Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.712
Filter
1.
Mediators Inflamm ; 2022: 5171525, 2022.
Article in English | MEDLINE | ID: mdl-36091666

ABSTRACT

Inflammation is the body's biological reaction to endogenous and exogenous stimuli. Recent studies have demonstrated several anti-inflammatory properties of Ferula species. In this paper, we decided to study the anti-inflammatory effect of ethanolic extract of Ferula assafoetida oleo-gum-resin (asafoetida) against TNF-α-stimulated human umbilical vein endothelial cells (HUVECs). HUVECs were cultured in a flat-bottom plate and then treated with ethanolic extract of asafoetida (EEA, 0-500 µg/ml) and TNF-α (0-100 ng/ml) for 24 h. We used the MTT test to assess cell survival. In addition, the LC-MS analysis was performed to determine the active substances. HUVECs were pretreated with EEA and then induced by TNF-α. Intracellular reactive oxygen species (ROS) and adhesion of peripheral blood mononuclear cells (PBMCs) to HUVECs were evaluated with DCFH-DA and CFSE fluorescent probes, respectively. Gene expression of intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin and surface expression of ICAM-1 protein were measured using real-time PCR and flow cytometry methods, respectively. While TNF-α significantly increased intracellular ROS formation and PBMC adhesion to TNF-α-induced HUVECs, the pretreatment of HUVECs with EEA (125 and 250 µg/ml) significantly reduced the parameters. In addition, EEA pretreatment decreased TNF-α-induced mRNA expression of VCAM-1 and surface protein expression of ICAM-1 in the target cells. Taken together, the results indicated that EEA prevented ROS generation, triggered by TNF-α, and inhibited the expression of VCAM-1 and ICAM-1, leading to reduced PBMC adhesion. These findings suggest that EEA can probably have anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents , Cell Adhesion Molecules , Ferula , Human Umbilical Vein Endothelial Cells , Plant Extracts , Anti-Inflammatory Agents/pharmacology , Cell Adhesion , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , E-Selectin/biosynthesis , E-Selectin/genetics , E-Selectin/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes, Mononuclear/immunology , Plant Extracts/pharmacology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
2.
J Pathol ; 257(3): 367-378, 2022 07.
Article in English | MEDLINE | ID: mdl-35302657

ABSTRACT

Most high-grade ovarian carcinomas (HGOCs) are sensitive to carboplatin (CBP)-based chemotherapy but frequently recur within 24 months. Recurrent tumors remain CBP-sensitive and acquire resistance only after several treatment rounds. Recurrences arise from a small number of residual tumor cells not amenable to investigation in patients. We developed patient-derived xenografts (PDXs) that allow the study of these different stages of CBP-sensitive recurrence and acquisition of resistance. We generated PDX models from CBP-sensitive and intrinsically resistant HGOC. PDXs were CBP- or mock-treated and tumors were sampled, after treatment and at recurrence. We also isolated models with acquired-resistance from CBP-sensitive PDXs. Tumors were characterized at the histological and transcriptome levels. PDX models reproduced treatment response seen in the patients. CBP-sensitive residual tumors contained nonproliferating tumor cell clusters embedded in a fibrotic mesh. In nontreated PDX tumors and treated CBP-resistant tumors, fibrotic tissue was not prevalent. Residual tumors had marked differences in gene expression when compared to naïve and recurrent tumors, indicating downregulation of the cell cycle and proliferation and upregulation of interferon response and the epithelial-mesenchymal transition. This gene expression pattern resembled that described in embryonal diapause and 'drug-tolerant persister' states. Residual and acquired-resistance tumors share the overexpression of three genes: CEACAM6, CRYAB, and SOX2. Immunostaining analysis showed strong CEACAM6, CRYAB, and SOX2 protein expression in CBP-sensitive residual and acquired-resistance PDX, thus confirming the RNA profiling results. In HGOC PDX, CBP-sensitive recurrences arise from a small population of quiescent, drug-tolerant, residual cells embedded in a fibrotic mesh. These cells overexpress CEACAM6, CRYAB, and SOX2, whose overexpression is also associated with acquired resistance and poor patient prognosis. CEACAM6, CRYAB, and SOX2 may thus serve as a biomarker to predict recurrence and emergence of resistant disease in CBP-treated HGOC patients. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Antigens, CD , Carcinoma, Ovarian Epithelial , Cell Adhesion Molecules , GPI-Linked Proteins , Ovarian Neoplasms , SOXB1 Transcription Factors , alpha-Crystallin B Chain , Antigens, CD/biosynthesis , Antigens, CD/genetics , Carboplatin/pharmacology , Carboplatin/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Drug Resistance, Neoplasm , Female , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Humans , Neoplasm Recurrence, Local , Neoplasm, Residual , Recurrence , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Xenograft Model Antitumor Assays , alpha-Crystallin B Chain/biosynthesis , alpha-Crystallin B Chain/genetics
3.
Clin. transl. oncol. (Print) ; 24(3): 568-577, marzo 2022.
Article in English | IBECS | ID: ibc-203551

