Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncol Lett ; 18(6): 6545-6553, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31807173

ABSTRACT

Mammary neoplasms are a heterogeneous form of disease, and in order to determine its course and biological features with more accuracy, investigations based on tumor phenotypes are required. The aim of the present study was to propose and validate a phenotypic classification for canine mammary tumors and to assess any association between clinicopathological characteristics, survival and prognosis. For the immunohistochemistry analysis, the primary antibodies against estrogen receptor-α, progesterone receptor, human epidermal growth factor receptor 2 (HER-2)/neu and E-cadherin were used. A total of 110 canine mammary tumors were investigated; 42 tumors were classified as luminal A, 41 as luminal B, 17 as triple-negative and 10 as HER-2-positive. The luminal A and B phenotypes were associated with improved prognosis, whereas HER-2positive and triple-negative tumors were more aggressive, and exhibited a significant association with the occurrence of metastasis, a worse Tumor-Node-Metastasis classification and shorter survival time (P<0.05). In addition, there were different levels of E-cadherin expression intensity observed among the four tumor profiles investigated. Luminal A and B phenotypes presented an upregulation of E-cadherin compared with the HER-2 positive and triple-negative phenotypes (P<0.05). From the results of the present study, the proposed immunohistochemical panel and phenotypic classification techniques could be useful diagnostic tools with a good technical applicability in veterinary oncology. The analysis of E-cadherin expression in the panel of tumor markers allowed a more accurate classification for determining the biological features of the mammary tumor.

2.
Life Sci ; 183: 98-109, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28624391

ABSTRACT

Mammary tumorigenesis can be modulated by melatonin, which has oncostatic action mediated by multiple mechanisms, including the inhibition of the activity of transcription factors such as NF-κB and modulation of interleukins (ILs) expression. IL-25 is an active cytokine that induces apoptosis in tumor cells due to differential expression of its receptor (IL-17RB). IL-17B competes with IL-25 for binding to IL-17RB in tumor cells, promoting tumorigenesis. This study purpose is to address the possibility of engaging IL-25/IL-17RB signaling to enhance the effect of melatonin on breast cancer cells. Breast cancer cell lines were cultured monolayers and 3D structures and treated with melatonin, IL-25, siIL-17B, each alone or in combination. Cell viability, gene and protein expression of caspase-3, cleaved caspase-3 and VEGF-A were performed by qPCR and immunofluorescence. In addition, an apoptosis membrane array was performed in metastatic cells. Treatments with melatonin and IL-25 significantly reduced tumor cells viability at 1mM and 1ng/mL, respectively, but did not alter cell viability of a non-tumorigenic epithelial cell line (MCF-10A). All treatments, alone and combined, significantly increased cleaved caspase-3 in tumor cells grown as monolayers and 3D structures (p<0.05). Semi-quantitative analysis of apoptosis pathway proteins showed an increase of CYTO-C, DR6, IGFBP-3, IGFBP-5, IGFPB-6, IGF-1, IGF-1R, Livin, P21, P53, TNFRII, XIAP and hTRA proteins and reduction of caspase-3 (p<0.05) after melatonin treatment. All treatments reduced VEGF-A protein expression in tumor cells (p<0.05). Our results suggest therapeutic potential, with oncostatic effectiveness, pro-apoptotic and anti-angiogenic properties for melatonin and IL-25-driven signaling in breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Interleukin-17/metabolism , Melatonin/metabolism , Receptors, Interleukin/metabolism , Apoptosis/physiology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival/physiology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Interleukin-17/administration & dosage , Melatonin/administration & dosage , Melatonin/pharmacology , Neovascularization, Pathologic/metabolism , Polymerase Chain Reaction , Receptors, Interleukin-17 , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/genetics
3.
J Mammary Gland Biol Neoplasia ; 22(1): 27-41, 2017 03.
Article in English | MEDLINE | ID: mdl-28078601

