Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pathol ; 235(4): 559-70, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25404354

ABSTRACT

We investigated the oncogenic role of SETDB1, focusing on non-small cell lung cancer (NSCLC), which has high expression of this protein. A total of 387 lung cancer cases were examined by immunohistochemistry; 72% of NSCLC samples were positive for SETDB1 staining, compared to 46% samples of normal bronchial epithelium (106 cases) (p <0.0001). The percentage of positive cells and the intensity of staining increased significantly with increased grade of disease. Forced expression of SETDB1 in NSCLC cell lines enhanced their clonogenic growth in vitro and markedly increased tumour size in a murine xenograft model, while silencing (shRNA) SETDB1 in NSCLC cells slowed their proliferation. SETDB1 positively stimulated activity of the WNT-ß-catenin pathway and diminished P53 expression, resulting in enhanced NSCLC growth in vitro and in vivo. Our finding suggests that therapeutic targeting of SETDB1 may benefit patients whose tumours express high levels of SETDB1.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Protein Methyltransferases/metabolism , Wnt Signaling Pathway , Animals , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , HCT116 Cells , Histone-Lysine N-Methyltransferase , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Neoplasm Grading , Neoplasm Transplantation , Protein Methyltransferases/genetics , RNA Interference , Time Factors , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway/genetics
2.
BMC Cancer ; 15: 655, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26445321

ABSTRACT

BACKGROUND: Anterior gradient 2 (AGR2) is a protein disulfide isomerase-like protein widely expressed in many normal tissues as well as cancers. In our study, non-neoplastic bronchial epithelial cells as well as non-small cell lung cancer (NSCLC) cells express AGR2 protein. METHODS: AGR2 expression was analyzed on lung tissue microarrays. Tumor staining was correlated with clinical outcomes. RESULTS: On a lung cancer tissue microarray using immunohistochemistry, expression levels in cancer showed generally decreasing intensities in order from adenocarcinomas with mucinous components, other adenocarcinomas, squamous carcinomas, to large cell carcinomas. The study cohort was comprised of 400 cases. As a group, there was a slight trend of lower expression with increasing tumor grade. AGR2 expression level was a significant predictor of overall survival in younger patients only. Patients under 65 with lower levels showed a significantly better survival for both men and women. Patients over 65, in contrast, showed no such trend. CONCLUSIONS: Nearly all NSCLC tumors show AGR2 expression. Lung cancer expression of AGR2 has prognostic value for younger patients.


Subject(s)
Biomarkers, Tumor , Gene Expression , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Proteins/genetics , Age Factors , Aged , Aged, 80 and over , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/diagnosis , Male , Middle Aged , Mucoproteins , Neoplasm Grading , Neoplasm Staging , Oncogene Proteins , Prognosis , Proportional Hazards Models , Proteins/metabolism , Risk Factors
3.
Cancers (Basel) ; 16(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38672563

ABSTRACT

Breast cancer (BC) remains among the most commonly diagnosed cancers in women worldwide. Triple-negative BC (TNBC) is a subset of BC characterized by aggressive behavior, a high risk of distant recurrence, and poor overall survival rates. Chemotherapy is the backbone for treatment in patients with TNBC, but outcomes remain poor compared to other BC subtypes, in part due to the lack of recognized functional targets. In this study, the expression of the tetraspan protein epithelial membrane protein 2 (EMP2) was explored as a predictor of TNBC response to standard chemotherapy. We demonstrate that EMP2 functions as a prognostic biomarker for patients treated with taxane-based chemotherapy, with high expression at both transcriptomic and protein levels following treatment correlating with poor overall survival. Moreover, we show that targeting EMP2 in combination with docetaxel reduces tumor load in syngeneic and xenograft models of TNBC. These results provide support for the prognostic and therapeutic potential of this tetraspan protein, suggesting that anti-EMP2 therapy may be beneficial for the treatment of select chemotherapy-resistant TNBC tumors.