ABSTRACT

ObjectivesRecently, the standard of care for advanced urothelial cancer (UC) has been changed by developing immune-checkpoint inhibitors (ICIs). However, its response rate is limited to 20–30%. The identification of biomarkers to predict the therapeutic effects of ICIs is urgently needed. The present study explored the association between immunohistochemical biomarkers and clinical outcomes in UC patients treated with pembrolizumab.Patients and methodsA total of 85 patients with UC who received pembrolizumab after chemotherapy from January 2018 to May 2020 were retrospectively reviewed. Tumor tissues were obtained for immunohistochemical study from 47 out of 85 patients. The protein expressions of PD-L1, WT1, Nectin-4, CD4, CD8, Foxp3, and CD68 in tumor cells and/or tumor infiltrating lymphocytes were immunohistochemically examined. The associations between protein expressions and overall survival (OS), progression-free survival (PFS), and disease control rate (DCR) were statistically analyzed.ResultsPatients with positive PD-L1 in tumor cells showed significantly worse OS (Log-rank test: HR 5.146, p = 0.001, Cox regression analysis: HR 4.331, p = 0.014) and PFS (Log-rank test: HR 3.31. p = 0.022), along with significantly lower DCR (14.3%) compared to the PD-L1 negative patients (67.5%). In addition, patients with strong expression of Nectin-4 in tumor cells showed significantly higher DCR (100%) than the other patients (50%).ConclusionPD-L1 expression in tumor cells was associated with poor prognosis (OS and PFS) and low DCR. Interestingly, the strong expression of Nectin-4 was correlated with high DCR. PD-L1 and Nectin-4 expression in tumor cells could be prognostic biomarkers useful for pembrolizumab in patients with advanced UC.


Subject(s)
Humans , Middle Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/biosynthesis , Cell Adhesion Molecules/biosynthesis , Urologic Neoplasms/drug therapy , Urologic Neoplasms/metabolism , Carcinoma , Retrospective Studies
4.
Cell Cycle ; 21(9): 921-933, 2022 05.
Article in English | MEDLINE | ID: mdl-35220890

ABSTRACT

Graves' disease (GD) is a kind of autoimmune diseases. The development of GD is closely related to the imbalance of Th1/Th2 generated by the differentiation of CD4+ T cells. This study was sought to clarify the role of lncRNA RUNX1-IT1 and explore the mechanism of its function. The expressions of RUNX1-IT1 and Neural cell adhesion molecule (NrCAM) in the peripheral blood of GD patients were detected by qRT-PCR and Western blot. We performed RNA pull down, RIP, and ChIP experiments to verify the correlation between p53 and RUNX1-IT1, p53 and NrCAM. The levels of Th1 cells differentiation markers were detected by Flow cytometry assay and ELISA. The expressions of lncRNA RUNX1-IT1 and NrCAM were most significantly up-regulated in CD4+ T cells of GD patients, and NrCAM expression was significantly positively correlated with RUNX1-IT1 expression. Furthermore, p53 was a potential transcription factor of NrCAM, which could interact with NrCAM. NrCAM level was up-regulated after the overexpression of p53 in CD4+ T cells, while knockdown of RUNX1-IT1 reversed this effect. Down-regulation of NrCAM and RUNX1-IT1 could decrease the mRNA and protein levels of transcriptional regulator T-bet and CXC chemokine ligand 10 (CXCL10) in CD4+ T cells. Our results suggested that RUNX1-IT1 regulated the expressions of the important Th1 factor T-bet, CXCL10, and interferon γ (IFN-γ) by regulating NrCAM transcription, thus participating in the occurrence and development of specific autoimmune disease GD.


Subject(s)
Cell Adhesion Molecules , Graves Disease , RNA, Long Noncoding , Th1 Cells , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chemokines, CXC/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Graves Disease/genetics , Graves Disease/immunology , Graves Disease/metabolism , Graves Disease/pathology , Humans , Neural Cell Adhesion Molecules/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Clin Transl Oncol ; 24(3): 568-577, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34687441

