Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 13: 1157461, 2023.
Article in English | MEDLINE | ID: mdl-37182191

ABSTRACT

Introduction: Non-small cell lung carcinomas (NSCLC) exhibit different microvessel patterns (MVPs). Basal (BA), diffuse (DA) and papillary (PA) patterns show signs of angiogenesis (new blood vessels), while an alveolar pattern indicates that tumors are co-opting existing normal vessels (non-angiogenic alveolar, NAA). NAA tumor growth is known to exist in NSCLC, but little is known about its prognostic impact in different histological subgroups, and about associations between MVPs and immune cell infiltration. Methods: Detailed patterns of angiogenic and non-angiogenic tumor growth were evaluated by CD34 immunohistochemistry in whole tissue slides from 553 surgically treated patients with NSCLC stage I-IIIB disease. Associations with clinicopathological variables and markers related to tumor immunology-, angiogenesis- and hypoxia/metabolism were explored, and disease-specific survival (DSS) was analyzed according to histological subtypes. Results: The predominant MVP was angiogenic in 82% of tumors: BA 40%, DA 34%, PA 8%, while a NAA pattern dominated in 18%. A contribution of the NAA pattern >5% (NAA+), i.e., either dominant or minority, was observed in 40.1% of tumors and was associated with poor disease-specific survival (DSS) (p=0.015). When stratified by histology, a significantly decreased DSS for NAA+ was found for adenocarcinomas (LUAD) only (p< 0.003). In multivariate analyses, LUAD NAA+ pattern was a significant independent prognostic factor; HR 2.37 (CI 95%, 1.50-3.73, p< 0.001). The immune cell density (CD3, CD4, CD8, CD45RO, CD204, PD1) added prognostic value in squamous cell carcinoma (LUSC) and LUAD with 0-5% NAA (NAA-), but not in LUAD NAA+. In correlation analyses, there were several significant associations between markers related to tumor metabolism (MCT1, MCT4, GLUT1) and different MVPs. Conclusion: The NAA+ pattern is an independent poor prognostic factor in LUAD. In NAA+ tumors, several immunological markers add prognostic impact in LUSC but not in LUAD.

2.
Cancers (Basel) ; 15(5)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36900240

ABSTRACT

Colon cancer is a common malignancy and a major contributor to human morbidity and mortality. In this study, we explore the expression and prognostic impact of IRS-1, IRS-2, RUNx3, and SMAD4 in colon cancer. Furthermore, we elucidate their correlations with miRs 126, 17-5p, and 20a-5p, which are identified as potential regulators of these proteins. Tumor tissue from 452 patients operated for stage I-III colon cancer was retrospectively collected and assembled into tissue microarrays. Biomarkers' expressions were examined by immunohistochemistry and analyzed using digital pathology. In univariate analyses, high expression levels of IRS1 in stromal cytoplasm, RUNX3 in tumor (nucleus and cytoplasm) and stroma (nucleus and cytoplasm), and SMAD4 in tumor (nucleus and cytoplasm) and stromal cytoplasm were related to increased disease-specific survival (DSS). In multivariate analyses, high expression of IRS1 in stromal cytoplasm, RUNX3 in tumor nucleus and stromal cytoplasm, and high expression of SMAD4 in tumor and stromal cytoplasm remained independent predictors of improved DSS. Surprisingly, with the exception of weak correlations (0.2 < r < 0.25) between miR-126 and SMAD4, the investigated markers were mostly uncorrelated with the miRs. However, weak to moderate/strong correlations (0.3 < r < 0.6) were observed between CD3 and CD8 positive lymphocyte density and stromal RUNX3 expression. High expression levels of IRS1, RUNX3, and SMAD4 are positive prognostic factors in stage I-III colon cancer. Furthermore, stromal expression of RUNX3 is associated with increased lymphocyte density, suggesting that RUNX3 is an important mediator during recruitment and activation of immune cells in colon cancer.