4.
Mod Pathol ; 26(6): 849-59, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23348903

ABSTRACT

The combination of expression patterns of AGR2 (anterior gradient 2) and CD10 by prostate cancer provided four phenotypes that correlated with clinical outcome. Based on immunophenotyping, CD10(low)AGR2(high), CD10(high)AGR2(high), CD10(low)AGR2(low), and CD10(high)AGR2(low) were distinguished. AGR2(+) tumors were associated with longer recurrence-free survival and CD10(+) tumors with shorter recurrence-free survival. In high-stage cases, the CD10(low)AGR2(high) phenotype was associated with a ninefold higher recurrence-free survival than the CD10(high)AGR2(low) phenotype. The CD10(high)AGR2(high) and CD10(low)AGR2(low) phenotypes were intermediate. The CD10(high)AGR2(low) phenotype was most frequent in high-grade primary tumors. Conversely, bone and other soft tissue metastases, and derivative xenografts, expressed more AGR2 and less CD10. AGR2 protein was readily detected in tumor metastases. The CD10(high)AGR2(low) phenotype in primary tumors is predictive of poor outcome; however, the CD10(low)AGR2(high) phenotype is more common in metastases. It appears that AGR2 has a protective function in primary tumors but may have a role in the distal spread of tumor cells.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Carcinoma/metabolism , Neprilysin/metabolism , Prostatic Neoplasms/metabolism , Proteins/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Carcinoma/genetics , Carcinoma/mortality , Carcinoma/secondary , Disease-Free Survival , Heterografts , Humans , Immunohistochemistry , Immunophenotyping , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mucoproteins , Multivariate Analysis , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Transplantation , Oncogene Proteins , Phenotype , Proportional Hazards Models , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Risk Factors , Time Factors , Tissue Array Analysis
5.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1150-8, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22345575

ABSTRACT

Diminished Na,K-ATPase expression has been reported in several carcinomas and has been linked to tumor progression. However, few studies have determined whether Na,K-ATPase function and expression are altered in lung malignancies. Because cigarette smoke (CS) is a major factor underlying lung carcinogenesis and progression, we investigated whether CS affects Na,K-ATPase activity and expression in lung cell lines. Cells exposed to CS in vitro showed a reduction of Na,K-ATPase activity. We detected the presence of reactive oxygen species (ROS) in cells exposed to CS before Na,K-ATPase inhibition, and neutralization of ROS restored Na,K-ATPase activity. We further determined whether Na,K-ATPase expression correlated with increasing grades of lung adenocarcinoma and survival of patients with smoking history. Immunohistochemical analysis of lung adenocarcinoma tissues revealed reduced Na,K-ATPase expression with increasing tumor grade. Using tissue microarray containing lung adenocarcinomas of patients with known smoking status, we found that high expression of Na,K-ATPase correlated with better survival. For the first time, these data demonstrate that CS is associated with loss of Na,K-ATPase function and expression in lung carcinogenesis, which might contribute to disease progression.


Subject(s)
Adenocarcinoma/enzymology , Lung Neoplasms/enzymology , Nicotiana , Smoke/adverse effects , Sodium-Potassium-Exchanging ATPase/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Cell Line, Tumor , Disease Progression , Disease-Free Survival , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Neoplasm Grading , Reactive Oxygen Species/metabolism , Smoking/adverse effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/biosynthesis
6.
BMC Cancer ; 11: 230, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21651811