ABSTRACT

OBJECTIVES: Recently, the standard of care for advanced urothelial cancer (UC) has been changed by developing immune-checkpoint inhibitors (ICIs). However, its response rate is limited to 20-30%. The identification of biomarkers to predict the therapeutic effects of ICIs is urgently needed. The present study explored the association between immunohistochemical biomarkers and clinical outcomes in UC patients treated with pembrolizumab. PATIENTS AND METHODS: A total of 85 patients with UC who received pembrolizumab after chemotherapy from January 2018 to May 2020 were retrospectively reviewed. Tumor tissues were obtained for immunohistochemical study from 47 out of 85 patients. The protein expressions of PD-L1, WT1, Nectin-4, CD4, CD8, Foxp3, and CD68 in tumor cells and/or tumor infiltrating lymphocytes were immunohistochemically examined. The associations between protein expressions and overall survival (OS), progression-free survival (PFS), and disease control rate (DCR) were statistically analyzed. RESULTS: Patients with positive PD-L1 in tumor cells showed significantly worse OS (Log-rank test: HR 5.146, p = 0.001, Cox regression analysis: HR 4.331, p = 0.014) and PFS (Log-rank test: HR 3.31. p = 0.022), along with significantly lower DCR (14.3%) compared to the PD-L1 negative patients (67.5%). In addition, patients with strong expression of Nectin-4 in tumor cells showed significantly higher DCR (100%) than the other patients (50%). CONCLUSION: PD-L1 expression in tumor cells was associated with poor prognosis (OS and PFS) and low DCR. Interestingly, the strong expression of Nectin-4 was correlated with high DCR. PD-L1 and Nectin-4 expression in tumor cells could be prognostic biomarkers useful for pembrolizumab in patients with advanced UC.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/biosynthesis , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/metabolism , Cell Adhesion Molecules/biosynthesis , Urologic Neoplasms/drug therapy , Urologic Neoplasms/metabolism , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Retrospective Studies
6.
Sci Rep ; 11(1): 21114, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702952

ABSTRACT

Non-small cell lung cancer (NSCLC) patients with idiopathic pulmonary fibrosis (IPF) show poor prognosis. Periostin is an extracellular matrix protein highly expressed in the lung tissues of IPF. This study aimed to investigate the possibility that periostin secreted by fibroblasts derived from IPF lung might affect proliferation of NSCLC cells. Periostin was more highly expressed and secreted by fibroblasts from diseased human lung with IPF (DIPF) than by normal human lung fibroblasts (NHLF). Cocultivation of NSCLC cells with conditioned media (CM) from DIPF increased proliferation of NSCLC cells through pErk signaling, with this proliferation attenuated by periostin-neutralizing antibodies. Knockdown of integrin ß3, a subunit of the periostin receptor, in NSCLC cells suppressed proliferation of NSCLC cells promoted by recombinant human periostin and CM of DIPF. On in vivo examination, DIPF promoted tumor progression more than NHLF, and knockdown of integrin ß3 in NSCLC cells suppressed tumor progression promoted by DIPF. Fibroblasts derived from surgical specimens from IPF patients also increased secretion of periostin compared to those from non-IPF patients. Periostin secreted from IPF-activated fibroblasts plays critical roles in the proliferation of NSCLC cells. The present study provides a solid basis for considering periostin-targeted therapy for NSCLC patients with IPF.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Adhesion Molecules/biosynthesis , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , A549 Cells , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Adhesion Molecules/genetics , Female , Humans , Idiopathic Pulmonary Fibrosis/genetics , Lung Neoplasms/genetics , Male , Neoplasm Proteins/genetics
7.
Eur J Immunol ; 51(9): 2317-2329, 2021 09.
Article in English | MEDLINE | ID: mdl-34272885

ABSTRACT

Endothelial cells are key regulators of transendothelial migration and their secretion of chemokines and expression of adhesion molecules facilitates lymphocyte entry into tissues. Previously, we demonstrated that Tregs can reduce transendothelial migration of T cells into tumors by decreasing endothelial CXCL10 secretion, but the mechanism by which this occurs is still not known. In this study, we aimed to define how Tregs decrease transendothelial migration into tumors. mRNA sequencing of intestinal tumor endothelial cells from Treg depleted mice identified neutral sphingomyelinase 2 (nSMase2) as a gene downregulated in the presence of Tregs. nSMase2 is expressed in human umbilical vein endothelial cells (HUVECs) and was decreased after coculture with Tregs. Furthermore, blocking of nSMase2 activity in vitro decreased VCAM1, CX3CL1, and CXCL10 expression in HUVECs, mirroring the same decrease found in Treg cocultures. In the APCmin/+ mouse model of intestinal cancer, nSMase2 is lower in tumor endothelial cells than in unaffected small intestine and chronic treatment with a nSMase2 inhibitor suppressed the increased migration that is otherwise seen in the absence of Tregs. We conclude that nSMase2 is an important mediator in endothelial cells supporting transendothelial migration, which may be targeted by Tregs to reduce T-cell migration into tumors.


Subject(s)
Chemokine CXCL10/metabolism , Colonic Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes, Regulatory/immunology , Transendothelial and Transepithelial Migration/physiology , Animals , Cell Adhesion Molecules/biosynthesis , Cell Line , Chemokine CX3CL1/biosynthesis , Chemokine CXCL10/biosynthesis , Colonic Neoplasms/immunology , Down-Regulation , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , Versicans/biosynthesis
8.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34299325

ABSTRACT

Extramammary Paget's disease (EMPD) is a rare skin cancer arising in the apocrine gland-rich areas. Most EMPD tumors are dormant, but metastatic lesions are associated with poor outcomes owing to the lack of effective systemic therapies. Trophoblast cell surface antigen 2 (Trop2), a surface glycoprotein, has drawn attention as a potential therapeutic target for solid tumors. Sacituzumab govitecan, an antibody-drug conjugate of Trop2, has recently entered clinical use for the treatment of various solid cancers. However, little is known about the role of Trop2 in EMPD. In this study, we immunohistochemically examined Trop2 expression in 116 EMPD tissue samples and 10 normal skin tissues. In normal skin, Trop2 was expressed in the epidermal keratinocytes, inner root sheaths, and infundibulum/isthmus epithelium of hair follicles, eccrine/apocrine glands, and sebaceous glands. Most EMPD tissues exhibited homogeneous and strong Trop2 expression, and high Trop2 expression was significantly associated with worse disease-free survival (p = 0.0343). These results suggest the potential use of Trop2-targeted therapy for EMPD and improve our understanding of the skin-related adverse effects of current Trop2-targeted therapies such as sacituzumab govitecan.


