproteins; HCT116 Cells; Homeodomain Proteins; Humans; Mice; Mice, Nude; Neoplasm Transplantation; Neoplastic Stem Cells; Peptides; RNA, Messenger; Spheroids, Cellular; Transcription Factor HES-1; Transplantation, Heterologous; Tumor Cells, Cultured; Up-Regulation

2014
Inhibition of gamma-secretase activity impedes uterine serous carcinoma growth in a human xenograft model.
    Gynecologic oncology, 2014, Volume: 133, Issue:3

    Uterine serous carcinoma (USC) represents an aggressive subtype of endometrial cancer. We sought to understand Notch pathway activity in USC and determine if pathway inhibition has anti-tumor activity.. Patient USC tissue blocks were obtained and used to correlate clinical outcomes with Notch1 expression. Three established USC cell lines were treated with gamma-secretase inhibitor (GSI) in vitro. Mice harboring cell line derived or patient derived USC xenografts (PDXs) were treated with vehicle, GSI, paclitaxel and carboplatin (P/C), or combination GSI and P/C. Levels of cleaved Notch1 protein and Hes1 mRNA were determined in GSI treated samples. Statistical analysis was performed using the Wilcoxon rank sum and Kaplan-Meier methods.. High nuclear Notch1 protein expression was observed in 58% of USC samples with no correlation with overall survival. GSI induced dose-dependent reductions in cell number and decreased levels of cleaved Notch1 protein and Hes1 mRNA in vitro. Treatment of mice with GSI led to decreased Hes1 mRNA expression in USC xenografts. In addition, GSI impeded tumor growth of cell line xenografts as well as UT1 USC PDXs. When GSI and P/C were combined, synergistic anti-tumor activity was observed in UT1 xenografts.. Notch1 is expressed in a large subset of USC. GSI-mediated Notch pathway inhibition led to both reduced cell numbers in vitro and decreased tumor growth of USC some xenograft models. When combined with conventional chemotherapy, GSI augmented anti-tumor activity in one USC PDX line suggesting that targeting of the Notch signaling pathway is a potential therapeutic strategy for future investigation.

    Topics: Adenocarcinoma; Aged; Amyloid Precursor Protein Secretases; Animals; Basic Helix-Loop-Helix Transcription Factors; Cell Line, Tumor; Cyclic S-Oxides; Female; Homeodomain Proteins; Humans; Mice; Receptor, Notch1; RNA, Messenger; Signal Transduction; Thiadiazoles; Transcription Factor HES-1; Tumor Cells, Cultured; Uterine Neoplasms; Xenograft Model Antitumor Assays

2014
Colocalization of β-catenin with Notch intracellular domain in colon cancer: a possible role of Notch1 signaling in activation of CyclinD1-mediated cell proliferation.
    Molecular and cellular biochemistry, 2014, Volume: 396, Issue:1-2

    The Wnt and Notch1 signaling pathways play major roles in intestinal development and tumorigenesis. Sub-cellular localization of β-catenin has been implicated in colorectal carcinogenesis. However, the β-catenin and Notch intracellular domain (NICD) interaction has to be addressed. Immunohistochemistries of β-catenin, NICD, and dual immunofluorescence of β-catenin and NICD were analyzed in colorectal tissues and HT29 cell line. Moreover, real-time PCR analysis of CyclinD1, Hes1 and MUC2 was done in HT29 cells upon N-[N-(3, 5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) treatment. Dual staining emphasized the strong interaction of β-catenin and NICD in adenoma and adenocarcinoma than in normal tissues. Hes1 transcript levels were decreased 1.5- and 7.1-fold in 12.5 and 25 µM DAPT-treated HT29 cells. CyclinD1 transcript levels decreased 1.2- and 1.6-fold, and MUC2 transcript level increased 4.3- and 7.5-fold in 12.5 and 25 µM DAPT-treated HT29 cells. The results of this study showed that the sub-cellular localization of β-catenin converges with NICD inducing proliferation through the activation of CyclinD1 and Hes1. Moreover, the inhibition of Notch1 signaling by DAPT leads to the arrest of cell proliferation and induces apoptosis leading to the upregulation of MUC2, a secretory cell lineage marker.