ABSTRACT

BACKGROUND: Tissue microarray (TMA) data are commonly used to validate the prognostic accuracy of tumor markers. For example, breast cancer TMA data have led to the identification of several promising prognostic markers of survival time. Several studies have shown that TMA data can also be used to cluster patients into clinically distinct groups. Here we use breast cancer TMA data to cluster patients into distinct prognostic groups. METHODS: We apply weighted correlation network analysis (WGCNA) to TMA data consisting of 26 putative tumor biomarkers measured on 82 breast cancer patients. Based on this analysis we identify three groups of patients with low (5.4%), moderate (22%) and high (50%) mortality rates, respectively. We then develop a simple threshold rule using a subset of three markers (p53, Na-KATPase-ß1, and TGF ß receptor II) that can approximately define these mortality groups. We compare the results of this correlation network analysis with results from a standard Cox regression analysis. RESULTS: We find that the rule-based grouping variable (referred to as WGCNA*) is an independent predictor of survival time. While WGCNA* is based on protein measurements (TMA data), it validated in two independent Affymetrix microarray gene expression data (which measure mRNA abundance). We find that the WGCNA patient groups differed by 35% from mortality groups defined by a more conventional stepwise Cox regression analysis approach. CONCLUSIONS: We show that correlation network methods, which are primarily used to analyze the relationships between gene products, are also useful for analyzing the relationships between patients and for defining distinct patient groups based on TMA data. We identify a rule based on three tumor markers for predicting breast cancer survival outcomes.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Sodium-Potassium-Exchanging ATPase/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , Neoplasm Proteins/genetics , Prognosis , Proportional Hazards Models , Protein Array Analysis , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Sodium-Potassium-Exchanging ATPase/genetics
7.
BMC Cancer ; 11: 259, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21689459

ABSTRACT

BACKGROUND: Raf-1 kinase inhibitor protein (RKIP) has been reported to negatively regulate signal kinases of major survival pathways. RKIP activity is modulated in part by phosphorylation on Serine 153 by protein kinase C, which leads to dissociation of RKIP from Raf-1. RKIP expression is low in many human cancers and represents an indicator of poor prognosis and/or induction of metastasis. The prognostic power has typically been based on total RKIP expression and has not considered the significance of phospho-RKIP. METHODS: The present study examined the expression levels of both RKIP and phospho-RKIP in human lung cancer tissue microarray proteomics technology. RESULTS: Total RKIP and phospho-RKIP expression levels were similar in normal and cancerous tissues. phospho-RKIP levels slightly decreased in metastatic lesions. However, the expression levels of phospho-RKIP, in contrast to total RKIP, displayed significant predictive power for outcome with normal expression of phospho-RKIP predicting a more favorable survival compared to lower levels (P = 0.0118); this was even more pronounced in more senior individuals and in those with early stage lung cancer. CONCLUSIONS: This study examines for the first time, the expression profile of RKIP and phospho-RKIP in lung cancer. Significantly, we found that phospho-RKIP was a predictive indicator of survival.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Phosphatidylethanolamine Binding Protein/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Disease Progression , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Phosphorylation , Prognosis , Survival Analysis
8.
Sci Rep ; 11(1): 11949, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099751

ABSTRACT

In type 1 endometrial cancer, unopposed estrogen stimulation is thought to lead to endometrial hyperplasia which precedes malignant progression. Recent data from our group and others suggest that ALDH activity mediates stemness in endometrial cancer, but while aldehyde dehydrogenase 1 (ALDH1) has been suggested as a putative cancer stem cell marker in several cancer types, its clinical and prognostic value in endometrial cancer remains debated. The aim of this study was to investigate the clinical value of ALDH1 expression in endometrial hyperplasia and to determine its ability to predict progression to endometrial cancer. Interrogation of the TCGA database revealed upregulation of several isoforms in endometrial cancer, of which the ALDH1 isoforms collectively constituted the largest group. To translate its expression, a tissue microarray was previously constructed which contained a wide sampling of benign and malignant endometrial samples. The array contained a metachronous cohort of samples from individuals who either developed or did not develop endometrial cancer. Immunohistochemical staining was used to determine the intensity and frequency of ALDH1 expression. While benign proliferative and secretory endometrium showed very low levels of ALDH1, slightly higher expression was observed within the stratum basalis. In disease progression, cytoplasmic ALDH1 expression showed a step-wise increase between endometrial hyperplasia, atypical hyperplasia, and endometrial cancer. ALDH1 was also shown to be an early predictor of EC development, suggesting that it can serve as an independent prognostic indicator of patients with endometrial hyperplasia with or without atypia who would progress to cancer (p = 0.012).