ABSTRACT

Epithelial mesenchymal transition (EMT) is a process by which epithelial cells acquire mesenchymal properties, generating metastases. Transforming growth factor beta (TGF-ß) is associated with this malignancy by having the ability to induce EMT. Metformin, has been shown to inhibit EMT in breast cancer cells. Based on this evidence we hypothesize that treatment with metformin and the silencing of TGF-ß, inhibits the EMT in cancer cells. Canine metastatic mammary tumor cell line CF41 was stably transduced with a shRNA-lentivirus, reducing expression level of TGF-ß1. This was combined with metformin treatment, to look at effects on cell migration and the expression of EMT markers. For in vivo study, unmodified or TGF-ß1sh cells were injected in the inguinal region of nude athymic female mice followed by metformin treatment. The mice's lungs were collected and metastatic nodules were subsequently assessed for EMT markers expression. The migration rate was lower in TGF-ß1sh cells and when combined with metformin treatment. Metformin treatment reduced N-cadherin and increased E-cadherin expression in both CF41 and TGF-ß1sh cells. Was demonstrated that metformin treatment reduced the number of lung metastases in animals bearing TGF-ß1sh tumors. This paralleled a decreased N-cadherin and vimentin expression, and increased E-cadherin and claudin-7 expression in lung metastases. This study confirms the benefits of TGF-ß1 silencing in addition to metformin as potential therapeutic agents for breast cancer patients, by blocking EMT process. To the best of our knowledge, we are the first to report metformin treatment in cells with TGF-ß1 silencing and their effect on EMT.


Subject(s)
Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Mammary Neoplasms, Animal/drug therapy , Metformin/pharmacology , Transforming Growth Factor beta/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Dogs , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Neoplasm Invasiveness/pathology
4.
Anticancer Agents Med Chem ; 17(8): 1113-1125, 2017.
Article in English | MEDLINE | ID: mdl-28042775

ABSTRACT

BACKGROUND: ROCK-1 expression is associated with the malignant character of tumors, while inhibiting this molecule results in a significant suppression of tumor metastasis. Likewise, transforming growth factor beta (TGF-ß) is associated with this malignancy by having the ability to induce epithelial-mesenchymal transition (EMT). Metformin, a drug used in the treatment of diabetes, has previously been shown to inhibit EMT in breast cancer cells. OBJECTIVE: The aim of this study is to evaluate the TGF-ß1 action model for induction of EMT and the action of metformin and ROCK-1 inhibitor (Y27632) in EMT process in breast cancer cell lines. METHOD: MCF-7 and MDA-MB-231 cell lines were treated with metformin and Y27632, after induction of EMT by TGF-ß1, to examine the effects on cell migration as well as the protein expression of the ROCK-1 markers, vimentin, E-cadherin, CD44 and CD24 by immunocitochemistry. RESULTS: There was a lower protein expression of ROCK-1, vimentin, CD44 and CD24 in both cell lines after treatment with metformin and Y27632. In MDA-MB-231 cells, E-cadherin expression was increased in all treatment groups. Treatment of MDA-MB-231 cell line with metformin and Y27632 significantly reduced the invasion of these cells. CONCLUSION: This study confirms the benefits of metformin and Y27632 as potential therapeutic agents in mammary tumors, by blocking EMT process and metastatic potential.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Metformin/pharmacology , Pyridines/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
5.
PLoS One ; 11(3): e0150407, 2016.
Article in English | MEDLINE | ID: mdl-26934679