3.
Sci Rep ; 12(1): 7080, 2022 04 30.
Article in English | MEDLINE | ID: mdl-35490164

ABSTRACT

In many types of cancer, microRNAs (miRs) are aberrantly expressed. The aim of this study was to explore the prognostic impact of miR-17-5p and miR-20a-5p in colon cancer. Tumor tissue from 452 stage I-III colon cancer patients was retrospectively collected and tissue microarrays constructed. miR-17-5p and miR-20a-5p expression was evaluated by in situ hybridization and analyzed using digital pathology. Cell line experiments, using HT-29 and CACO-2, were performed to assess the effect of miR-17-5p and miR-20a-5p over expression on viability, invasion and migration. In multivariate analyses, high miR-17-5p expression in tumor (HR = 0.43, CI 0.26-0.71, p < 0.001) and high expression of miR-20a-5p in tumor (HR = 0.60, CI 0.37-0.97, p = 0.037) and stroma (HR = 0.63, CI 0.42-0.95, p = 0.027) remained independent predictors of improved disease-specific survival. In cell lines, over expression of both miRs resulted in mitigated migration without any significant effect on viability or invasion. In conclusion, in stage I-III colon cancer, high expression of both miR-17-5p and miR-20a-5p are independent predictors of favorable prognosis.


Subject(s)
Colonic Neoplasms , MicroRNAs , Caco-2 Cells , Colonic Neoplasms/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Prognosis , Retrospective Studies
4.
Int J Mol Sci ; 22(3)2021 Jan 25.
Article in English | MEDLINE | ID: mdl-33503805

ABSTRACT

Castration-resistant prostate cancer (CRPC) is an advanced and androgen-independent form of prostate cancer. Recent studies of rapid actions mediated by estrogen in the prostate and its relationship with CRPC are emerging. We have previously shown that estrogen receptor (ER) promotes migration and invasion of the androgen-independent prostate cancer cells PC-3, but the signaling pathways involved in these events remain to be elucidated. Therefore, this study aimed to analyze the role of ERα and ERß in the activation of SRC, and the involvement of SRC and PI3K/AKT on invasion and colony formation of the PC-3 cells. Our results showed that the activation of ERα (using ERα-selective agonist PPT) and ERß (using ERß-selective agonist DPN) increased phosphorylation of SRC in PC-3 cells. In the presence of the selective inhibitor for SRC-family kinases PP2, the effects of DPN and PPT on transmigration and soft agar colony formation assays were decreased. Furthermore, SRC is involved in the expression of the non-phosphorylated ß-catenin. Finally, using PI3K specific inhibitor Wortmannin and AKT inhibitor MK2206, we showed that PI3K/AKT are also required for invasion and colony formation of PC-3 cells simulated by ER. This study provides novel insights into molecular mechanisms of ER in PC-3 cells by demonstrating that ER, located outside the cell nucleus, activates rapid responses molecules, including SRC and PI3K/AKT, which enhance the tumorigenic potential of prostate cancer cells, increasing cell proliferation, migration, invasion, and tumor formation.


Subject(s)
Androgens/metabolism , Prostatic Neoplasms/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Humans , Immunohistochemistry , Male , PC-3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , src-Family Kinases/metabolism
5.
Article in English | MEDLINE | ID: mdl-32328032

ABSTRACT

Prostate cancer is initially dependent on the androgen, gradually evolves into an androgen-independent form of the disease, also known as castration-resistant prostate cancer (CRPC). At this stage, current therapies scantily improve survival of the patient. Androgens and estrogens are involved in normal prostate and prostate cancer development. The mechanisms by which estrogens/estrogen receptors (ERs) induce prostate cancer and promote prostate cancer progression have not yet been fully identified. Our laboratory has shown that androgen-independent prostate cancer cells PC-3 express both ERα and ERß. The activation of ERß increases the expression of ß-catenin and proliferation of PC-3 cells. We now report that the activation of ERß promotes the increase of migration, invasion and anchorage-independent growth of PC-3 cells. Furthermore, the activation of ERα also plays a role in invasion and anchorage-independent growth of PC-3 cells. These effects are blocked by pretreatment with PKF 118-310, compound that disrupts the complex ß-catenin/TCF/LEF, suggesting that ERs/ß-catenin are involved in all cellular characteristics of tumor development in vitro. Furthermore, PKF 118-310 also inhibited the upregulation of vascular endothelial growth factor A (VEGFA) induced by activation of ERs. VEGF also is involved on invasion of PC-3 cells. In conclusion, this study provides novel insights into the signatures and molecular mechanisms of ERß in androgen-independent prostate cancer cells PC-3. ERα also plays a role on invasion and colony formation of PC-3 cells.


Subject(s)
Adenocarcinoma/pathology , Cell Movement , Cell Proliferation , Prostatic Neoplasms/pathology , Receptors, Estrogen/physiology , Androgens/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Estradiol/pharmacology , Humans , Male , Neoplasm Invasiveness , PC-3 Cells , Signal Transduction/drug effects , Tumor Stem Cell Assay , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
6.
Mol Cell Endocrinol ; 483: 11-23, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30660702

ABSTRACT

The aim of the present study was to investigate the subcellular localization of estrogen receptors ERα and ERß in androgen-independent prostate cancer cell line DU-145, and the possible role of exportin CRM1 on ERs distribution. In addition, we evaluated the ERs contribution to activation of ERK1/2 and AKT. Immunostaining of ERα and ERß was predominantly found in the extranuclear regions of DU-145 cells. CRM1 inhibitor Leptomycin B reduced drastically the presence of ERα and ERß in the extranuclear regions and increased in the nuclei, indicating the possible involvement of CRM1 on ERs nuclear-cytoplasmic shuttling. 17ß-estradiol (E2), ERα-selective agonist PPT and ERß-selective agonist DPN induced a rapid increase on ERK1/2 phosphorylation. E2-induced ERK1/2 activation was partially inhibited when cells were pretreated with ERα- or ERß-selective antagonists, and blocked by simultaneous pretreatment with both antagonists, suggesting ERα/ß heterodimers formation. Furthermore, E2 treatment did not activate AKT pathway. Therefore, we highlighted a possible crosstalk between extranuclear and nuclear ERs and their upstream and downstream signaling molecules as an important mechanism to control ER function as a potential therapeutic target in prostate cancer cells.


Subject(s)
Cytoplasm/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Karyopherins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Estradiol/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Male , Phosphorylation/drug effects , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Exportin 1 Protein
7.
Int J Biochem Cell Biol ; 96: 40-50, 2018 03.
Article in English | MEDLINE | ID: mdl-29341930

ABSTRACT

The aim of the present study was to investigate the impact of the activation of estrogen receptors on expression and localization of N-cadherin, E-cadherin and non-phosphorylated ß-catenin in androgen-independent prostate cancer cells (PC-3 and DU-145) and in human post pubertal prostate epithelial cells (PNT1A). Expression of N-cadherin was detected in PNT1A and PC-3 cells, but not in DU-145 cells. E-cadherin was detected only in DU-145 cells and ß-catenin was detected in all cells studied. N-cadherin and ß-catenin were located preferentially in the cellular membrane of PNT1A cells and in the cytoplasm of PC-3 cells. E-cadherin and ß-catenin were located preferentially in the cellular membrane of DU-145 cells. 17ß-estradiol (E2) or the ERα-selective agonist PPT did not affect the content and localization of N-cadherin in PC-3 and PNT1A cells or E-cadherin in DU-145 cells. In PC-3 cells, ERß-selective agonist DPN decreased the expression of N-cadherin. DPN-induced downregulation of N-cadherin was blocked by pretreatment with the ERß-selective antagonist (PHTPP), indicating that ERß1 is the upstream receptor regulating the expression of N-cadherin. In DU-145 cells, the activation of ERß1 by DPN increased the expression of E-cadherin. Taken together, these results suggest that activation of ERß1 is required to maintain an epithelial phenotype in PC-3 and DU-145 cells. The activation of ERß1 also increased the expression of ß-catenin in cytoplasm of PC-3 and in the cellular membrane of DU-145 cells. In conclusion, our results indicate differential expression and localization of N-cadherin, E-cadherin and ß-catenin in androgen-independent prostate cancer cells. The reduction of N-cadherin content by activation of ERß, exclusively observed in androgen-independent prostate cancer cells (PC-3), may be related to the activation of signaling pathways, such as the release of ß-catenin into the cytoplasm, translocation of ß-catenin to the nucleus and activation of gene transcription.