Subject(s)
Aldehyde Dehydrogenase 1 Family/genetics , Biomarkers, Tumor/genetics , Endometrial Hyperplasia/genetics , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Precancerous Conditions/genetics , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase 1 Family/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Progression , Endometrial Hyperplasia/enzymology , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Prognosis
9.
Am J Pathol ; 174(5): 1619-28, 2009 May.
Article in English | MEDLINE | ID: mdl-19349354

ABSTRACT

Cancer cells exhibit alterations in histone modification patterns at individual genes and globally at the level of single nuclei in individual cells. We demonstrated previously that lower global/cellular levels of histone H3 lysine 4 dimethylation (H3K4me2) and H3K18 acetylation (ac) predict a higher risk of prostate cancer recurrence. Here we show that the cellular levels of both H3K4me2 and H3K18ac also predict clinical outcome in both lung and kidney cancer patients, with lower levels predicting significantly poorer survival probabilities in both cancer groups. We also show that lower cellular levels of H3K9me2, a modification associated with both gene activity and repression, is also prognostic of poorer outcome for individuals with either prostate or kidney cancers. The predictive power of these histone modifications was independent of tissue-specific clinicopathological variables, the proliferation marker Ki-67, or a p53 tumor suppressor mutation. Chromatin immunoprecipitation experiments indicated that the lower cellular levels of histone modifications in more aggressive cancer cell lines correlated with lower levels of modifications at DNA repetitive elements but not with gene promoters across the genome. Our results suggest that lower global levels of histone modifications are predictive of a more aggressive cancer phenotype, revealing a surprising commonality in prognostic epigenetic patterns of adenocarcinomas of different tissue origins.


Subject(s)
Adenocarcinoma/diagnosis , Histones/metabolism , Kidney Neoplasms/diagnosis , Lung Neoplasms/diagnosis , Methylation , Prostatic Neoplasms/diagnosis , Acetylation , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Cohort Studies , Female , Histones/genetics , Humans , Immunoenzyme Techniques , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Repetitive Sequences, Nucleic Acid , Tissue Array Analysis
10.
BMC Cancer ; 10: 680, 2010 Dec 13.
Article in English | MEDLINE | ID: mdl-21144054

ABSTRACT

BACKGROUND: The protein AGR2 is a putative member of the protein disulfide isomerase family and was first identified as a homolog of the Xenopus laevis gene XAG-2. AGR2 has been implicated in a number of human cancers. In particular, AGR2 has previously been found to be one of several genes that encode secreted proteins showing increased expression in prostate cancer cells compared to normal prostatic epithelium. METHODS: Gene expression levels of AGR2 were examined in prostate cancer cells by microarray analysis. We further examined the relationship of AGR2 protein expression to histopathology and prostate cancer outcome on a population basis using tissue microarray technology. RESULTS: At the RNA and protein level, there was an increase in AGR2 expression in adenocarcinoma of the prostate compared to morphologically normal prostatic glandular epithelium. Using a tissue microarray, this enhanced AGR2 expression was seen as early as premalignant PIN lesions. Interestingly, within adenocarcinoma samples, there was a slight trend toward lower levels of AGR2 with increasing Gleason score. Consistent with this, relatively lower levels of AGR2 were highly predictive of disease recurrence in patients who had originally presented with high-stage primary prostate cancer (P = 0.009). CONCLUSIONS: We have shown for the first time that despite an increase in AGR2 expression in prostate cancer compared to non-malignant cells, relatively lower levels of AGR2 are highly predictive of disease recurrence following radical prostatectomy.