    Topics: Adenocarcinoma; Adenoma; Basic Helix-Loop-Helix Transcription Factors; beta Catenin; Cell Proliferation; Colonic Neoplasms; Colorectal Neoplasms; Cyclin D1; Dipeptides; Dose-Response Relationship, Drug; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; HT29 Cells; Humans; Mucin-2; Protein Structure, Tertiary; Receptor, Notch1; Reference Values; Signal Transduction; Transcription Factor HES-1

2014
Notch1-Hes1 signalling axis in the tumourigenesis of biliary neuroendocrine tumours.
    Journal of clinical pathology, 2013, Volume: 66, Issue:5

    Biliary neuroendocrine tumours (NETs) are rare and mostly exist as a component of mixed adenoneuroendocrine carcinomas (MANECs). Although the NET component in biliary MANECs is generally more malignant and clinically more important to the prognosis than the ordinary adenocarcinomatous component, the histogenesis of biliary NET has not been clarified. In this study, the role of the Notch1-Hes1 signalling axis in the histogenesis of biliary NETs was examined.. Immunohistochemistry for Notch1, its ligand Jagged1 and Hes1 was performed using surgical specimens from 11 patients with biliary MANEC. Moreover, after the knock-down of Notch1 mRNA expression in a cholangiocarcinoma cell line, the expression of chromogranin A (a neuroendocrine marker) and Ascl1 (a neuroendocrine-inducing molecule inhibited by activated Hes1) was examined by quantitative PCR.. Histological examination revealed that the adenocarcinomatous components were predominately located at the luminal surface of the MANEC and the majority of stromal invasion involved NET components. Ordinary adenocarcinomas and non-neoplastic biliary epithelium constantly expressed Notch1, Jagged1 and Hes1, but the expression of Notch1 and Hes1 was decreased or absent in NET components, suggesting interference with the Notch1-Hes1 signalling axis in biliary NET. Moreover, in the cholangiocarcinoma cell line in which the expression of Notch1 mRNA was knocked down, the mRNA expression of Ascl1 and chromogranin A was increased.. The Notch1-Hes1 signalling axis suppresses neuroendocrine differentiation and maintains tubular/acinar features in adenocarcinoma and non-neoplastic epithelium in the biliary tree. Moreover, a disruption of this signalling axis may be associated with the tumourigenesis of NETs in biliary MANEC.

    Topics: Adenocarcinoma; Basic Helix-Loop-Helix Transcription Factors; Biliary Tract Neoplasms; Carcinoma, Neuroendocrine; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Homeodomain Proteins; Humans; Immunohistochemistry; Receptor, Notch1; Signal Transduction; Transcription Factor HES-1; Transfection

2013
Notch pathway activity identifies cells with cancer stem cell-like properties and correlates with worse survival in lung adenocarcinoma.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2013, Apr-15, Volume: 19, Issue:8

    The cancer stem cell theory postulates that tumors contain a subset of cells with stem cell properties of self-renewal, differentiation, and tumor initiation. The purpose of this study is to determine the role of Notch activity in identifying lung cancer stem cells.. We investigated the role of Notch activity in lung adenocarcinoma using a Notch GFP reporter construct and a γ-secretase inhibitor (GSI), which inhibits Notch pathway activity.. Transduction of lung cancer cells with Notch GFP reporter construct identified a subset of cells with high Notch activity (GFP-bright). GFP-bright cells had the ability to form more tumor spheres in serum-free media and were able to generate both GFP-bright and GFP-dim (lower Notch activity) cell populations. GFP-bright cells were resistant to chemotherapy and were tumorigenic in serial xenotransplantation assays. Tumor xenografts of mice treated with GSI had decreased expression of downstream effectors of Notch pathway and failed to regenerate tumors upon reimplantation in NOD/SCID mice. Using multivariate analysis, we detected a statistically significant correlation between poor clinical outcome and Notch activity (reflected in increased Notch ligand expression or decreased expression of the negative modulators), in a group of 443 patients with lung adenocarcinoma. This correlation was further confirmed in an independent group of 89 patients with adenocarcinoma in which Hes-1 overexpression correlated with poor overall survival.. Notch activity can identify lung cancer stem cell-like population and its inhibition may be an appropriate target for treating lung adenocarcinoma.