Subject(s)
Antigens, Neoplasm/biosynthesis , Cell Adhesion Molecules/biosynthesis , Paget Disease, Extramammary/metabolism , Skin Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apocrine Glands/metabolism , Biomarkers, Tumor , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Hair Follicle/metabolism , Humans , Immunoconjugates/pharmacology , Keratinocytes/metabolism , Male , Membrane Glycoproteins/metabolism , Middle Aged , Paget Disease, Extramammary/drug therapy , Paget Disease, Extramammary/genetics , Paget Disease, Extramammary/pathology , Sebaceous Glands/metabolism , Skin/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology
9.
Exp Biol Med (Maywood) ; 246(23): 2522-2532, 2021 12.
Article in English | MEDLINE | ID: mdl-34308659

ABSTRACT

Cardiovascular disease is the leading cause of morbidity, mortality, and health care costs in the USA, and around the world. Among the various risk factors of cardiovascular disease, environmental and dietary exposures to methyl mercury, a highly toxic metal traditionally labeled as a neurotoxin, have been epidemiologically linked to human cardiovascular disease development. However, its role in development and promotion of atherosclerosis, an initial step in more immediately life-threatening cardiovascular diseases, remains unclear. This study was conducted to examine the role that methyl mercury plays in the adhesion of monocytes to human microvascular endothelial cells (HMEC-1), and the underlying mechanisms. Methyl mercury treatment significantly induced the adhesion of monocyte to HMEC-1 endothelial cells, a critical step in atherosclerosis, while also upregulating the expression of proinflammatory cytokines interleukin-6, interleukin-8. Further, methyl mercury treatment also upregulated the chemotactic cytokine monocyte chemoattractant protein-1 and intercellular adhesion molecule-1. These molecules are imperative for the firm adhesion of leukocytes to endothelial cells. Additionally, our results further demonstrated that methyl mercury stimulated a significant increase in NF-κB activation. These findings suggest that NF-κB signaling pathway activation by methyl mercury is an important factor in the binding of monocytes to endothelial cells. Finally, by using flow cytometric analysis, methyl mercury treatment caused a significant increase in necrotic cell death only at higher concentrations without initiating apoptosis. This study provides new insights into the molecular actions of methyl mercury that can lead to endothelial dysfunction, inflammation, and subsequent atherosclerotic development.


Subject(s)
Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/pathology , Cell Adhesion/drug effects , Endothelial Cells/metabolism , Leukocytes/metabolism , Methylmercury Compounds/toxicity , Atherosclerosis/pathology , Cell Adhesion/physiology , Cell Adhesion Molecules/biosynthesis , Cell Death/drug effects , Cell Line , Chemokine CCL2/metabolism , Endothelial Cells/drug effects , Environmental Exposure/adverse effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Leukocytes/drug effects , NF-kappa B/metabolism
10.
Cells ; 10(6)2021 06 14.
Article in English | MEDLINE | ID: mdl-34198584

ABSTRACT

MAGI1 is a cytoplasmic scaffolding protein initially identified as a component of cell-to-cell contacts stabilizing cadherin-mediated cell-cell adhesion in epithelial and endothelial cells. Clinical-pathological and experimental evidence indicates that MAGI1 expression is decreased in some inflammatory diseases, and also in several cancers, including hepatocellular carcinoma, colorectal, cervical, breast, brain, and gastric cancers and appears to act as a tumor suppressor, modulating the activity of oncogenic pathways such as the PI3K/AKT and the Wnt/ß-catenin pathways. Genomic mutations and other mechanisms such as mechanical stress or inflammation have been described to regulate MAGI1 expression. Intriguingly, in breast and colorectal cancers, MAGI1 expression is induced by non-steroidal anti-inflammatory drugs (NSAIDs), suggesting a role in mediating the tumor suppressive activity of NSAIDs. More recently, MAGI1 was found to localize at mature focal adhesion and to regulate integrin-mediated adhesion and signaling in endothelial cells. Here, we review MAGI1's role as scaffolding protein, recent developments in the understanding of MAGI1 function as tumor suppressor gene, its role in endothelial cells and its implication in cancer and vascular biology. We also discuss outstanding questions about its regulation and potential translational implications in oncology.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Adhesion Molecules , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Guanylate Kinases , Mutation , Neoplasms , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Focal Adhesions/genetics , Focal Adhesions/metabolism , Guanylate Kinases/biosynthesis , Guanylate Kinases/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
11.
Sci Rep ; 11(1): 7344, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795719

ABSTRACT

Nectin-4, upregulated in various cancer cells, cis-interacts with ErbB2 and its trastuzumab-resistant splice variants, p95-ErbB2 and ErbB2∆Ex16, enhancing DNA synthesis through the PI3K-AKT signaling in human breast cancer T47D cells in an adherent culture. We found here that nectin-4 and p95-ErbB2, but not nectin-4 and either ErbB2 or ErbB2∆Ex16, cooperatively enhanced SOX2 gene expression and cell proliferation in a suspension culture. This enhancement of T47D cell proliferation in a suspension culture by nectin-4 and p95-ErbB2 was dependent on the SOX2 gene expression. In T47D cells, nectin-4 and any one of p95-ErbB2, ErbB2, or ErbB2∆Ex16 cooperatively activated the PI3K-AKT signaling, known to induce the SOX2 gene expression, to similar extents. However, only a combination of nectin-4 and p95-ErbB2, but not that of nectin-4 and either ErbB2 or ErbB2∆Ex16, cooperatively enhanced the SOX2 gene expression. Detailed studies revealed that only nectin-4 and p95-ErbB2 cooperatively activated the Hippo signaling. YAP inhibited the SOX2 gene expression in this cell line and thus the MST1/2-LATS1/2 signaling-mediated YAP inactivation increased the SOX2 gene expression. These results indicate that only the combination of nectin-4 and p95-ErbB2, but not that of nectin-4 and either ErbB2 or ErbB2∆Ex16, cooperatively regulates the Hippo signaling-dependent SOX2 gene expression, enhancing anchorage-independent T47D cell proliferation.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion Molecules/biosynthesis , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/biosynthesis , Receptor, ErbB-2/biosynthesis , SOXB1 Transcription Factors/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cytosol/metabolism , Female , Gene Expression Profiling , Hippo Signaling Pathway , Humans , Phosphatidylinositol 3-Kinases/metabolism , Plasmids/metabolism , RNA, Small Interfering/metabolism , Receptor, ErbB-2/chemistry , Signal Transduction
12.
Aging (Albany NY) ; 13(8): 12224-12238, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33882457

ABSTRACT

Unmasking the complex regulatory pathways that mediate the malignant phenotypes of cancer cells can provide novel targets for therapies that could limit the recurrence and metastasis of gastric cancer (GC). Herein, we intended to clarify the role of embryonic ectoderm development protein (EED), microRNA-228-5p (miR-338-5p), methyltransferase like 3 (METTL3) and CUB domain containing protein 1 (CDCP1) in GC. Differentially expressed miRNAs and their target genes were extracted by in silico analysis. The studies revealed high expression of EED in GC tissues and cell lines and it high expression in GC patients was shown to be associated with poor prognosis. The chromatin immunoprecipitation assay identified that EED methylated miR-338-5p to inhibit its expression. EED knockdown could restrain the proliferative and invasive abilities of GC cells by inducing miR-338-5p. Furthermore, miR-338-5p targeted m6A methylase METTL3, while METTL3 amplified the translation of CDCP1 via m6A activity which led to accelerated proliferation and invasion of GC cells. Moreover, in vivo experiments validated that EED promoted the progression of GC through mediating the miR-338-5p/METTL3/CDCP1 axis. Collectively, EED downregulated miR-338-5p through histone methylation, which in turn impaired miR-338-5p-dependent METTL3 inhibition and enhanced CDCP1 translation, therefore contributing to the development of GC.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasm Recurrence, Local/epidemiology , Polycomb Repressive Complex 2/metabolism , Stomach Neoplasms/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Aged , Animals , Antigens, Neoplasm/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease-Free Survival , Female , Follow-Up Studies , Gastrectomy , Gene Knockdown Techniques , Humans , Male , Methylation , Methyltransferases/metabolism , Mice , MicroRNAs/genetics , Middle Aged , Neoplasm Recurrence, Local/genetics , Polycomb Repressive Complex 2/genetics , Prognosis , Protein Biosynthesis/genetics , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Xenograft Model Antitumor Assays
13.
Appl Immunohistochem Mol Morphol ; 29(8): 619-625, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33901032

ABSTRACT

The antibody-drug conjugate enfortumab-vedotin acts by targeting nectin-4, a protein that is nearly ubiquitously expressed in conventional urothelial cancer. However, expression of nectin-4 in morphologic variants of urothelial carcinoma and nonurothelial histotypes was unknown. Immunohistochemistry for nectin-4 using was performed on 169 patients including 83 with nonmuscle invasive bladder cancer and 86 patients with muscle invasive bladder cancer. Staining was scored for intensity (0 to 3) and extent (% positive cells) using the histological score system, where >15 was considered positive. Overall, 72/83 (87%) samples of nonmuscle invasive urothelial carcinoma were positive, including 29/30 (97%) noninvasive papillary urothelial carcinomas, 7/8 (87.5%) carcinomas in situ, 36/45 (80%) papillary urothelial carcinomas invading the lamina propria. Overall, 50/86 muscle invasive tumors were positive, including 15/22 (68.2%) urothelial carcinomas, 7/10 (70%) squamous cell carcinomas, 3/11 (28%) micropapillary tumors, 4/6 (66%) adenocarcinomas, 2/4 (50%) nested carcinomas, 5/8 (63%) plasmacytoid, 1/10 (10%) sarcomatoid carcinomas, and 0/15 (0%) small cell carcinomas. Whole transcriptome RNA sequencing revealed that compared with conventional urothelial carcinomas, most sarcomatoid carcinomas and all but 2 small cell carcinomas expressed very low levels of nectin-4 mRNA but expressed significant levels of either trop2 or ERBB2, which are the molecular targets of 2 other antibody-drug conjugates-sacituzumab gavitecan (trop2) or trastuzumab deruxtecan (ERBB2/HER2). In summary, our study demonstrates that there is heterogeneity of expression of nectin-4 in morphologic variants of urothelial cancer and nonurothelial histotypes, and suggests that testing expression of nectin-4 should be considered in morphologic variants or nonurothelial histotypes found to have lower expression.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , RNA-Seq , Retrospective Studies , Urinary Bladder/pathology , Urinary Bladder Neoplasms/classification , Urinary Bladder Neoplasms/pathology , Urothelium/pathology
14.
BMC Nephrol ; 22(1): 3, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33407253

ABSTRACT

OBJECTIVE: The present study aims to examine the expression of leukocyte adhesion molecules and renal metabolic factors in diabetic mouse kidneys with periodontal pathogen Pg-LPS-induced nephropathy. BACKGROUND: We recently reported that the glomerular endothelium expresses toll-like receptor (TLR)2 and TLR4 in diabetic environments and TLR2/4 ligand Porphyromonas (P.) gingivalis lipopolysaccharides (Pg-LPS) induce nephropathy in diabetic mice. It is thought that Pg-LPS promotes the chronic inflammation with the overexpression of leukocyte adhesion molecules and renal-specific metabolic enzymes by the recognition of Pg-LPS via TLR in the diabetic kidneys. There have been no reports of the effects of periodontopathic bacteria on the expression of leukocyte adhesion molecules and the accumulation of physiologically active substances in the kidney. METHODS: The immunohistochemical investigation was performed on diabetic mouse kidney with Pg-LPS-induced nephropathy with glomerulosclerosis in glomeruli. RESULTS: There were no vessels which expressed vascular cell adhesion molecule-1 (VCAM-1), E-selectin, or fibroblast growth factor (FGF) 23 in streptozotocin (STZ)-induced diabetic ICR mice (STZ-ICR), or in healthy ICR mice administered Pg-LPS (LPS-ICR). However, in diabetic ICR mouse kidneys with Pg-LPS-induced nephropathy (LPS-STZ) the expression of VCAM-1 and the accumulation of FGF23 were observed in renal tubules and glomeruli, and the expression of E-selectin was observed in renal parenchyma and glomeruli. The angiotensin-converting enzyme 2 (ACE2) was detected in the proximal tubules but not in other regions of ICR, STZ-ICR, or LPS-ICR. In LPS-STZ ACE2 was detected both in renal tubules as well as in glomeruli. The Mac-1 and podoplanin-positive cells increased in the renal parenchyma with diabetic condition and there was the distribution of a large number of Mac-1-positive cells in LPS-STZ. CONCLUSIONS: The Pg-LPS may induce diabetic renal inflammation such as glomerulosclerosis and tubulitis with infiltration of Mac-1/podoplanin positive macrophages via glomerular overexpression of VCAM-1 and E-selectin, resulting in accumulation of both ACE2 and FGF23 which were unmetabolized with the inflammation-induced kidney damage under the diabetic condition. Periodontitis may be a critical factor in the progress of nephropathy in diabetic patients.


Subject(s)
Angiotensin-Converting Enzyme 2/biosynthesis , Cell Adhesion Molecules/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Fibroblast Growth Factor-23/biosynthesis , Angiotensin-Converting Enzyme 2/analysis , Animals , Cell Adhesion Molecules/analysis , Diabetes Mellitus, Experimental/etiology , Diabetic Nephropathies/etiology , Fibroblast Growth Factor-23/analysis , Immunohistochemistry , Lipopolysaccharides , Male , Mice , Mice, Inbred ICR , Porphyromonas gingivalis
15.
Invest New Drugs ; 39(2): 304-316, 2021 04.
Article in English | MEDLINE | ID: mdl-32949323

ABSTRACT

Epidermal growth factor-like domain multiple 6 (EGFL6) is implicated in tumor growth, metastasis and angiogenesis, and its ectopic alteration has been detected in aggressive malignancies. However, the pathophysiologic roles and molecular mechanisms of EGFL6 in gastric cancer (GC) remain to be elucidated. In this study, we investigated EGFL6 expression in GC cell lines and tissues using western blotting and immunohistochemistry. We found that EGFL6 was elevated expression in GC cell lines and tissues. The high expression of EGFL6 significantly was correlated with histological grade, depth of invasion, lymph node involvement, distant metastasis and TNM stage in GC and predicted poorer prognosis, and it could act an independent prognostic factor for GC patients. EGFL6 enhanced the proliferation, migration and invasion of GC cells. In addition, we identified the possible molecular mechanisms of EGFL6-involved epithelial-mesenchymal transition (EMT). EGFL6 regulated EMT process and induced metastasis partly through FAK/PI3K/AKT/mTOR, Notch and MAPK signaling pathways. In conclusion, EGFL6 confers an oncogenic function in GC progression and may be proposed as a potential therapeutic target for GC.


Subject(s)
Calcium-Binding Proteins/biosynthesis , Cell Adhesion Molecules/biosynthesis , Epithelial-Mesenchymal Transition/physiology , Stomach Neoplasms/pathology , Cell Line, Tumor , Humans , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Signal Transduction/physiology
16.
J Ethnopharmacol ; 268: 113562, 2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33217519

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Shen-Fu Decoction (SFD), a classic Traditional Chinese paired herb formulation, has been widely used for the treatment of sepsis in China. This study was carried out to assess the effects of SFD in sepsis-induced intestinal permeability and intestinal epithelial tight junction damage in rats with sepsis. MATERIALS AND METHODS: A rat model of sepsis was created by cecal ligation and puncture (CLP). Rats in Sham and CLP + vehicle groups received equal distilled water, while rats in SFD group were treated by gavage of SFD (3 mg/kg, twice a day) for 72h. Mortality, sepsis-induced peritoneal inflammation, intestinal and liver histopathology damage, intestinal permeability (serum FITC-dextran and D-lactate), serum LPS, serum inflammation (PCT, TNF-α, and IL-6), and liver function (AST and ALT) were evaluated. The levels of zonula occluden (ZO-1), Occludin, Claudin-1 were analyzed by Real-time quantitative PCR and Western blotting (WB) respectively. Vasodilator-stimulated phosphoprotein (VASP) and p-VASP in intestinal epithelium were analyzed by WB. RESULTS: Our study showed that SFD markedly reduced the mortality rate of CLP rats, prevented intestine and liver damage, relieved sepsis-induced intestinal permeability and inflammation elevation, ameliorated sepsis-induced impaired intestinal permeability by regulating the expression of ZO-1, Occludin, Claudin-1 and p-VASP. CONCLUSIONS: The herbal formula SFD may be useful for reducing sepsis-induced organic damage and mortality by ameliorating the condition of sepsis-induced intestinal permeability by regulating tight junction proteins and p-VASP.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Drugs, Chinese Herbal/therapeutic use , Intestinal Mucosa/drug effects , Microfilament Proteins/biosynthesis , Phosphoproteins/biosynthesis , Sepsis/drug therapy , Tight Junction Proteins/biosynthesis , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Drugs, Chinese Herbal/pharmacology , Gene Expression , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Microfilament Proteins/antagonists & inhibitors , Permeability/drug effects , Phosphoproteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sepsis/metabolism , Sepsis/pathology , Tight Junction Proteins/antagonists & inhibitors
17.
Technol Cancer Res Treat ; 19: 1533033820977535, 2020.
Article in English | MEDLINE | ID: mdl-33302812

ABSTRACT

Periostin (POSTN) is a protein secreted by mesenchymal cells. Periostin is upregulated in several cancer types and overexpression is associated with poor prognosis. However, the functional role and molecular underpinnings of periostin in epithelial ovarian cancer (EOC) is unknown. In the present study, periostin was found to be significantly upregulated in EOC stroma. Functional studies revealed that periostin could decrease cisplatin (DDP)-induced apoptosis in EOC. Periostin led to DDP resistance in EOC cells, potentially through the PI3K/Akt signaling pathway. We generated periostin-overexpressing fibroblasts and found that EOC cells were resistant to DDP when co-cultured with periostin-overexpressing fibroblasts. The findings of the present study indicated that periostin secreted by cancer-associated stromal cells may be a potential therapeutic target for EOC.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cell Adhesion Molecules/biosynthesis , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Databases, Genetic , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Platinum/pharmacology , Signal Transduction/drug effects
18.
Biomolecules ; 10(11)2020 11 18.
Article in English | MEDLINE | ID: mdl-33218117

ABSTRACT

BACKGROUND: The pathological features of chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) tissues include an eosinophilic infiltration pattern (eosinophilic CRS (ECRS)) or a less eosinophilic pattern (non-ECRS). Recently, it has been suggested that 15-lipoxygenase 1 (15-LOX-1) may have significant roles in allergic disease; however, the significance of 15-LOX-1 in CRS is not well understood. The objective of this study was to demonstrate the expression of 15-LOX-1 in CRS. METHODS: The mRNA expression levels of 15-LOX-1 and periostin in nasal tissues were measured by quantitative real-time polymerase chain reaction. We also performed an immunofluorescence study of nasal tissues. Cells of the Eol-1 eosinophilic leukemic cell line were stimulated with interleukin-33 to test the induction of 15-LOX-1. RESULTS: The expression level of 15-LOX-1 mRNA in nasal polyps (NPs) was significantly higher in ECRS patients than in non-ECRS patients. The immunofluorescence study revealed that both airway epithelial cells and eosinophils in NPs expressed 15-LOX-1. A significant correlation was seen between the number of eosinophils and the mRNA expression levels of 15-LOX-1 and periostin in nasal polyps. Moreover, interleukin-33 enhanced 15-LOX-1 expression in Eol-1 cells. CONCLUSIONS: 15-LOX-1 was shown to be a significant molecule that facilitates eosinophilic inflammation in ECRS.


Subject(s)
Arachidonate 15-Lipoxygenase/biosynthesis , Cell Adhesion Molecules/biosynthesis , Eosinophils/metabolism , Rhinorrhea/metabolism , Sinusitis/metabolism , Adult , Aged , Arachidonate 15-Lipoxygenase/genetics , Cell Adhesion Molecules/genetics , Chronic Disease , Eosinophils/pathology , Female , Gene Expression , Humans , Male , Middle Aged , Rhinorrhea/genetics , Rhinorrhea/pathology , Sinusitis/genetics , Sinusitis/pathology
19.
J Bioenerg Biomembr ; 52(5): 343-354, 2020 10.
Article in English | MEDLINE | ID: mdl-32929607

ABSTRACT

Cell adhesion molecule 1 (CADM1) is frequently silenced in lung, prostate, liver, stomach, pancreatic and breast carcinomas and other forms of human carcinomas. However, it is unclear regarding the role of CADM1 in irritable bowel syndrome with diarrhoea (IBS-D) that is the most common gastrointestinal diagnosis and may contribute to impaired intestinal barrier function. The aim of the present study is to explore the potential mechanism of CADM1 in regulating intestinal barrier function in IBS-D. A rat model with IBS-D induced by the combination method of mother-infant separation, acetic acid and restraint stress was initially established. The defecation frequency, faecal water content (FWC), total intestinal permeability, sIgA, endotoxin, D-lactic acid and diamine oxidase (DAO) were then measured. Next, positive expression of CADM1 protein was detected in distal colonic tissue of rats by immunohistochemistry. The expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in distal colonic mucosa, CADM1, Janus kinase 1 (JAK1), STAT3, p-JAK1, p-STAT3, Claudin-1and Claudin-2 were evaluated using ELISA, RT-qPCR and western blot analysis. IBS-D rats exhibited low CADM1 expression and activated STAT3 signaling pathway. Overexpression of CADM1 in rats was shown to increase Claudin-1 expression, while decreasing expression of STAT3, Claudin-2, TNF-α and IL-6. In addition, silencing of CADM1 or inhibition of the STAT3 signaling pathway was demonstrated to improve the intestinal barrier function. Our study provides evidence that CADM1 can potentially improve intestinal barrier function in rats with IBS-D by inhibiting the STAT3 signaling pathway.


Subject(s)
Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Inflammatory Bowel Diseases/metabolism , STAT3 Transcription Factor/metabolism , Animals , Case-Control Studies , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Disease Models, Animal , Female , HEK293 Cells , HeLa Cells , Humans , Immunoglobulins/biosynthesis , Immunoglobulins/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction
20.
Mol Biol Rep ; 47(10): 8235-8241, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32886326

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is the most frequent histological type of malignancy in the pancreas. Extracellular matrix (ECM), plays a critical role during the process of human carcinogenesis and the possible diversity in matricellular proteins composition of ECM may have a significant impact on the clinical course of PDAC. Aim of this paper was to evaluate the expression of three matricellular proteins, including Periostin (POSTN), Tenascin (TNS) and Osteopontin (OPN), in PDAC from long-survival (LS) and non-long survival (NLS) patients. A total of 30 PDAC were analyzed, 15 from patients that survived more than 60 months after surgery (LS) and 15 that died from the disease within 24 (NLS). RNA was extracted and OPN, TNS and POSTN mRNA levels were evaluated by qRT-PCR. LS and NLS samples showed the same type of POSTN and TN isoforms. On the contrary, OPN seems to be preferentially expressed in NLS PDAC. Moreover, OPNb and OPNc isoforms were expressed exclusively in NLS samples. In conclusion, Our data led to hypothesize a possible relationship between the expression of different isoforms of each of these proteins and the clinical outcome of patients with PDAC.


Subject(s)
Adenocarcinoma , Cell Adhesion Molecules/biosynthesis , Neoplasm Proteins/biosynthesis , Osteopontin/biosynthesis , Pancreatic Neoplasms , Tenascin/biosynthesis , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Adult , Disease-Free Survival , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Pilot Projects , Protein Isoforms/biosynthesis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...