Subject(s)
Adenocarcinoma/enzymology , Precancerous Conditions/enzymology , Prostatic Neoplasms/enzymology , Proteins/analysis , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Aged , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Los Angeles , Lymphatic Metastasis , Male , Middle Aged , Mucoproteins , Neoplasm Recurrence, Local , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Oncogene Proteins , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proportional Hazards Models , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Proteins/genetics , RNA, Messenger/analysis , Risk Assessment , Risk Factors , Tissue Array Analysis/methods , Treatment Outcome
11.
Mol Cancer Ther ; 19(8): 1682-1695, 2020 08.
Article in English | MEDLINE | ID: mdl-32451329

ABSTRACT

Little is known about the role of epithelial membrane protein-2 (EMP2) in breast cancer development or progression. In this study, we tested the hypothesis that EMP2 may regulate the formation or self-renewal of breast cancer stem cells (BCSC) in the tumor microenvironment. In silico analysis of gene expression data demonstrated a correlation of EMP2 expression with known metastasis-related genes and markers of cancer stem cells (CSC) including aldehyde dehydrogenase (ALDH). In breast cancer cell lines, EMP2 overexpression increased and EMP2 knockdown decreased the proportion of stem-like cells as assessed by the expression of the CSC markers CD44+/CD24-, ALDH activity, or by tumor sphere formation. In vivo, upregulation of EMP2 promoted tumor growth, whereas knockdown reduced the ALDHhigh CSC population as well as retarded tumor growth. Mechanistically, EMP2 functionally regulated the response to hypoxia through the upregulation of HIF-1α, a transcription factor previously shown to regulate the self-renewal of ALDHhigh CSCs. Furthermore, in syngeneic mouse models and primary human tumor xenografts, mAbs directed against EMP2 effectively targeted CSCs, reducing the ALDH+ population and blocking their tumor-initiating capacity when implanted into secondary untreated mice. Collectively, our results show that EMP2 increases the proportion of tumor-initiating cells, providing a rationale for the continued development of EMP2-targeting agents.


Subject(s)
Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Membrane Glycoproteins/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment/immunology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Placenta ; 81: 9-17, 2019 06.
Article in English | MEDLINE | ID: mdl-31138432

ABSTRACT

OBJECTIVE: Intrauterine growth restriction (IUGR) is a complication of pregnancy that has both short- and long-term sequelae for affected mothers and offspring. The pathophysiology of disease stems from poor nutrient and oxygen provision to the fetus, resulting in increased oxidative stress within the placenta. As the milieu within the local microenvironment alters macrophage differentiation, we hypothesized that macrophage plasticity may be altered in placentas associated with IUGR, and that macrophages would show hallmarks of lipid peroxidation including altered aldehyde metabolism. METHODS: In human placentas taken from normal pregnancies resulting in appropriate-for-gestational-age (AGA) newborns and placentas associated with IUGR, placental macrophages were evaluated by immunohistochemistry and shown in IUGR to resemble pro-inflammatory activated M1-type macrophages. To link oxidative stress to macrophages, the expression of aldehyde dehydrogenase (ALDHs) isozymes ALDH1, ALDH2, and ALDH3 was assessed. RESULTS: All three isozymes displayed preferential staining for distinct cellular populations within the term human placenta. ALDH1 and ALDH2 were strongly expressed in placental Hofbauer and decidual stromal cells. ALDH3, in contrast, was present in extravillous trophoblasts. Comparing AGA and IUGR-associated placentas, ALDH1 and ALDH2 trended to have greater expression in macrophage populations but lower expression in decidual cell populations in IUGR-associated placentas. ALDH3 had higher expression in IUGR-associated placentas but localized specifically to extravillous trophoblast populations. CONCLUSION: Therefore, we speculate that specific ALDH isozymes have cell-specific functions related to differentiation, inflammation, or oxidative stress responses that are altered in IUGR-associated term human placentas. This family of isozymes may be a novel method to identify human placentas affected by placental insufficiency/IUGR.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Fetal Growth Retardation/enzymology , Macrophages/metabolism , Placenta/enzymology , Adult , Female , Fetal Growth Retardation/immunology , Humans , Pregnancy , Protein Isoforms/metabolism
13.
Cell Rep ; 14(7): 1590-1601, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876179

