Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Histochem Cytochem ; 59(12): 1087-100, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22034518

ABSTRACT

Progress in our understanding of heterotypic cellular interaction in the tumor microenvironment, which is recognized to play major roles in cancer progression, has been hampered due to unavailability of an appropriate in vitro co-culture model. The aim of this study was to generate an in vitro 3-dimensional human breast cancer model, which consists of cancer cells and fibroblasts. Breast cancer cells (UACC-893) and fibroblasts at various densities were co-cultured in a rotating suspension culture system to establish co-culture parameters. Subsequently, UACC-893, BT.20, or MDA.MB.453 were co-cultured with fibroblasts for 9 days. Co-cultures resulted in the generation of breast cancer histoid (BCH) with cancer cells showing the invasion of fibroblast spheroids, which were visualized by immunohistochemical (IHC) staining of sections (4 µm thick) of BCH. A reproducible quantitative expression of C-erbB.2 was detected in UACC-893 cancer cells in BCH sections by IHC staining and the Automated Cellular Imaging System. BCH sections also consistently exhibited qualitative expression of pancytokeratins, p53, Ki-67, or E-cadherin in cancer cells and that of vimentin or GSTPi in fibroblasts, fibronectin in the basement membrane and collagen IV in the extracellular matrix. The expression of the protein analytes and cellular architecture of BCH were markedly similar to those of breast cancer tissue.


Subject(s)
Breast Neoplasms/pathology , Bioreactors , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , Fibroblasts/cytology , Humans , Immunohistochemistry , Neoplasm Invasiveness , Receptor, ErbB-2/metabolism , Tumor Microenvironment
2.
Mol Cell Biochem ; 336(1-2): 85-95, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19826768

ABSTRACT

Although safe in most cases, ancient treatments are ignored because neither their active components nor their molecular targets are well defined. This is not the case, however, with curcumin, a yellow-pigment substance and component of turmeric (Curcuma longa), which was identified more than a century ago. Recently, extensive research has addressed the chemotherapeutic potential of this relatively nontoxic-plant-derived polyphenol. Because most cancers are caused by deregulation of as many as 500 different genes, agents that target multiple gene products are needed for prevention and treatment of cancer. In this regard, curcumin has been reported to have immense potentiality for being used in cancer chemotherapy because of its control over the machineries of cell survival, proliferation, invasion, and angiogenesis. The mechanisms implicated are diverse and appear to involve a combination of cell signaling pathways at multiple levels. This review seeks to summarize the unique multifocal signal modulatory properties of the "ancient weapon," curcumin, which may be exploited for successful clinical cancer prevention.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/pharmacology , Neoplasms/drug therapy , Neoplasms/prevention & control , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Curcuma/chemistry , Curcumin/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medicine, Ayurvedic , Neoplasm Metastasis/prevention & control , Neoplasms/metabolism , Rhizome/chemistry , Signal Transduction/drug effects
3.
Carcinogenesis ; 31(2): 259-68, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19969555

ABSTRACT

The most common alterations found in breast cancer are inactivation or mutation of tumor suppressor gene p53. The present study revealed that theaflavins induced p53-mutated human breast cancer cell apoptosis. Pharmacological inhibition of caspase-8 or expression of dominant-negative (Dn)-caspase-8/Fas-associated death domain (FADD) partially inhibited apoptosis, whereas caspase-9 inhibitor completely blocked the killing indicating involvement of parallel pathways that converged to mitochondria. Further studies demonstrated theaflavin-induced Fas upregulation through the activation of c-jun N-terminal kinase, Fas-FADD interaction in a Fas ligand-independent manner, caspase-8 activation and t-Bid formation. A search for the parallel pathway revealed theaflavin-induced inhibition of survival pathway, mediated by Akt deactivation and Bcl-xL/Bcl-2-associated death promoter dephosphorylation. These well-defined routes of growth control converged to a common process of mitochondrial transmembrane potential loss, cytochrome c release and activation of the executioner caspase-9 and -3. Overexpression of either constitutively active myristylated-Akt (Myr-Akt) or Dn-caspase-8 partially blocked theaflavin-induced mitochondrial permeability transition and apoptosis of p53-mutated cells, whereas cotransfection of Myr-Akt and Dn-caspase-8 completely abolished theaflavin effect thereby negating the possibility of existence of third pathways. These results and other biochemical correlates established the concept that two distinct signaling pathways were regulated by theaflavins to induce mitochondrial death cascade, eventually culminating to apoptosis of p53-mutated human breast cancer cells that are strongly resistant to conventional therapies.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Biflavonoids/pharmacology , Breast Neoplasms/pathology , Catechin/pharmacology , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/metabolism , Caspase Inhibitors , Cytochromes c/metabolism , Fas-Associated Death Domain Protein/antagonists & inhibitors , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , bcl-Associated Death Protein/antagonists & inhibitors , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism , fas Receptor/antagonists & inhibitors , fas Receptor/genetics , fas Receptor/metabolism
4.
PLoS One ; 4(10): e7382, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19812686

ABSTRACT

BACKGROUND: Many tumors are associated with decreased cellular immunity and elevated levels of prostaglandin E2 (PGE2), a known inhibitor of CD4+ T cell activation and inducer of type-2 cytokine bias. However, the role of this immunomodulator in the survival of T helper cells remained unclear. Since CD4+ T cells play critical roles in cell-mediated immunity, detail knowledge of the effect tumor-derived PGE2 might have on CD4+ T cell survival and the underlying mechanism may, therefore, help to overcome the overall immune deviation in cancer. METHODOLOGY/PRINCIPAL FINDINGS: By culturing purified human peripheral CD4+ T cells or Jurkat cells with spent media of theaflavin- or celecoxib-pre-treated MCF-7 cells, we show that tumor-shed PGE2 severely impairs interleukin 2 receptor gammac (IL2Rgammac)-mediated survival signaling in CD4+ T cells. Indeed, tumor-shed PGE2 down-regulates IL2Rgammac expression, reduces phosphorylation as well as activation of Janus kinase 3 (Jak-3)/signal transducer and activator of transcription 5 (Stat-5) and decreases Bcl-2/Bax ratio thereby leading to activation of intrinsic apoptotic pathway. Constitutively active Stat-5A (Stat-5A1 6) over-expression efficiently elevates Bcl-2 levels in CD4+ T cells and protects them from tumor-induced death while dominant-negative Stat-5A over-expression fails to do so, indicating the importance of Stat-5A-signaling in CD4+ T cell survival. Further support towards the involvement of PGE2 comes from the results that (a) purified synthetic PGE2 induces CD4+ T cell apoptosis, and (b) when knocked out by small interfering RNA, cyclooxygenase-2 (Cox-2)-defective tumor cells fail to initiate death. Interestingly, the entire phenomena could be reverted back by theaflavins that restore cytokine-dependent IL2Rgammac/Jak-3/Stat-5A signaling in CD4+ T cells thereby protecting them from tumor-shed PGE2-induced apoptosis. CONCLUSIONS/SIGNIFICANCE: These data strongly suggest that tumor-shed PGE2 is an important factor leading to CD4+ T cell apoptosis during cancer and raise the possibility that theaflavins may have the potential as an effective immunorestorer in cancer-bearer.


Subject(s)
Biflavonoids/chemistry , CD4-Positive T-Lymphocytes/immunology , Catechin/chemistry , Dinoprostone/metabolism , Gene Expression Regulation , Interleukin Receptor Common gamma Subunit/metabolism , Neoplasms/metabolism , Adult , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , Cell Survival , Humans , Immune System , Jurkat Cells , Lymphocyte Activation , STAT5 Transcription Factor/metabolism , Transfection , Tumor Suppressor Proteins/metabolism
5.
Int J Cancer ; 125(1): 91-103, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19358280

ABSTRACT

Human papilloma virus is a causative factor in the etiology of cervical cancer with HPV16 being the most prevalent genotype associated with it. Intratype variations in oncogenic E6/E7 and capsid L1 proteins of HPV 16 besides being of phylogenetic importance, are associated with risk of viral persistence and progression. The objective of this multicentric study was to identify HPV-16 E6, E7 and L1 variants prevalent in India and their possible biological effects. Squamous cell cervical cancer biopsies were collected from 6 centres in India and examined for the presence of HPV 16. Variants of HPV-16 were characterized by full length sequence analysis of L1, E6 and E7 genes in 412 samples. Similar distribution of the variants was seen from the different centres/regions, with the European variant E350G being the most prevalent (58%), followed by American Asian variant (11.4%). Fifty six changes were seen in E6 region, 31 being nonsynonymous. The most frequent being L83V (72.3%), Q14H (13.1%) and H78Y (12.1%). Twenty-nine alterations were seen in E7 region, with 12 being nonsynonymous. The most frequent being F57V (9%). L1 region showed 204 changes, of which 67 were nonsynonymous. The most frequent being 448insS (100%), and 465delD (100%), H228D (94%), T292A (85%). The identified variants some new and some already reported can disrupt pentamer formation, transcriptional regulation of the virus, L1 protein interface interaction, B and T cell epitopes, p53 degradation, and thus their distribution is important for development of HPV diagnostics, vaccine, and for therapeutic purpose.


Subject(s)
Capsid Proteins/genetics , Genetic Variation , Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Repressor Proteins/genetics , Uterine Cervical Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Female , Human papillomavirus 16/classification , Humans , India , Middle Aged , Papillomavirus E7 Proteins , Papillomavirus Infections/epidemiology , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/virology
6.
Neuropathology ; 29(5): 521-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19019178

ABSTRACT

J1-31 is one of the astrocytic proteins, the expression of which has not been evaluated in astrocytomas. In the present study, we studied the expression of J1-31 protein in astrocytes and astrocytomas in comparison with GFAP, p53 and Ki-67. Materials consisted of formalin-fixed paraffin-embedded tissue specimens that included five cases of normal brain, 17 of gliosis, 15 of pilocytic astrocytoma (WHO grade I), 26 of low-grade diffuse astrocytoma (WHO grade II), four of anaplastic astrocytoma (WHO grade III), and eight of glioblastoma (WHO grade IV). GFAP was highly expressed in all specimens examined. The anti-J1-31 antibody exhibited strong cytoplasmic staining of reactive gliosis in 17/17 (100%) cases with a higher intensity of staining than that observed in the adjacent normal astrocytes. The antibody showed reactivity with tumor cells in 12/15 (80%) cases of pilocytic astrocytoma, although intensity of staining was generally weaker and more focal than observed in reactive gliosis. J1-31-positive tumor cells were detected in only 9/26 (35%) cases of the low-grade diffuse astrocytoma and none of the cases of anaplastic astrocytoma and glioblastoma. Increasing Ki-67 indices paralleled advancing tumor grades. p53 protein was expressed more commonly in infiltrating astrocytomas compared to pilocytic astrocytoma. In conclusion, down-regulation of J1-31 expression correlates with advancing grade of astrocytomas. The result suggests this protein plays some role in astrocytes that is progressively lost in malignant progression. The anti-J1-31 antibody may help further our understanding of astrocytes in disease and may be useful as an aid in the pathologic diagnosis of astrocytic lesions.


Subject(s)
Astrocytes/metabolism , Astrocytoma/metabolism , Nerve Tissue Proteins/metabolism , Cytoplasm/metabolism , Down-Regulation , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/metabolism , Gliosis/metabolism , Humans , Ki-67 Antigen/metabolism , Neoplasm Staging , Tumor Suppressor Protein p53/metabolism
7.
Prostate ; 68(15): 1681-8, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18712716

ABSTRACT

BACKGROUND: The concurrent determination of methylation status of E-cadherin gene and E-cadherin protein expression remains scant in metastatic prostate cancer cells in bone, the most prevalent site for metastatic growth. Therefore, the study was undertaken to ascertain the methylation status of E-cadherin gene, a most frequent and known epigenetic mechanism of its regulation, and the protein expression in prostate tissue biopsy specimen. METHODS: The methylation of E-cadherin gene was determined by methylation specific-PCR and the protein expression by immunohistochemical method in the consecutive sections of each prostate tissue biopsy specimen. RESULTS: The unmethylated E-cadherin gene and homogeneous E-cadherin protein expression was significantly associated with BPH as compared to the primary prostate carcinoma (Fisher's Exact P < 0.001). A significant association was observed between the concurrent methylated gene and markedly reduced expression of the protein in the primary prostate cancer cells as compared to the BPH cells, suggesting methylation-dependent regulation of the gene expression in these cases. In contrast to the primary cancer, a highly significant increase in the frequency of metastatic prostate cancer cells in bone exhibited the concurrent expression of unmethylated gene and homogeneous protein (Fisher's Exact P < 0.001). CONCLUSIONS: The study clearly demonstrated a significant association of the concurrent expression of unmethylated E-cadherin gene and E-cadherin protein with metastatic prostate cancer cells in bone, and that its expression may have a role in the intercellular adhesion in the formation of metastatic lesions in bone.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cadherins/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Biopsy , Bone Neoplasms/pathology , Cadherins/genetics , Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Methylation , Polymerase Chain Reaction , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/genetics , Tissue Distribution
8.
Apoptosis ; 13(6): 771-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18454316

ABSTRACT

Theaflavins, the bioactive flavonoids of black tea, have been demonstrated to inhibit proliferation and induce apoptosis in a variety of cancer cells. However, the contribution of p53 in mammary epithelial carcinoma cell apoptosis by theaflavins remains unclear. It has been reported that p53 triggers apoptosis by inducing mitochondrial outer membrane permeabilization through transcription-dependent and -independent mechanisms. Using wild-type and mutant p53-expressing as well as p53-null cells we found a strong correlation between p53 status and theaflavin-induced breast cancer cell apoptosis. Apoptogenic effect was more pronounced in functional p53-expressing cells in which theaflavins raised p53 protein levels that harmonized with Bax up-regulation and migration to mitochondria. However, in the same cells, when p53-mediated transactivation was inhibited by pifithrin-alpha, theaflavins not only failed to increase transcription but also to induce apoptosis although p53 up-regulation was not altered. In contrast, Bax over-expression restored back theaflavin-induced apoptosis in pifithrin-alpha-inhibited/dominant-negative p53-expressing cells. Inhibition of Bax by RNA-interference also reduced theaflavin-induced apoptosis. These results not only indicated the requirement of p53-mediated transcriptional activation of Bax but also its role as down-stream effecter in theaflavin-induced apoptosis. Bax up-regulation resulted in mitochondrial transmembrane potential loss and cytochrome c release followed by activation of caspase cascade. In contrast, mitochondrial translocation of p53 and its interaction with Bcl-2 family proteins or activation of caspase-8 could not be traced thereby excluding the involvement of p53-mediated transcription-independent pathways. Together these findings suggest that in breast cancer cells, p53 promotes theaflavin-induced apoptosis in a transcription-dependent manner through mitochondrial death cascade.


Subject(s)
Apoptosis/drug effects , Biflavonoids/pharmacology , Breast Neoplasms/pathology , Catechin/pharmacology , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/physiology , Apoptosis/physiology , Benzothiazoles/pharmacology , Caspases/metabolism , Cyclosporine/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Toluene/analogs & derivatives , Toluene/pharmacology , Transcriptional Activation/physiology , Up-Regulation
9.
Prostate ; 68(1): 78-84, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18008331

ABSTRACT

BACKGROUND: The expression of E-cadherin in the intercellular adhesion of metastatic prostate cancer cells in bone, which is the most prevalent site of metastatic growth, remains elusive. METHODS: The aim of the study was to compare the concurrent membranous expression of E-cadherin and beta-catenin proteins, the state which is known to be associated with the cellular adhesion function of E-cadherin, in prostate biopsy tissue specimens by immunohistochemical staining method. The expression patterns of E-cadherin or beta-catenin were classified as homogeneous (most cells exhibiting positively), heterogeneous (a few scattered patches of cells with positivity) or negative. RESULTS: Benign prostate hyperplasia cells exhibited homogeneous expression of both E-cadherin and beta-catenin in 9 of 11 (82%), whereas the primary prostate cancer cells were homogeneously positive for both proteins only in 4 of 22 (18%) of the cases. The results are similar to those reported in literature. However, in contrast to the primary cancer, a significantly increased frequency of the metastatic prostate cancer cells in bone exhibited homogeneous expression of E-cadherin and beta-catenin in 12 of 17 (71%) of the cases. A statistically significant association was observed between the overexpression of both proteins and the metastatic prostate cancer cells in bone (Fisher's exact P < 0.001). CONCLUSIONS: The result of the study demonstrated for the first time that the membranous overexpression of E-cadherin and beta-catenin are significantly associated with the metastatic prostate cancer cells in bone and that the high frequency of expression suggest their involvement in the intercellular adhesion of the metastatic cells in bone.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cadherins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , beta Catenin/metabolism , Biopsy , Humans , Immunohistochemistry , Male , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology
10.
Int J Gynecol Pathol ; 26(3): 214-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17581401

ABSTRACT

The expression of the catalytic subunit of telomerase protein (human telomerase reverse transcriptase [hTERT]), which is associated with telomerase activity, was evaluated as a potential marker of the high-grade premalignant cervical intraepithelial neoplasia (CIN 2/3) lesions. For comparison, cases of normal cervical squamous mucosa, low-grade CIN1 lesion, and cervical squamous cell carcinoma were included. The hTERT expression was also compared with Ki-67 and topoisomerase II-alpha (TPII-alpha) to determine the proliferative activity of the hTERT-positive dysplastic cells by a quantitative immunohistochemical staining method and was classified as follows: negative, 5% or less; moderate, 6% to 50%; or high, greater than 50% of the positive cells. The hTERT-positive cells were detected in a patchy pattern in the lower parabasal layers and in much of the basal layer in normal squamous mucosa. A similar frequency of Ki-67- or TPII-alpha-positive cells was observed, with the exception of the basal layer cells that were mostly negative. It is worthy to note that the recognizable intact basal layer cells in cases of CIN lesions were also consistently positive for the expression of hTERT, but rarely for Ki-67 or TPII-alpha. The expression of hTERT was detected in a less patchy pattern at a high or moderate percentage of the dysplastic epithelial cells each in 28.5% of cases of CIN1 lesions. A similar frequency, high and moderate percentage combined, of the TPII-alpha-positive dysplastic cell was also observed. In contrast, a high percentage of the hTERT-positive dysplastic cells were detected as diffuse basal or full-length thickness in 87.5% or 95% of cases of CIN2 or CIN3, respectively. A similar frequency of Ki-67 or TPII-alpha expression was observed in the dysplastic cells of CIN3 lesions. The pattern of hTERT-positive malignant cells in squamous cell carcinoma and dysplastic cells in the high-grade CIN lesions, to a greater extent, and dysplastic cells in the low-grade CIN lesion, to a lesser extent, was distinct from that of the normal cervical squamous mucosa. The results suggest that the progressive increase in the hTERT expression, together with the proliferative activity of the dysplastic epithelial cells of the high-grade CIN lesions, represents an early genetic abnormality in cervical pathogenesis.


Subject(s)
Biomarkers, Tumor/biosynthesis , Telomerase/biosynthesis , Uterine Cervical Dysplasia/enzymology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/pathology , Antigens, Neoplasm/biosynthesis , Cell Growth Processes/physiology , DNA Topoisomerases, Type II/biosynthesis , DNA-Binding Proteins/biosynthesis , Disease Progression , Female , Humans , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Retrospective Studies , Stromal Cells/enzymology , Stromal Cells/pathology , Telomerase/metabolism
11.
J Biol Chem ; 282(22): 15954-64, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17392282

ABSTRACT

Patients with advanced cancer exhibit multifaceted defects in their immune capacity, which are likely to contribute to an increased susceptibility to infections and disease progression. We demonstrated earlier that curcumin inhibits tumor growth and prevents immune cell death in tumor-bearing hosts. Here we report that tumor-induced immunodepletion involves apoptosis of thymic CD4+/CD8+ single/double positive cells as well as loss of circulating CD4+/CD8+ T cells. Administration of curcumin to tumor-bearing animals resulted in restoration of progenitor, effecter, and circulating T cells. In fact, tumor burden decreased the expression level of the pro-proliferative protein Bcl-2 while increasing the pro-apoptotic protein Bax in T cells. Curcumin down-regulated the Bax level while augmenting Bcl-2 expression in these cells, thereby protecting the immunocytes from tumor-induced apoptosis. A search for the upstream mechanism revealed down-regulation of the common cytokine receptor gamma chain (gammac) expression in T cells by tumor-secreted prostaglandin E2. As a result, Jak-3 and Stat-5a phosphorylation and to a lesser extent Stat-5b phosphorylation were also decreased in T cells. These entire phenomena could be reverted back by curcumin, indicating that this phytochemical restored the cytokine-dependent Jak-3/Stat-5a signaling pathway in T cells of tumor bearers. Overexpressed Stat-5a/constitutively active Stat-5a1*6 but not Stat-5b could efficiently elevate Bcl-2 levels and protect T cells from tumor-induced death, whereas C-terminal truncated Stat-5a713 overexpression failed to do so, indicating the importance of Stat-5a signaling in T cell survival. Thus, these results raise the possibility of inclusion of curcumin in successful therapeutic regimens against cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Curcumin/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT5 Transcription Factor/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Curcumin/therapeutic use , Dinoprostone/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Janus Kinase 3/metabolism , Lymphocyte Depletion , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Recovery of Function/drug effects , Recovery of Function/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/immunology , bcl-2-Associated X Protein/metabolism
12.
Anticancer Res ; 27(6B): 3903-8, 2007.
Article in English | MEDLINE | ID: mdl-18225549

ABSTRACT

AIM: The aim of the present study was to evaluate E-cadherin, whose expression remains poorly understood in the intercellular adhesion of metastatic breast cancer cells in bone, the most prevalent site for metastatic growth. MATERIALS AND METHODS: An immunohistochemical staining method was used for the localization of E-cadherin protein in tissue biopsy specimens of normal breast (n = 9) and well- (n = 8), moderately (n = 8) or poorly (n = 14) differentiated invasive primary breast cancer and metastatic breast cancer in bone (n = 17). The expression patterns of E-cadherin were classified as homogeneous (most cells exhibiting positivity), heterogeneous (a few scattered patches of cells with positivity) or negative (cells with undetectable positivity). RESULTS: Normal breast epithelial cells showed homogeneous overexpression of E-cadherin in all cases. A progressive and statistically significant reduction of E-cadherin expression was detected in the histologically well- to moderately to poorly differentiated breast cancer cells (p < 0.001). The clumps of invasive primary breast cancer cells in CD-31-positive blood vessels exhibited E-cadherin expression. Moreover, as compared to the poorly differentiated breast cancer cells, a significantly increased frequency of the metastatic breast cancer cells in bone exhibited homogeneous expression of E-cadherin in 15 out of 17 and heterogeneous expression in the remaining 2 cases (McNemar Exact p < 0.001). This is the first demonstration of membranous overexpression of E-cadherin on metastatic breast cancer cells in bone; the high frequency of its expression may have a role in the intercellular adhesion of metastatic cells in bone.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/biosynthesis , Female , Humans , Immunohistochemistry
13.
Int J Oncol ; 27(4): 949-56, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142310

ABSTRACT

This study examines the role of LEF1, a component of the Wnt signaling pathway, in human breast and murine mammary carcinoma and its relationship to ErbB2 (her-2/neu) expression. Mammary tissue and tumors from 5 different Wnt pathway-activated transgenic mouse strains and 5 different ErbB2 pathway-activated transgenic mouse strains were studied for the amount and distribution of expression of beta-catenin and LEF1. Fourteen samples of human infiltrating ductal breast cancer arising from a background of ductal carcinoma in situ (DCIS) were analyzed for LEF1, estrogen and progesterone receptor (ER and PR) and her-2/neu expression. in vitro, the effect of estradiol on LEF1 protein expression was examined in several breast cancer cell lines. The functional role of LEF1 was analyzed by a Matrigel invasion assay following transfection of breast cancer cell lines with either an LEF1 expression construct or a dominant-negative LEF1 construct. A significant (p=0.023) negative correlation between the expression of LEF1 and her-2/neu was observed in human breast cancer. LEF1 was strongly expressed, and beta-catenin had nuclear localization, in mammary tumors derived from Wnt pathway transgenic mice but not in ErbB2 pathway transgenic mice. In estrogen-receptor-positive breast cancer cell lines, LEF1 protein expression increased significantly following estradiol incubation (>200% of baseline). Following transient transfection, overexpression of LEF1 promoted and dominant-negative LEF1 inhibited tumor cell invasion. LEF1, a downstream component of the Wnt signaling pathway, defines a distinct, her-2/neu negative (non-overexpressing) subset of breast/mammary cancers in both humans and mice, mediates breast cancer cell invasion, and may be regulated in part by estradiol.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoid Enhancer-Binding Factor 1/physiology , Receptor, ErbB-2/biosynthesis , Wnt Proteins/metabolism , Animals , Blotting, Western , Breast/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Line, Tumor , Collagen/pharmacology , Down-Regulation , Drug Combinations , Estradiol/metabolism , Estrogens/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Jurkat Cells , Laminin/pharmacology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Neoplasm Invasiveness , Proteoglycans/pharmacology , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Time Factors , Transfection , Up-Regulation , beta Catenin/metabolism
14.
Anticancer Res ; 24(4): 2391-400, 2004.
Article in English | MEDLINE | ID: mdl-15330189

ABSTRACT

A retrospective study was undertaken to determine and compare the prognostic significance of LEA-135 protein expression by immunohistochemistry with other prognostic pathological parameters, with respect to recurrence and overall survival. This study was conducted in freshly-frozen tissue sections from a cohort of 367 patients having primary invasive breast cancer, with axillary lymph node metastasis. The association of LEA-135 expression was compared with estrogen and progesterone receptor status, segmentectomy or radical mastectomy and hormonal therapy or chemotherapy in terms of recurrence or disease-free survival. Pathologic parameters including tumor size, histological tumor type and histological grade, as well as age of patients at the time of initial diagnosis, and the treatments, together with a median follow-up of 8.8 years were contemplated for the study. Among these parameters, tumor size and histological grade were individually and significantly associated with an increased probability of recurrence (log rank p<0.001 in both cases) and short survival (log ranks p<0.001 and p=0.002, respectively), whereas age was only significantly associated with an increased probability of recurrence (log rank p=0.002) by univariate analysis. By multivariate analysis, both tumor size and histological grade remained statistically significant for recurrence (log rank p<0.001 and p=0.013, respectively) and overall survival (log ranks p<0.001 and p=0.016, respectively). Among the prognostic biomarkers, both ER and PR expression were associated with a decreased rate of recurrence (log ranks p<0.001 and p=0.008, respectively) and overall survival (log ranks p<0.001 and p=0.002, respectively) by univariate analysis. By multivariate analysis, only the ER expression remained significantly associated with a decreased recurrence and increased overall survival (log ranks p=0.023 and p=0.002, respectively). Patients with high (>50% positive cells) or moderate (5-50% positive cells) number of LEA-135-positive cells had a lower probability (46%) of recurrence at 10 years after surgery compared to 76% in LEA-135-negative patients (log rank p<0.001) by univariate analysis. Moreover, the probability of overall survival was higher in patients with high or moderate expression of LEA-135 (46% and 47%, respectively) compared to LEA-135-negative patients (24%) by univariate analysis (log rank p=0.009). By multivariate analysis, the association remained statistically significant for recurrence (log rank p<0.001) and survival (log rank p=0.002). However, there was no significant association between LEA-135 and any of the pathological parameters, age, hormone receptor status, the mode of surgery or the form of therapy (chemo- and/or hormonal) received by this cohort of patients. The results show that an improved prognosis was directly associated with the density of LEA-135-positive cancer cells, while loss of LEA-135 expression was associated with an aggressive phenotype of cancer cells during breast cancer progression. Thus, LEA-135 expression can be implicated as a significant and independent biomarker to identify and distinguish high- from low-risk patients with lymph node-positive invasive breast cancer for an aggressive treatment. Moreover, according to the present results, LEA-135 expression appears to be associated with the tumor cells that have retained certain normal biological characteristics, leading to their lack of aggressiveness and hence a better prognosis.


Subject(s)
Breast Neoplasms/metabolism , Membrane Glycoproteins/biosynthesis , Neoplasm Recurrence, Local/metabolism , Age Factors , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Humans , Immunohistochemistry , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Risk Factors , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...