ABSTRACT

Cancer stem cells (CSCs) have been associated with metastasis and therapeutic resistance and can be generated via epithelial mesenchymal transition (EMT). Some studies suggest that the hormone melatonin acts in CSCs and may participate in the inhibition of the EMT. The objectives of this study were to evaluate the formation of mammospheres from the canine and human breast cancer cell lines, CMT-U229 and MCF-7, and the effects of melatonin treatment on the modulation of stem cell and EMT molecular markers: OCT4, E-cadherin, N-cadherin and vimentin, as well as on cell viability and invasiveness of the cells from mammospheres. The CMT-U229 and MCF-7 cell lines were subjected to three-dimensional culture in special medium for stem cells. The phenotype of mammospheres was first evaluated by flow cytometry (CD44(+)/CD24(low/-) marking). Cell viability was measured by MTT colorimetric assay and the expression of the proteins OCT4, E-cadherin, N-cadherin and vimentin was evaluated by immunofluorescence and quantified by optical densitometry. The analysis of cell migration and invasion was performed in Boyden Chamber. Flow cytometry proved the stem cell phenotype with CD44(+)/CD24(low/-) positive marking for both cell lines. Cell viability of CMT-U229 and MCF-7 cells was reduced after treatment with 1mM melatonin for 24 h (P<0.05). Immunofluorescence staining showed increased E-cadherin expression (P<0.05) and decreased expression of OCT4, N-cadherin and vimentin (P<0.05) in both cell lines after treatment with 1 mM melatonin for 24 hours. Moreover, treatment with melatonin was able to reduce cell migration and invasion in both cell lines when compared to control group (P<0.05). Our results demonstrate that melatonin shows an inhibitory role in the viability and invasiveness of breast cancer mammospheres as well as in modulating the expression of proteins related to EMT in breast CSCs, suggesting its potential anti-metastatic role in canine and human breast cancer cell lines.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Melatonin/pharmacology , Neoplasm Invasiveness/prevention & control , Neoplastic Stem Cells/drug effects , Animals , Breast/drug effects , Breast/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Dogs , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/pathology
6.
Mediators Inflamm ; 2016: 9512743, 2016.
Article in English | MEDLINE | ID: mdl-26989335

ABSTRACT

Inflammation results in the production of cytokines, such as interleukin- (IL-) 4 and IL-10 with immunosuppressive properties or IL-6 and TNF-α with procarcinogenic activity. Furthermore, NF-κB is the major link between inflammation and tumorigenesis. This study verified the interaction between active inflammatory cytokines in the tumor microenvironment and serum of female dogs with mammary tumors and their correlation with the clinicopathological characteristics and overall survival. Measurement of gene expression was performed by qPCR and protein levels by ELISA/Luminex. High gene and protein expression levels of NF-κB, IL-6, and TNF-α were found in association with characteristics that reflect worse prognosis and a negative correlation between TNF-α protein expression and survival time was observed (p < 0.05). In contrast, high gene and protein expression levels of IL-4 and IL-10 were associated with characteristics of better prognosis and an increased level of IL-4 and a longer survival time of animals were obtained (p < 0.05). In addition, there was a positive correlation between TNF-α and IL-6 expression in association with NF-κB. The results show a significant correlation of these cytokines with tumor development, associated with NF-κB expression and cytokines promodulation, showing that these biological factors could be used as predictive and prognostic markers in breast cancer.


Subject(s)
Cytokines/metabolism , NF-kappa B/metabolism , Animals , Dogs , Female , Interleukin-10/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
J Pineal Res ; 60(1): 3-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26292662

ABSTRACT

The occurrence of metastasis, an important breast cancer prognostic factor, depends on cell migration/invasion mechanisms, which can be controlled by regulatory and effector molecules such as Rho-associated kinase protein (ROCK-1). Increased expression of this protein promotes tumor growth and metastasis, which can be restricted by ROCK-1 inhibitors. Melatonin has shown oncostatic, antimetastatic, and anti-angiogenic effects and can modulate ROCK-1 expression. Metastatic and nonmetastatic breast cancer cell lines were treated with melatonin as well as with specific ROCK-1 inhibitor (Y27632). Cell viability, cell migration/invasion, and ROCK-1 gene expression and protein expression were determined in vitro. In vivo lung metastasis study was performed using female athymic nude mice treated with either melatonin or Y27832 for 2 and 5 wk. The metastases were evaluated by X-ray computed tomography and single photon emission computed tomography (SPECT) and by immunohistochemistry for ROCK-1 and cytokeratin proteins. Melatonin and Y27632 treatments reduced cell viability and invasion/migration of both cell lines and decreased ROCK-1 gene expression in metastatic cells and protein expression in nonmetastatic cell line. The numbers of 'hot' spots (lung metastasis) identified by SPECT images were significantly lower in treated groups. ROCK-1 protein expression also was decreased in metastatic foci of treated groups. Melatonin has shown to be effective in controlling metastatic breast cancer in vitro and in vivo, not only via inhibition of the proliferation of tumor cells but also through direct antagonism of metastatic mechanism of cells rendered by ROCK-1 inhibition. When Y27632 was used, the effects were similar to those found with melatonin treatment.