ABSTRACT

Monocarboxylate transporter 1 (MCT1) inhibition is thought to block tumor growth through disruption of lactate transport and glycolysis. Here, we show MCT1 inhibition impairs proliferation of glycolytic breast cancer cells co-expressing MCT1 and MCT4 via disruption of pyruvate rather than lactate export. MCT1 expression is elevated in glycolytic breast tumors, and high MCT1 expression predicts poor prognosis in breast and lung cancer patients. Acute MCT1 inhibition reduces pyruvate export but does not consistently alter lactate transport or glycolytic flux in breast cancer cells that co-express MCT1 and MCT4. Despite the lack of glycolysis impairment, MCT1 loss-of-function decreases breast cancer cell proliferation and blocks growth of mammary fat pad xenograft tumors. Our data suggest MCT1 expression is elevated in glycolytic cancers to promote pyruvate export that when inhibited, enhances oxidative metabolism and reduces proliferation. This study presents an alternative molecular consequence of MCT1 inhibitors, further supporting their use as anti-cancer therapeutics.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Monocarboxylic Acid Transporters/genetics , Muscle Proteins/genetics , Pyruvic Acid/metabolism , Symporters/genetics , Animals , Antineoplastic Agents/pharmacology , Biological Transport , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Profiling , Glycolysis/drug effects , Glycolysis/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Oxidative Phosphorylation/drug effects , Pyrimidinones/pharmacology , Signal Transduction , Symporters/antagonists & inhibitors , Symporters/metabolism , Thiophenes/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Crit Rev Oncog ; 20(5-6): 349-55, 2015.
Article in English | MEDLINE | ID: mdl-27279234

ABSTRACT

Lung cancer is the leading cause of cancer deaths in men and women, both because of its high incidence rates and poor prognosis without effective therapies. Environmental carcinogens, most predominantly tobacco smoke, play a significant role. There are continuously emerging data to suggest the biological process differs between lung cancers in men and women. Differences are seen in a variety of cellular pathways and responses to carcinogens and therapies. Particular note in this article is made of carcinogen processing by cytochrome P450s, estrogen receptor pathways, epidermal growth factor receptor mutations, and how these are not necessarily independent cellular processes. These topics are only very briefly summarized here and it was not possible to include many important references. The heterogeneity of lung cancers in men and women, as well as smokers and nonsmokers, are likely to become more apparent with further studies. Work previously done in our laboratory (EDRN, PIs David Chia & Lee Goodglick) served to further emphasize these differences. This report is dedicated to the memory of Lee Goodglick with whom I had the privilege to work for many years prior to his untimely death.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Carcinogens/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cytochrome P-450 Enzyme System/metabolism , ErbB Receptors/genetics , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/epidemiology , Lung Neoplasms/physiopathology , Male , Mutation , Receptors, Estrogen/metabolism , Sex Factors , Signal Transduction , Treatment Outcome
15.
Biomed Res Int ; 2015: 925703, 2015.
Article in English | MEDLINE | ID: mdl-25874233

ABSTRACT

Triple-negative breast cancer (TNBC) occurs in 10-15% of patients yet accounts for almost half of all breast cancer deaths. TNBCs lack expression of estrogen and progesterone receptors and HER-2 overexpression and cannot be treated with current targeted therapies. TNBCs often occur in African American and younger women. Although initially responsive to some chemotherapies, TNBCs tend to relapse and metastasize. Thus, it is critical to find new therapeutic targets. A second ER gene product, termed ERß, in the absence of ERα may be such a target. Using human TNBC specimens with known clinical outcomes to assess ERß expression, we find that ERß1 associates with significantly worse 5-year overall survival. Further, a panel of TNBC cell lines exhibit significant levels of ERß protein. To assess ERß effects on proliferation, ERß expression in TNBC cells was silenced using shRNA, resulting in a significant reduction in TNBC proliferation. ERß-specific antagonists similarly suppressed TNBC growth. Growth-stimulating effects of ERß may be due in part to downstream actions that promote VEGF, amphiregulin, and Wnt-10b secretion, other factors associated with tumor promotion. In vivo, insulin-like growth factor-2 (IGF-2), along with ERß1, is significantly expressed in TNBC and stimulates high ERß mRNA in TNBC cells. This work may help elucidate the interplay of metabolic and growth factors in TNBC.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor beta/biosynthesis , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/metabolism , Neoplasm Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Cell Proliferation , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Female , Humans , Insulin-Like Growth Factor II/antagonists & inhibitors , Insulin-Like Growth Factor II/genetics , MCF-7 Cells , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics
16.
Crit Rev Oncog ; 20(5-6): 373-90, 2015.
Article in English | MEDLINE | ID: mdl-27279236