    Topics: Adenocarcinoma; Animals; Apoptosis; Basic Helix-Loop-Helix Transcription Factors; Cell Cycle; Cell Line, Tumor; Cyclic S-Oxides; Flow Cytometry; Gene Expression Regulation, Neoplastic; Green Fluorescent Proteins; Homeodomain Proteins; Humans; Immunohistochemistry; Kaplan-Meier Estimate; Lung Neoplasms; Mice; Mice, Inbred NOD; Mice, Nude; Mice, SCID; Multivariate Analysis; Neoplastic Stem Cells; Protein Isoforms; Receptors, Notch; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Thiadiazoles; Transcription Factor HES-1; Xenograft Model Antitumor Assays

2013
Aberrant expression and association of VEGF and Dll4/Notch pathway molecules under hypoxia in patients with lung cancer.
    Histology and histopathology, 2013, Volume: 28, Issue:2

    Tumor angiogenesis plays important roles in the pathogenesis and prognosis of lung cancer. Both vascular endothelial growth factor (VEGF) and Dll4/Notch pathways are critical for angiogenesis, whereas their relationship under hypoxia in lung cancer remains unknown. Thus, in the present study, we evaluated the expression of VEGF and Dll4/Notch signaling molecules, and assessed their association with the microvessel density (CD31) and hypoxia (HIF1a) in lung cancer and normal lung tissues using immunohistochemical and Real-time RT-PCR techniques. Then, we investigated the biological function of Dll4 by transfecting Dll4 into HUVECs. In lung cancer tissues, Notch pathway molecules (HES1) and VEGF pathway molecules (VEGFR1 and VEGFR2) were significantly up-regulated, while the ratio of VEGFR1/VEGFR2 was decreased. CD31 and HIF1a were also found to be elevated in lung cancer. VEGFR1 was negatively correlated with Notch1 while positively correlated with Dll4. CD31 was positively correlated with HIF1a but negatively correlated with VEGFR1. Moreover, HIF1a was nearly positively correlated with HES1 in lung cancer tissues. After transfection, Dll4, Notch1 and VEGFR1 were up-regulated while VEGF and VEGFR2 were down-regulated in Dll4-transfected HUVECs compared with controls. Also, our findings suggest that the expression of VEGF and VEGFR2 increased gradually with the disease progression of lung cancer. In summary, VEGF and Notch signaling pathway molecules were overexpressed in lung cancer, which positively correlates with hypoxia (HIF1a) and angiogenesis (CD31). There might be a negative feedback loop between VEGF and Dll4/Notch signaling pathway in lung tumor angiogenesis.

    Topics: Adaptor Proteins, Signal Transducing; Adenocarcinoma; Adenocarcinoma of Lung; Adult; Aged; Aged, 80 and over; Basic Helix-Loop-Helix Transcription Factors; Calcium-Binding Proteins; Carcinoma, Squamous Cell; Cells, Cultured; Disease Progression; Female; Follow-Up Studies; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Hypoxia; Hypoxia-Inducible Factor 1, alpha Subunit; Intercellular Signaling Peptides and Proteins; Lung; Lung Neoplasms; Male; Middle Aged; Neovascularization, Pathologic; Platelet Endothelial Cell Adhesion Molecule-1; Receptors, Notch; Retrospective Studies; Signal Transduction; Transcription Factor HES-1; Umbilical Veins; Up-Regulation; Vascular Endothelial Growth Factor A

2013
Notch signaling pathway and Cdx2 expression in the development of Barrett's esophagus.
    Laboratory investigation; a journal of technical methods and pathology, 2012, Volume: 92, Issue:6

    Cdx2 expression in esophageal stem cells induced by reflux bile acids may be an important factor for development of Barrett's esophagus, whereas Notch signaling is a molecular signaling pathway that plays an important role in the determination of cell differentiation. ATOH1 (a factor associated with Notch signaling) plays an important role in differentiation of stem cells into goblet cells. However, the relationship between the Notch signaling pathway and Cdx2 expression in the development of Barrett's esophagus has not been explored. The aim of this study was to investigate the interrelationship between Notch signaling and Cdx2 in esophageal epithelial cells. The expressions of Cdx2, MUC2, and intracellular signaling molecules related to Notch signaling (Notch1, Hes1, and ATOH1) were examined using real-time polymerase chain reaction (PCR) and immunohistochemical staining with biopsy specimens obtained from esophageal intestinal metaplasia (IM) with goblet cells (IM⁺) and columnar epithelium not accompanied by goblet cells (IM⁻). For in vitro experiments, we employed human esophageal epithelial cell lines (OE33, OE19, and Het-1A). After forced Cdx2 expression by applying a Cdx2 expression vector to the cells, changes in the expressions of Notch1, Hes1, ATOH1, Cdx2, and MUC2 were analyzed by real-time PCR and western blot analysis. Changes in expressions of Notch1, Hes1, ATOH1, Cdx2, and MUC2 in cells were analyzed following stimulation with bile acids in the presence or absence of Cdx2 blocking with Cdx2-siRNA. Suppressed Hes1 and enhanced ATOH1 and MUC2 expressions were identified in IM⁺ specimens. Forced expression of Cdx2 in cells suppressed Hes1, and enhanced ATOH1 and MUC2 expressions, whereas bile acids suppressed Hes1, and enhanced ATOH1, Cdx2, and MUC2 expressions. On the other hand, these effects were blocked by siRNA-based Cdx2 downregulation. Enhanced expression of Cdx2 by stimulation with bile acids may induce intestinal differentiation of esophageal columnar cells by interaction with the Notch signaling pathway.

    Topics: Adenocarcinoma; Aged; Barrett Esophagus; Basic Helix-Loop-Helix Transcription Factors; CDX2 Transcription Factor; Cell Line, Tumor; Cholic Acid; Deoxycholic Acid; Epithelial Cells; Esophageal Neoplasms; Esophagus; Female; Gene Expression; Gene Silencing; Goblet Cells; Homeodomain Proteins; Humans; Male; Metaplasia; Mucin-2; Real-Time Polymerase Chain Reaction; Receptor, Notch1; RNA, Small Interfering; Signal Transduction; Transcription Factor HES-1; Transfection

2012
Chemokine 25-induced signaling suppresses colon cancer invasion and metastasis.
    The Journal of clinical investigation, 2012, Volume: 122, Issue:9

    Chemotactic cytokines (chemokines) can help regulate tumor cell invasion and metastasis. Here, we show that chemokine 25 (CCL25) and its cognate receptor chemokine receptor 9 (CCR9) inhibit colorectal cancer (CRC) invasion and metastasis. We found that CCR9 protein expression levels were highest in colon adenomas and progressively decreased in invasive and metastatic CRCs. CCR9 was expressed in both primary tumor cell cultures and colon-cancer-initiating cell (CCIC) lines derived from early-stage CRCs but not from metastatic CRC. CCL25 stimulated cell proliferation by activating AKT signaling. In vivo, systemically injected CCR9+ early-stage CCICs led to the formation of orthotopic gastrointestinal xenograft tumors. Blocking CCR9 signaling inhibited CRC tumor formation in the native gastrointestinal CCL25+ microenvironment, while increasing extraintestinal tumor incidence. NOTCH signaling, which promotes CRC metastasis, increased extraintestinal tumor frequency by stimulating CCR9 proteasomal degradation. Overall, these data indicate that CCL25 and CCR9 regulate CRC progression and invasion and further demonstrate an appropriate in vivo experimental system to study CRC progression in the native colon microenvironment.

    Topics: Adenocarcinoma; Animals; Basic Helix-Loop-Helix Transcription Factors; Cell Line, Tumor; Chemokines, CC; Chemotaxis; Colorectal Neoplasms; Gene Expression Regulation, Neoplastic; Gene Regulatory Networks; Homeodomain Proteins; Mice; Mice, Inbred NOD; Mice, SCID; Neoplasm Invasiveness; Neoplasm Transplantation; Receptors, CCR; Signal Transduction; Transcription Factor HES-1

2012
Dysfunctional transforming growth factor-β signaling with constitutively active Notch signaling in Barrett's esophageal adenocarcinoma.
    Cancer, 2011, Aug-15, Volume: 117, Issue:16

    Esophageal adenocarcinoma is often considered to arise from a clonal stem-like population of cells, which is potentially responsible for its poor prognosis. Transforming growth factor β (TGF-β) and Notch signaling pathways play important roles in regulating self-renewal of stem cells and cell-fate determination. Both pathways are frequently implicated in gastrointestinal carcinogenesis. However, their contributions to esophageal adenocarcinoma remain unclear.. We evaluated TGF-β and Notch signaling components in normal esophagus, Barrett's esophagus, and adenocarcinoma tissues and cell lines via immunohistochemical analysis and immunoblotting; Hes-1 transcription was assayed using a Hes-1 luciferase reporter.. We observed loss of Smad4 (P<.05) and β2 spectrin (β2SP) (P<.01) in 5/10 Barrett's esophagus and 17/22 adenocarcinoma tissue sections. Concomitantly, dramatically raised levels of Notch signaling components Hes1 and Jagged1 occurred in adenocarcinoma tissues and cell lines compared with normal tissues. In normal esophagus, Oct3/4-positive cells are located in the basal layer (2-3 per cluster), representing a pool of progenitor cells. We observed an expansion of this pool of Oct3/4 positive cells in esophageal adenocarcinoma (15 per cluster). Furthermore, a panel of SOXs proteins documented for stem cell markers exhibit increased expression in tumor cells, indicating expansion of putative cancer stem cells. Finally, we observed growth inhibition in BE3 cells with a γ-secretase inhibitor, but not in SKGT-4 cells. Unlike SKGT-4 cells, BE3 cells have activated Notch signaling with disruption of TGF-β signaling.. Our findings demonstrated a potential therapeutic value for targeted therapy in esophageal adenocarcinoma in the setting of loss of β2SP/TGF-β with concomitant constitutively active Notch signaling.

    Topics: Adenocarcinoma; Barrett Esophagus; Basic Helix-Loop-Helix Transcription Factors; Calcium-Binding Proteins; Cell Line, Tumor; Cell Proliferation; Core Binding Factor Alpha 3 Subunit; Cyclin-Dependent Kinase 4; Esophageal Neoplasms; Esophagus; Homeodomain Proteins; Humans; Intercellular Signaling Peptides and Proteins; Jagged-1 Protein; Membrane Proteins; Octamer Transcription Factor-3; Receptors, Notch; Serrate-Jagged Proteins; Signal Transduction; Transcription Factor HES-1; Transforming Growth Factor beta

2011
Differential Notch1 and Notch2 expression and frequent activation of Notch signaling in gastric cancers.
    Archives of pathology & laboratory medicine, 2011, Volume: 135, Issue:4