Subject(s)
Amides/pharmacology , Breast Neoplasms/drug therapy , Melatonin/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Single Photon Emission Computed Tomography Computed Tomography , Xenograft Model Antitumor Assays , rho-Associated Kinases/metabolism
8.
Anticancer Agents Med Chem ; 16(3): 347-58, 2016.
Article in English | MEDLINE | ID: mdl-25963143

ABSTRACT

Angiogenesis is the process of new blood vessel formation, regulated by a number of pro- and antiangiogenic factors and usually begins in response to hypoxia. Exogenous administration of melatonin has shown numerous anti-tumor effects and appears to inhibit tumor angiogenesis. However, many factors involved in the anti-angiogenic effect of melatonin are still under investigation. Here, we evaluate the effects of melatonin on cell viability and expression of angiogenic factors in MCF-7 and MDA-MB-231 breast cancer cells under hypoxic conditions. Cell viability was investigated by MTT and gene and protein expression of the hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF-A) were verified by qPCR and immunocytochemistry after melatonin treatment (1 mM) under hypoxic conditions. Additionally, a protein array with 20 different cytokines/factors was performed on tumor cell lysates. The results showed that 1 mM of melatonin reduced the viability of MCF-7 and MDA-MB-231 cells (p < .05). This treatment also decreased both gene and protein expression of HIF-1α and VEGF-A under hypoxic conditions (p < .05). Among the proteins evaluated by protein array, melatonin treatment during hypoxia reduced VEGF-C, VEGFR receptors (VEGFR2 and VEGFR3), matrix metalloproteinase 9 (MMP9) and Angiogenin in MCF-7 cells. In MDA-MB-231 cells, a significant decrease was observed in VEGFR2, epidermal growth factor receptor (EGFR) and Angiogenin (p < .05). Taken together, these results showed that melatonin acts in the regulation of angiogenic factors in breast tumor cells and suggests an anti-angiogenic activity, particularly under hypoxic conditions.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antioxidants/pharmacology , Breast Neoplasms/blood supply , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melatonin/pharmacology , Neovascularization, Pathologic/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cytokines/metabolism , ErbB Receptors/metabolism , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Ribonuclease, Pancreatic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3
9.
Oncol Rep ; 33(5): 2345-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25779537

ABSTRACT

Mammary tumors are the most common type of tumor in dogs, with approximately half of these tumors being malignant. Hypoxia, characterized by oxygen levels below normal, is a known adverse factor to cancer treatment. The hypoxia-inducible factor 1α (HIF-1α) is a central regulator of the pathophysiological response of mammalian cells to low oxygen levels. HIF-1α activates the transcription of vascular endothelial growth factor (VEGF), which in turn promotes angiogenesis through its ability to stimulate the growth, migration and invasion of endothelial cells to form new blood vessels, contributing to tumor progression. In this study, we evaluated the serum concentration and gene expression of VEGF and HIF-1α linking them with clinicopathological parameters and survival of dogs with mammary tumors in order to infer the possible prognostic value of these factors. We collected blood and tumor fragments of 24 female dogs with malignant mammary tumors (study group) and 26 non-affected female dogs (control group) to verify the gene expression of VEGF and HIF-1α by quantitative real-time PCR (qPCR) and the serum levels by ELISA (enzyme-linked immunosorbent). The results showed high serum levels of VEGF in the study group and its correlation between abundant vascularization, lymph node involvement, metastasis, death rate and low survival (p<0.05). The serum percentage of HIF-1α in female dogs with mammary neoplasia was lower than that in the control group and higher in female dogs with tumor metastasis and history of tumor recurrence (p<0.05). Regarding gene expression, there was a gene overexpression of VEGFA in female dogs with poor outcome, in contrast to the gene underexpression of HIF-1A. Taken together, these results suggested that VEGF is important in tumor progression and can be used as a potential prognostic marker in the clinic and may be useful in predicting tumor progression in dogs with mammary neoplasia.


Subject(s)
Carcinoma/genetics , Dog Diseases/genetics , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mammary Neoplasms, Animal/genetics , Neovascularization, Pathologic/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Carcinoma/mortality , Carcinoma/pathology , Dog Diseases/mortality , Dog Diseases/pathology , Dogs , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymph Nodes/pathology , Mammary Neoplasms, Animal/mortality , Mammary Neoplasms, Animal/pathology , Neoplasm Staging , Neovascularization, Pathologic/pathology , Prognosis , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism
10.
Oncol Rep ; 33(1): 311-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25384569

ABSTRACT

Mammary neoplasias are the most common tumors observed in female dogs. Identification of these tumors is valuable in order to identify beneficial therapeutic agents as alternative treatments for this tumor type. Oral administration of melatonin appears to exert an oncostatic effect on mammary neoplasia and may have a possible mechanism of action through its interaction with estrogen receptors on epithelial cells. Hence, we analyzed the potential therapeutic value of melatonin in tumors that are estrogen-dependent or -independent, and established a relationship of its action with the expression of the melatonin receptors MT1 and MT2. Furthermore, we analyzed the rate of cell proliferation and apoptosis after treatment with melatonin. Cell cultures were performed using 10 canine mammary tumor fragments and were divided into estrogen receptor (ER)-positive and ER-negative tumors. The results showed that both ER-positive and ER-negative tumors had decreased cell viability and proliferation after treatment with melatonin (p<0.05), although treatment was more effective in the ER-positive tumors. Analysis of the relative expression of the MT1 and MT2 genes by quantitative PCR was performed and the data were compared with the expression of ER in 24 canine mammary tumors and the cellular response to melatonin in 10 samples. MT1 was overexpressed in ER-positive tumors (p<0.05), whereas MT2 was not expressed. Furthermore, melatonin treatment in ER-positive tumors showed an efficient oncostatic effect by inhibiting cell viability and proliferation and inducing apoptosis. These results suggest that melatonin decreased neoplastic mammary cell proliferation and viability and induced apoptosis, with greater efficacy in ER-positive tumors that have a high expression of melatonin receptor MT1. This is a strong evidence for the use of melatonin as a therapeutic agent for estrogen-dependent canine mammary tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Dog Diseases/drug therapy , Mammary Neoplasms, Animal/drug therapy , Melatonin/pharmacology , Animals , Apoptosis , Cell Proliferation/drug effects , Cell Survival/drug effects , Dog Diseases/metabolism , Dogs , Drug Screening Assays, Antitumor , Female , Gene Expression , Mammary Neoplasms, Animal/metabolism , Primary Cell Culture , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Tumor Cells, Cultured
11.
Anticancer Agents Med Chem ; 14(9): 1302-11, 2014.
Article in English | MEDLINE | ID: mdl-25323035

ABSTRACT

BACKGROUND: Oral cancer is the most common type of head and neck cancer and its high rate of mortality and morbidity is closely related to the processes of angiogenesis and tumor metastasis. The overexpression of the pro-angiogenic genes, HIF-1α and VEGF, and pro-metastatic gene, ROCK-1, are associated with unfavorable prognosis in oral carcinoma. Melatonin has oncostatic, antiangiogenic and antimetastatic properties in several types of neoplasms, although its relationship with oral cancer has been little explored. This study aims to analyze the expression of the genes HIF-1α, VEGF and ROCK-1 in cell lines of squamous cell carcinoma of the tongue, after treatment with melatonin. METHODS: SCC9 and SCC25 cells were cultured and cell viability was assessed by MTT assay, after treatment with 100 µM of CoCl2 to induce hypoxia and with melatonin at different concentrations. The analysis of quantitative RT-PCR and the immunocytochemical analysis were performed to verify the action of melatonin under conditions of normoxia and hypoxia, on gene and protein expression of HIF-1α, VEGF and ROCK-1. RESULTS: The MTT assay showed a decrease in cell viability in both cell lines, after the treatment with melatonin. The analysis of quantitative RT-PCR indicated an inhibition of the expression of the pro-angiogenic genes HIF-1α (P < 0.001) and VEGF (P < 0.001) under hypoxic conditions, and of the pro-metastatic gene ROCK-1 (P < 0.0001) in the cell line SCC9, after treatment with 1 mM of melatonin. In the immunocytochemical analysis, there was a positive correlation with gene expression data, validating the quantitative RT-PCR results for cell line SCC9. Treatment with melatonin did not demonstrate inhibition of the expression of genes HIF-1α, VEGF and ROCK-1 in line SCC25, which has different molecular characteristics and greater degree of malignancy when compared to the line SCC9. CONCLUSION: Melatonin affects cell viability in the SCC9 and SCC25 lines and inhibits the expression of the genes HIF-1α, VEGF and ROCK-1 in SCC9 line. Additional studies may confirm the potential therapeutic effect of melatonin in some subtypes of oral carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Melatonin/pharmacology , Mouth Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasm Metastasis , Vascular Endothelial Growth Factor A/metabolism , rho-Associated Kinases/metabolism
12.
Acta Histochem ; 116(1): 148-57, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23899963

ABSTRACT

The expression of prognostic markers in cancer has become important in diagnostic routine and research. A high mitotic rate compromises the individual cell access to oxygen and nutrients, due to reduced blood supply. Cells undertake adaptive measures such as vascular endothelial growth factor (VEGF), expressed under the control of hypoxia-inducible factor-1α (HIF-1α). CD34 is an endothelial marker which can show the presence and distribution of blood vessels. This study evaluated the presence and relative expression of VEGF, HIF-1α and CD34 using immunohistochemistry of 60 breast tumors and double staining, correlating the findings with clinical and pathological variables. High VEGF expression was correlated with low cell proliferation, lymph node-negative, smaller tumor size and patients not receiving hormone therapy. High HIF-1α expression predominated in younger (<50-year) patients, subjected to neo-adjuvant therapy and in p53-negative tumors. Absence of metastasis, radiotherapy or hormone treatment, and estrogen receptor (ER)-positive tumors showed high CD34 immunoreactivity. We suggest that the angiogenic factors VEGF, HIF-1α and CD34 are important in breast cancer progression and their abundance in breast tumors has prognostic and predictive value.


Subject(s)
Antigens, CD34/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/secondary , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , ROC Curve , Tissue Array Analysis
13.
Oncol Rep ; 30(3): 1119-28, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23765060

ABSTRACT

The use of prognostic markers for breast cancer allows therapeutic strategies to be defined more efficiently. The expression of glutathione (GSH) and glutathione peroxidase (GPX) in tumor cells has been evaluated as a predictor of prognosis and response to cytotoxic treatments. Its immunoexpression was assessed in 63 women diagnosed with invasive ductal carcinoma in a retrospective study. The results showed that high GSH expression was associated with tumors negative for the estrogen receptor (ER) (P<0.05), and GPX expression was associated with tumors negative for the progesterone receptor (PR) and patient mortality. Focusing on the 37 patients who received adjuvant chemotherapy/radiotherapy (Group I), high expression of GPX was associated with a high rate of patient mortality (P<0.05). The 19 patients who received only adjuvant chemotherapy (Group II) showed high expression of GSH in relation to metastasis (P<0.05). In addition, high levels of GPX expression were significantly associated with a shorter overall survival (P<0.05). To confirm this, the expression of precursor genes of GSH [glutamate cysteine ligase (GCLC) and glutathione synthetase (GSS)] and the GPX gene was analyzed using quantitative PCR in cultured neoplastic mammary cells treated with doxorubicin. Doxorubicin treatment was able to eliminate tumor cells without alterations in the gene expression of GSS, but led to underexpression of the GCLC and GPX genes. Our results suggest that high levels of GPX may be related to the development of resistance to chemotherapy in these tumors, response to treatment and the clinical course of the breast cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Glutathione Peroxidase/metabolism , Glutathione/metabolism , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/pathology , Cell Proliferation/drug effects , Chemotherapy, Adjuvant , Doxorubicin/pharmacology , Female , Follow-Up Studies , Glutathione Peroxidase/genetics , Humans , Immunoenzyme Techniques , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Glutathione Peroxidase GPX1
14.
Vet Immunol Immunopathol ; 146(2): 106-12, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22405680

ABSTRACT

Mammary gland tumors in female dogs are an excellent model for the clinic-pathological, diagnostic and prognostic investigation of mammary neoplasias. Prognostic and predictive markers are effective in research and routine diagnosis. Interleukins play a fundamental role in cancer, with a particular function in tumor growth, invasion and metastatic potential. Interleukin-8 (IL-8) is known to possess tumorigenic and pro-angiogenic properties, and its overexpression is seen in a number of human tumors. IL-8 serum levels were determined and correlated with the clinic-pathological features and clinical evolution of mammary gland neoplasias in female dogs. IL-8 was measured by an immunoenzymatic assay in 30 female dogs with mammary neoplasias within a 12 month follow-up and in 50 control animals. The correlation between IL-8 concentration and clinical parameters was investigated. A statistically significant difference in the IL-8 serum levels was found in tumor-bearing dogs compared to the controls. In addition, when the individual parameters were evaluated, IL-8 content showed a positive correlation with the tumor progression, lymph node involvement, recurrence and death. Single and multivariate analyses showed associations between tumor recurrence, metastasis, high clinical staging and high IL-8, and also with the death risk. This was also consistent with the high IL-8 content in dogs showing tumor recurrence and metastasis. IL-8 superexpression has been detected in a number of human tumors, usually associated with a poor prognostic. Besides promoting angiogenesis, IL-8 is strongly related with the metastatic phenotype of mammary tumor cells. High IL-8 concentration was found in mammary gland cancer patients with advanced disease stages. Our results show that IL-8 can be used as a non-invasive prognostic marker for mammary gland cancer, and can be useful for the prediction of disease progression and recurrence in dogs with mammary neoplasias. The increased level of this cytokine acts as an independent prognostic marker of survival and the identification of animals with the poor prognostic.


Subject(s)
Biomarkers, Tumor/immunology , Dog Diseases/immunology , Interleukin-8/immunology , Mammary Neoplasms, Animal/immunology , Animals , Biomarkers, Tumor/blood , Disease Progression , Dog Diseases/blood , Dogs , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Interleukin-8/blood , Kaplan-Meier Estimate , Mammary Neoplasms, Animal/blood , Predictive Value of Tests , ROC Curve
15.
Acta Histochem ; 114(5): 510-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22000861

ABSTRACT

Breast cancer is the most frequent cancer in women worldwide. Prognostic markers are important for diagnosis, allowing therapeutic strategies to be defined more efficiently. The expression of the glutathione S-transferase pi isoenzyme (GSTpi) in tumor cells has been evaluated as a predictor of prognosis and in response to cytotoxic treatments. Its immunoexpression was assessed in 63 women diagnosed with invasive ductal carcinoma in a retrospective study. The results were statistically correlated with clinicopathological parameters of patients. The results showed that high GSTpi expression was related to p53-positive tumors, grade III histology, large tumor size and death (p<0.05). The 37 patients who received adjuvant treatment, checked separately, showed high expression of GSTpi in relation to local recurrence, metastasis and death (p<0.05). In addition, high levels of GSTpi expression were significantly associated with a shorter overall survival (p<0.05). To confirm this suspicion, GSTpi gene expression was checked by Real-time PCR in neoplastic mammary cells cultured and subjected to treatment with doxorubicin. Our results suggest that high levels of GSTpi may be related to the development of resistance to chemotherapy in these tumors, the response of these tumors to treatment and the clinical course of the patients involved.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glutathione S-Transferase pi/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , Female , Humans , Immunohistochemistry , Middle Aged , Predictive Value of Tests , Prognosis , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...