ABSTRACT

Triple-negative breast cancers (TNBCs) lack estrogen receptor-α (ERα), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) amplification and account for almost half of all breast cancer deaths. This breast cancer subtype largely affects women who are premenopausal, African-American, or have BRCA1/2 mutations. Women with TNBC are plagued with higher rates of distant metastasis that significantly diminish their overall survival and quality of life. Due to their poor response to chemotherapy, patients with TNBC would significantly benefit from development of new targeted therapeutics. Research suggests that the insulin-like growth factor (IGF) family and estrogen receptor beta-1 (ERß1), due to their roles in metabolism and cellular regulation, might be attractive targets to pursue for TNBC management. Here, we review the current state of the science addressing the roles of ERß1 and the IGF family in TNBC. Further, the potential benefit of metformin treatment in patients with TNBC as well as areas of therapeutic potential in the IGF-ERß1 pathway are highlighted.


Subject(s)
Antineoplastic Agents/therapeutic use , Estrogen Receptor beta/drug effects , Molecular Targeted Therapy , Somatomedins/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Female , Humans
17.
Aging (Albany NY) ; 7(5): 294-306, 2015 May.
Article in English | MEDLINE | ID: mdl-26000617

ABSTRACT

Studies that elucidate why some human tissues age faster than others may shed light on how we age, and ultimately suggest what interventions may be possible. Here we utilize a recent biomarker of aging (referred to as epigenetic clock) to assess the epigenetic ages of up to 30 anatomic sites from supercentenarians (subjects who reached an age of 110 or older) and younger subjects. Using three novel and three published human DNA methylation data sets, we demonstrate that the cerebellum ages more slowly than other parts of the human body. We used both transcriptional data and genetic data to elucidate molecular mechanisms which may explain this finding. The two largest superfamilies of helicases (SF1 and SF2) are significantly over-represented (p=9.2x10-9) among gene transcripts that are over-expressed in the cerebellum compared to other brain regions from the same subject. Furthermore, SNPs that are associated with epigenetic age acceleration in the cerebellum tend to be located near genes from helicase superfamilies SF1 and SF2 (enrichment p=5.8x10-3). Our genetic and transcriptional studies of epigenetic age acceleration support the hypothesis that the slow aging rate of the cerebellum is due to processes that involve RNA helicases.


Subject(s)
Aged, 80 and over , Aging/genetics , Cerebellum/physiology , Epigenesis, Genetic , RNA Helicases/genetics , Adult , Humans , Middle Aged , Transcriptome
18.
PLoS One ; 10(5): e0127600, 2015.
Article in English | MEDLINE | ID: mdl-26001082

ABSTRACT

BACKGROUND: Ribonucleotide reductase catalyzes the conversion of ribonucleotide diphosphates to deoxyribonucleotide diphosphates. The functional enzyme consists of two subunits - one large (RRM1) and one small (RRM2 or RRM2b) subunit. Expression levels of each subunit have been implicated in prognostic outcomes in several different types of cancers. EXPERIMENTAL DESIGN: Immunohistochemistry for RRM1 and RRM2 was performed on a lung cancer tissue microarray (TMA) and analyzed. 326 patients from the microarray were included in this study. RESULTS: In non-small cell lung cancer (NSCLC), RRM2 expression was strongly predictive of disease-specific survival in women, non-smokers and former smokers who had quit at least 10 years prior to being diagnosed with lung cancer. Higher expression was associated with worse survival. This was not the case for men, current smokers and those who had stopped smoking for shorter periods of time. RRM1 was not predictive of survival outcomes in any subset of the patient group. CONCLUSION: RRM2, but not RRM1, is a useful predictor of survival outcome in certain subsets of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Prognosis , Ribonucleoside Diphosphate Reductase/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Sex Factors , Smoking , Survival Rate
19.
Lung Cancer ; 74(2): 318-25, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21511357