    The biologic effects of Notch1 and Notch2 vary with cancer types and their potential role(s) in gastric cancers (GCs) remains largely unknown.. This study aimed to address the previously mentioned issue by checking the expression of Notch1, Notch2, and Notch target gene Hes1 in GCs, premalignant gastric lesions, and noncancerous endoscopic gastric mucosa and by inhibiting Notch signal transduction in GC cells.. The status of Notch1, Notch2, and Hes1 expression in 74 GC surgical specimens, 10 endoscopic samples, and 4 human GC cell lines was evaluated by tissue microarray-based immunohistochemical staining, Western blotting, and reverse transcription-polymerase chain reaction, and the importance of Notch signaling was elucidated by treating 2 GC cell lines with 2 γ-secretase inhibitors.. Notch1 was undetectable in noncancerous gastric mucosa but was expressed with nuclear translocation in 16.7% (4 of 24) of chronic gastritis, 50.0% (9 of 18) of intestinal metaplasia, 54.2% (26 of 48) of intestinal GC, and 23.1% (6 of 26) of diffuse GC, showing distinct differences of Notch1 detection rates between either intestinal metaplasia and chronic gastritis or intestinal GCs and diffuse GCs (P  =  .03; P  =  .005, respectively). Notch2 nuclear translocation frequencies were 10.0% (1 of 10) in noncancerous endoscopic mucosa, 71.4% (30 of 42) in premalignant lesions, and 97.3% (72 of 74) in GC tissues, demonstrating a correlation of Notch2 expression with both intestinal GC and diffuse GC formation (P < .001). The rates of nuclear-Hes1 labeling were 1 of 10 among noncancerous, 42.9% premalignant, and 81.1% cancer tissues, which were closely correlated with Notch2 (P < .001) rather than Notch1 (P  =  .42) nuclear translocation. Only Notch2 was expressed accompanied with Hes1 nuclear labeling in the 4 GC cell lines established from diffuse GC cases. Inhibition of Notch signaling with γ-secretase inhibitors, L-685,458 and DAPT, prevented Hes1 nuclear translocation but neither suppressed growth nor induced cell death.. This study demonstrated a close correlation of Notch2 expression with GC formation and the potential link of Notch1 upregulation with intestinal-like phenotypes of gastric lesions. Although inhibition of Notch activity failed to achieve anti-GC effects, the activated Notch signaling may reflect a potential GC risk.

    Topics: Adenocarcinoma; Basic Helix-Loop-Helix Transcription Factors; Cell Line, Tumor; Gastrectomy; Gastric Mucosa; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Precancerous Conditions; Receptor, Notch1; Receptor, Notch2; RNA, Messenger; Signal Transduction; Stomach Neoplasms; Tissue Array Analysis; Transcription Factor HES-1

2011
Essential role of Notch signaling in apoptosis of human pancreatic tumoral cells mediated by exosomal nanoparticles.
    International journal of cancer, 2009, Sep-01, Volume: 125, Issue:5

    We previously reported that exosomal nanoparticles secreted by human pancreatic tumoral cell lines decrease tumoral cell proliferation through the mitochondria-dependent apoptotic pathway, because of activation of pro-apoptotic phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and of glucose synthase kinase-3beta (GSK-3beta). Interactions between exosomal nanoparticles and cells are thought to involve membrane lipid rafts. However, the underlying mechanism is unknown. Here, we report that the interaction of exosomal nanoparticles with pancreatic cancer cells led to decreased expression of hairy and enhancer-of-split homolog-1 (Hes-1), the intranuclear target of Notch-1 signaling pathway, and to activation of the apoptotic pathway after a cell cycle arrest in G(0)G(1) phase. Strikingly, the expression level of Notch-1 pathway components was critical, because exosomal nanoparticles decreased the proliferation of cells in which these partners are either weakly represented, in differentiated adenocarcinoma cells, or inhibited, in poorly differentiated carcinoma cells, by blocking presenilin in the gamma-secretase complex that regulates the Notch-1 pathway. Overexpression of Notch-1 intracellular domain resulted in the reversion of the cell proliferation inhibition promoted by exosomal nanoparticles. Blocking presenilin unexpectedly resulted in activation of PTEN and GSK-3beta. Conversely, inhibiting either PTEN or GSK-3beta increased Hes-1 expression and partially counteracted the inhibition of proliferation promoted by exosomal nanoparticles, highlighting reciprocal regulations between Notch signaling and PTEN/GSK-3beta. We concluded that interactions of exosomal nanoparticles with target cells, at lipid rafts where Notch-1 pathway partners are localized, hampered the functioning of the Notch-1 survival pathway and activated the apoptotic pathway, which determines tumoral cell fate.

    Topics: Adenocarcinoma; Amyloid Precursor Protein Secretases; Apoptosis; Basic Helix-Loop-Helix Transcription Factors; Blotting, Western; Carbamates; Caspase 3; Cell Differentiation; Cell Proliferation; Dipeptides; Exosomes; Flow Cytometry; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Homeodomain Proteins; Humans; Mitochondria; Nanoparticles; Pancreatic Neoplasms; PTEN Phosphohydrolase; Receptors, Notch; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Transcription Factor HES-1; Tumor Cells, Cultured

2009
Activation of Notch signaling in human colon adenocarcinoma.
    International journal of oncology, 2008, Volume: 33, Issue:6

    Notch and Wnt signaling function together to regulate colonic progenitor cell division and differentiation. Studies in mice have also shown that Notch signaling is required for adenoma formation in response to elevated Wnt-pathway signaling that occurs in the APCMin mouse model of human adenomatous polyposis coli. We therefore used in situ hybridization to analyze expression of Notch ligands, receptors and fringe genes, as well as the Notch target gene, HES1, in human colorectal cancer (CRC). In a small cohort of tumors, JAGGED ligands, NOTCH1, LFNG and HES1 were expressed at levels similar to, or higher than, levels observed in the crypt. To explore the possibility that Notch signaling may play a quantitative role in human CRC we next analyzed HES1 mRNA expression in 130 tumors, each associated with outcome data. The vast majority of these tumors expressed HES1, although at varying levels. Absolute expression levels did not correlate with patient survival. These results establish that JAG ligands and NOTCH1, as well as Notch receptor activation are consistent features of human CRC and support the notion that many of these tumors, like the APCMin mouse, may respond to anti-Notch therapeutic regimes.

    Topics: Adenocarcinoma; Basic Helix-Loop-Helix Transcription Factors; Calcium-Binding Proteins; Cell Differentiation; Colonic Neoplasms; Gene Expression Regulation, Neoplastic; Germany; Glycosyltransferases; Homeodomain Proteins; Humans; In Situ Hybridization; Intercellular Signaling Peptides and Proteins; Jagged-1 Protein; Kaplan-Meier Estimate; Membrane Proteins; Ontario; Prognosis; Receptor, Notch1; Receptors, Notch; Registries; RNA, Messenger; Serrate-Jagged Proteins; Signal Transduction; Transcription Factor HES-1

2008
The Notch pathway in ovarian carcinomas and adenomas.
    British journal of cancer, 2005, Sep-19, Volume: 93, Issue:6

    Elements of the Notch pathway regulate differentiation; we investigated the expression of such elements in epithelial ovarian tumours. A total of 32 ovarian tumour samples (17 adenocarcinomas, three borderline tumours, 12 adenomas), two human ovarian cancer (A2780, OVCAR3), and one ovarian surface (IOSE 144) cell lines were analysed. The expression of Notch pathway elements was assessed by RT-PCR, real-time PCR (Notch 1), and by immunoblots (Notch 1 extracellular domain (EC), HES1). The proliferation and colony formation of A2780 cells were measured after stable transfection with activated Notch 1 (intracellular domain). Jagged 2, Delta-like-1, Manic Fringe, and TSL1 were expressed more frequently in adenocarcinomas whereas Deltex, Mastermind, and Radical Fringe were more frequent in adenomas. Quantitative PCR revealed decreased Notch 1 mRNA in ovarian adenocarcinomas compared with adenomas. The expression of Notch 1-EC protein was similar in benign and malignant tumours. HES1 protein was strongly expressed in 18/19 ovarian cancers and borderline tumours but not in adenomas. Transfecting A2780 cells with active Notch 1-IC resulted in a proliferative and colony formation advantage compared to mock transfected cells. Thus, Notch pathway elements are expressed in ovarian epithelial tumours and some of them are differentially expressed between adenomas and carcinomas. The Notch pathway could be a target for the development of therapies for ovarian cancer.

    Topics: Adenocarcinoma; Adenoma; Adult; Aged; Aged, 80 and over; Basic Helix-Loop-Helix Transcription Factors; Cell Proliferation; Female; Homeodomain Proteins; Humans; Membrane Proteins; Middle Aged; Neoplasms, Glandular and Epithelial; Ovarian Neoplasms; Ovary; Receptors, Notch; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Transcription Factor HES-1; Tumor Cells, Cultured

2005