Subject(s)
Antigens, CD/biosynthesis , Cadherins/biosynthesis , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , beta Catenin/biosynthesis , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Estrogen Receptor beta/genetics , Humans , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction , beta Catenin/genetics
8.
Mol Cell Endocrinol ; 430: 12-24, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27107935

ABSTRACT

The aim of the present study was to characterize the mechanism underlying estrogen effects on the androgen-independent prostate cancer cell line PC-3. 17ß-estradiol and the ERß-selective agonist DPN, but not the ERα-selective agonist PPT, increased the incorporation of [methyl-(3)H]thymidine and the expression of Cyclin D2, suggesting that ERß mediates the proliferative effect of estrogen on PC-3 cells. In addition, upregulation of Cyclin D2 and incorporation of [methyl-(3)H]thymidine induced by 17ß-estradiol and DPN were blocked by the ERß-selective antagonist PHTPP in PC-3 cells. Upregulation of Cyclin D2 and incorporation of [methyl-(3)H]thymidine induced by DPN were also blocked by PKF118-310, a compound that disrupts ß-catenin-TCF (T-cell-specific transcription factor) complex, suggesting the involvement of ß-catenin in the estradiol effects in PC-3 cells. A diffuse immunostaining for non-phosphorylated ß-catenin was detected in the cytoplasm of PC-3 cells. Low levels of non-phosphorylated ß-catenin immunostaining were also detected near the plasma membrane and in nuclei. Treatment of PC-3 cells with 17ß-estradiol or DPN markedly increased non-phosphorylated ß-catenin expression. These effects were blocked by pretreatment with the ERß-selective antagonist PHTPP, PI3K inhibitor Wortmannin or AKT inhibitor MK-2206, indicating that ERß-PI3K/AKT mediates non-phosphorylated ß-catenin expression. Cycloheximide blocked the DPN-induced upregulation of non-phosphorylated ß-catenin, suggesting de novo synthesis of this protein. In conclusion, these results suggest that estrogen may play a role in androgen-independent prostate cancer cell proliferation through a novel pathway, involving ERß-mediated activation of ß-catenin.


Subject(s)
Estrogen Receptor beta/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin D2/metabolism , Cycloheximide/pharmacology , Estradiol/pharmacology , Estrogen Receptor beta/agonists , Humans , Male , Nitriles/pharmacology , Phenols/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Thymidine/metabolism
9.
Spermatogenesis ; 3(1): e23181, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23687614

ABSTRACT

The Wnt/ß-catenin signaling pathway controls several biological processes throughout development and adult life. Dysregulation of Wnt/ß-catenin signaling underlies a wide range of pathologies in animals and humans, including cancer in different tissues. In this review, we provide an update of the Wnt/ß-catenin signaling pathway and the possible roles of the Wnt/ß-catenin signaling in the biology of testis, epididymis and prostate. Data from our laboratory suggest the involvement of 17ß-estradiol and estrogen receptors (ERs) on the regulation of ß-catenin expression in rat Sertoli cells. We also provide emerging evidences of the involvement of Wnt/ß-catenin pathway in testis and prostate cancer. Our understanding of the role of Wnt/ß-Catenin signaling in male reproductive tissues is still evolving, and several questions are open to be addressed in the future.

SELECTION OF CITATIONS
SEARCH DETAIL
...