ABSTRACT

Estrogen signaling pathways may play a significant role in the pathogenesis of non-small cell lung cancers (NSCLC) as evidenced by the expression of aromatase and estrogen receptors (ERα and ERß) in many of these tumors. Here we examine whether ERα and ERß levels in conjunction with aromatase define patient groups with respect to survival outcomes and possible treatment regimens. Immunohistochemistry was performed on a high-density tissue microarray with resulting data and clinical information available for 377 patients. Patients were subdivided by gender, age and tumor histology, and survival data was determined using the Cox proportional hazards model and Kaplan-Meier curves. Neither ERα nor ERß alone was predictor of survival in NSCLC. However, when coupled with aromatase expression, higher ERß levels predicted worse survival in patients whose tumors expressed higher levels of aromatase. Although this finding was present in patients of both genders, it was especially pronounced in women ≥ 65 years old, where higher expression of both ERß and aromatase indicated a markedly worse survival rate than that determined by aromatase alone. Expression of ERß together with aromatase has predictive value for survival in different gender and age subgroups of NSCLC patients. This predictive value is stronger than each individual marker alone. Our results suggest treatment with aromatase inhibitors alone or combined with estrogen receptor modulators may be of benefit in some subpopulations of these patients.


Subject(s)
Aromatase/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Estrogen Receptor beta/metabolism , Lung Neoplasms/diagnosis , Aged , Aromatase/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Estrogen Receptor beta/genetics , Female , Humans , Immunohistochemistry , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Male , Microarray Analysis , Middle Aged , Predictive Value of Tests , Prognosis , Smoking , Survival Analysis
20.
Steroids ; 76(9): 910-20, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21600232

ABSTRACT

Lung cancer is the most common cause of cancer mortality in male and female patients in the US. Although it is clear that tobacco smoking is a major cause of lung cancer, about half of all women with lung cancer worldwide are never-smokers. Despite a declining smoking population, the incidence of non-small cell lung cancer (NSCLC), the predominant form of lung cancer, has reached epidemic proportions particularly in women. Emerging data suggest that factors other than tobacco, namely endogenous and exogenous female sex hormones, have a role in stimulating NSCLC progression. Aromatase, a key enzyme for estrogen biosynthesis, is expressed in NSCLC. Clinical data show that women with high levels of tumor aromatase (and high intratumoral estrogen) have worse survival than those with low aromatase. The present and previous studies also reveal significant expression and activity of estrogen receptors (ERα, ERß) in both extranuclear and nuclear sites in most NSCLC. We now report further on the expression of progesterone receptor (PR) transcripts and protein in NSCLC. PR transcripts were significantly lower in cancerous as compared to non-malignant tissue. Using immunohistochemistry, expression of PR was observed in the nucleus and/or extranuclear compartments in the majority of human tumor specimens examined. Combinations of estrogen and progestins administered in vitro cooperate in promoting tumor secretion of vascular endothelial growth factor and, consequently, support tumor-associated angiogenesis. Further, dual treatment with estradiol and progestin increased the numbers of putative tumor stem/progenitor cells. Thus, ER- and/or PR-targeted therapies may offer new approaches to manage NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , AC133 Antigen , Aldehyde Dehydrogenase/metabolism , Animals , Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Culture Media, Conditioned , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Estradiol/pharmacology , Estradiol/physiology , Estrogens/pharmacology , Estrogens/physiology , Female , Glycoproteins/metabolism , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , Mifepristone/pharmacology , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Progestins/antagonists & inhibitors , Progestins/pharmacology , Receptors, Progesterone/genetics , Transcription, Genetic , Umbilical Cord/cytology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL