Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Clin Invest ; 129(12): 5343-5356, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31682236

ABSTRACT

Both miRNAs and A-to-I RNA editing, a widespread nucleotide modification mechanism, have recently emerged as key players in cancer pathophysiology. However, the functional impact of RNA editing of miRNAs in cancer remains largely unexplored. Here, we focused on an ADAR2-catalyzed RNA editing site within the miR-379-5p seed region. This site was under-edited in tumors relative to normal tissues, with a high editing level being correlated with better patient survival times across cancer types. We demonstrated that in contrast to wild-type miRNA, edited miR-379-5p inhibited cell proliferation and promoted apoptosis in diverse tumor contexts in vitro, which was due to the ability of edited but not wild-type miR-379-5p to target CD97. Importantly, through nanoliposomal delivery, edited miR-379-5p mimics significantly inhibited tumor growth and extended survival of mice. Our study indicates a role of RNA editing in diversifying miRNA function during cancer progression and highlights the translational potential of edited miRNAs as a new class of cancer therapeutics.


Subject(s)
Antigens, CD/physiology , Apoptosis , MicroRNAs/physiology , Neoplasms/therapy , RNA Editing , Receptors, G-Protein-Coupled/physiology , Animals , Antigens, CD/genetics , Cell Line, Tumor , Cell Proliferation , Female , Mice , Neoplasms/pathology , Receptors, G-Protein-Coupled/genetics
2.
Cancer Cell ; 33(5): 817-828.e7, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29706454

ABSTRACT

Adenosine (A) to inosine (I) RNA editing introduces many nucleotide changes in cancer transcriptomes. However, due to the complexity of post-transcriptional regulation, the contribution of RNA editing to proteomic diversity in human cancers remains unclear. Here, we performed an integrated analysis of TCGA genomic data and CPTAC proteomic data. Despite limited site diversity, we demonstrate that A-to-I RNA editing contributes to proteomic diversity in breast cancer through changes in amino acid sequences. We validate the presence of editing events at both RNA and protein levels. The edited COPA protein increases proliferation, migration, and invasion of cancer cells in vitro. Our study suggests an important contribution of A-to-I RNA editing to protein diversity in cancer and highlights its translational potential.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Proteomics/methods , RNA Editing , Adenosine/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Inosine/genetics , Sequence Analysis, RNA , Tandem Mass Spectrometry
3.
Mol Cell Proteomics ; 17(6): 1245-1258, 2018 06.
Article in English | MEDLINE | ID: mdl-29531020

ABSTRACT

Molecular analysis of tumors forms the basis for personalized cancer medicine and increasingly guides patient selection for targeted therapy. Future opportunities for personalized medicine are highlighted by the measurement of protein expression levels via immunohistochemistry, protein arrays, and other approaches; however, sample type, sample quantity, batch effects, and "time to result" are limiting factors for clinical application. Here, we present a development pipeline for a novel multiplexed DNA-labeled antibody platform which digitally quantifies protein expression from lysate samples. We implemented a rigorous validation process for each antibody and show that the platform is amenable to multiple protocols covering nitrocellulose and plate-based methods. Results are highly reproducible across technical and biological replicates, and there are no observed "batch effects" which are common for most multiplex molecular assays. Tests from basal and perturbed cancer cell lines indicate that this platform is comparable to orthogonal proteomic assays such as Reverse-Phase Protein Array, and applicable to measuring the pharmacodynamic effects of clinically-relevant cancer therapeutics. Furthermore, we demonstrate the potential clinical utility of the platform with protein profiling from breast cancer patient samples to identify molecular subtypes. Together, these findings highlight the potential of this platform for enhancing our understanding of cancer biology in a clinical translation setting.


Subject(s)
Antibodies/chemistry , DNA/chemistry , Neoplasms/metabolism , Proteins/metabolism , Cell Line, Tumor , Female , Humans , Proteomics
4.
Cancer Cell ; 33(3): 450-462.e10, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29533785

ABSTRACT

The functional impact of the vast majority of cancer somatic mutations remains unknown, representing a critical knowledge gap for implementing precision oncology. Here, we report the development of a moderate-throughput functional genomic platform consisting of efficient mutant generation, sensitive viability assays using two growth factor-dependent cell models, and functional proteomic profiling of signaling effects for select aberrations. We apply the platform to annotate >1,000 genomic aberrations, including gene amplifications, point mutations, indels, and gene fusions, potentially doubling the number of driver mutations characterized in clinically actionable genes. Further, the platform is sufficiently sensitive to identify weak drivers. Our data are accessible through a user-friendly, public data portal. Our study will facilitate biomarker discovery, prediction algorithm improvement, and drug development.


Subject(s)
Biomarkers, Tumor/genetics , Mutation/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Algorithms , Genomics , High-Throughput Nucleotide Sequencing/methods , Humans , Precision Medicine , Proteomics
5.
Genome Res ; 27(7): 1112-1125, 2017 07.
Article in English | MEDLINE | ID: mdl-28411194

ABSTRACT

RNA editing, a widespread post-transcriptional mechanism, has emerged as a new player in cancer biology. Recent studies have reported key roles for individual miRNA editing events, but a comprehensive picture of miRNA editing in human cancers remains largely unexplored. Here, we systematically characterized the miRNA editing profiles of 8595 samples across 20 cancer types from miRNA sequencing data of The Cancer Genome Atlas and identified 19 adenosine-to-inosine (A-to-I) RNA editing hotspots. We independently validated 15 of them by perturbation experiments in several cancer cell lines. These miRNA editing events show extensive correlations with key clinical variables (e.g., tumor subtype, disease stage, and patient survival time) and other molecular drivers. Focusing on the RNA editing hotspot in miR-200b, a key tumor metastasis suppressor, we found that the miR-200b editing level correlates with patient prognosis opposite to the pattern observed for the wild-type miR-200b expression. We further experimentally showed that, in contrast to wild-type miRNA, the edited miR-200b can promote cell invasion and migration through its impaired ability to inhibit ZEB1/ZEB2 and acquired concomitant ability to repress new targets, including LIFR, a well-characterized metastasis suppressor. Our study highlights the importance of miRNA editing in gene regulation and suggests its potential as a biomarker for cancer prognosis and therapy.


Subject(s)
Genes, Tumor Suppressor , MicroRNAs/metabolism , Neoplasms/metabolism , RNA Editing , RNA, Neoplasm/metabolism , Adenosine/genetics , Adenosine/metabolism , Female , Humans , Inosine/genetics , Inosine/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Male , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , RNA, Neoplasm/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
6.
Sci Rep ; 6: 27391, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27272709

ABSTRACT

Mutation of PPP2R1A has been observed at high frequency in endometrial serous carcinomas but at low frequency in ovarian clear cell carcinoma. However, the biological role of mutation of PPP2R1A in ovarian and endometrial cancer progression remains unclear. In this study, we found that PPP2R1A expression is elevated in high-grade primary tumor patients with papillary serous tumors of the ovary. To determine whether increased levels or mutation of PPP2R1A might contribute to cancer progression, the effects of overexpression or mutation of PPP2R1A on cell proliferation, migration, and PP2A phosphatase activity were investigated using ovarian and endometrial cancer cell lines. Among the mutations, PPP2R1A-W257G enhanced cell migration in vitro through activating SRC-JNK-c-Jun pathway. Overexpression of wild type (WT) PPP2R1A increased its binding ability with B56 regulatory subunits, whereas PPP2R1A-mutations lost the ability to bind to most B56 subunits except B56δ. Total PP2A activity and PPP2R1A-associated PP2Ac activity were significantly increased in cells overexpressing PPP2R1A-WT. In addition, overexpression of PPP2R1A-WT increased cell proliferation in vitro and tumor growth in vivo.


Subject(s)
MAP Kinase Signaling System , Mutation , Neoplasm Metastasis/genetics , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Cell Proliferation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation
7.
Cell Rep ; 15(7): 1493-1504, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160903

ABSTRACT

3q26.2 amplification in high-grade serous ovarian cancer leads to increased expression of mature microRNA miR551b-3p, which is associated with poor clinical outcome. Importantly, miR551b-3p contributes to resistance to apoptosis and increased survival and proliferation of cancer cells in vitro and in vivo. miR551b-3p upregulates STAT3 protein levels, and STAT3 is required for the effects of miR551b-3p on cell proliferation. Rather than decreasing levels of target mRNA as expected, we demonstrate that miR551b-3p binds a complementary sequence on the STAT3 promoter, recruiting RNA polymerase II and the TWIST1 transcription factor to activate STAT3 transcription, and thus directly upregulates STAT3 expression. Furthermore, anti-miR551b reduced STAT3 expression in ovarian cancer cells in vitro and in vivo and reduced ovarian cancer growth in vivo. Together, our data demonstrate a role for miR551b-3p in transcriptional activation. Thus, miR551b-3p represents a promising candidate biomarker and therapeutic target in ovarian cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/genetics , Up-Regulation/genetics , Animals , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation/genetics , Cell Self Renewal , Cell Survival/genetics , Down-Regulation/genetics , Female , Gene Amplification , Gene Knockdown Techniques , Gene Silencing , Humans , Mice, Nude , Neoplasm Metastasis , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , STAT3 Transcription Factor/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription, Genetic , Treatment Outcome , Tumor Burden/genetics , Twist-Related Protein 1/metabolism
8.
Cancer Cell ; 28(4): 515-528, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26439496

ABSTRACT

Adenosine-to-inosine (A-to-I) RNA editing is a widespread post-transcriptional mechanism, but its genomic landscape and clinical relevance in cancer have not been investigated systematically. We characterized the global A-to-I RNA editing profiles of 6,236 patient samples of 17 cancer types from The Cancer Genome Atlas and revealed a striking diversity of altered RNA-editing patterns in tumors relative to normal tissues. We identified an appreciable number of clinically relevant editing events, many of which are in noncoding regions. We experimentally demonstrated the effects of several cross-tumor nonsynonymous RNA editing events on cell viability and provide the evidence that RNA editing could selectively affect drug sensitivity. These results highlight RNA editing as an exciting theme for investigating cancer mechanisms, biomarkers, and treatments.


Subject(s)
Adenosine/metabolism , Inosine/metabolism , Neoplasms/genetics , RNA Editing , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Genome, Human , Humans , Neoplasms/pathology
9.
Cancer Cell ; 26(6): 863-879, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25490449

ABSTRACT

Small noncoding miRNAs represent underexplored targets of genomic aberrations and emerging therapeutic targets. The 3q26.2 amplicon is among the most frequent genomic aberrations in multiple cancer lineages including ovarian and breast cancers. We demonstrate that hsa-miR-569 (hereafter designated as miR569), which is overexpressed in a subset of ovarian and breast cancers, at least in part due to the 3q26.2 amplicon, alters cell survival and proliferation. Downregulation of TP53INP1 expression by miR569 is required for the effects of miR569 on survival and proliferation. Targeting miR569 sensitizes ovarian and breast cancer cells overexpressing miR569 to cisplatin by increasing cell death both in vitro and in vivo. Thus targeting miR569 could potentially benefit patients with the 3q26.2 amplicon and subsequent miR569 elevation.


Subject(s)
Breast Neoplasms/genetics , Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , MicroRNAs/metabolism , Neoplasms, Glandular and Epithelial/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromosomes, Human, Pair 3 , Cisplatin/pharmacology , Female , Gene Amplification , Gene Duplication , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasms, Experimental , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology
10.
Cancer Cell ; 26(4): 479-94, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25284480

ABSTRACT

PIK3R1 (p85α regulatory subunit of PI3K) is frequently mutated across cancer lineages. Herein, we demonstrate that the most common recurrent PIK3R1 mutation PIK3R1(R348∗) and a nearby mutation PIK3R1(L370fs), in contrast to wild-type and mutations in other regions of PIK3R1, confers an unexpected sensitivity to MEK and JNK inhibitors in vitro and in vivo. Consistent with the response to inhibitors, PIK3R1(R348∗) and PIK3R1(L370fs) unexpectedly increase JNK and ERK phosphorylation. Surprisingly, p85α R348(∗) and L370fs localize to the nucleus where the mutants provide a scaffold for multiple JNK pathway components facilitating nuclear JNK pathway activation. Our findings uncover an unexpected neomorphic role for PIK3R1(R348∗) and neighboring truncation mutations in cellular signaling, providing a rationale for therapeutic targeting of these mutant tumors.


Subject(s)
MAP Kinase Signaling System/drug effects , Mutation , Phosphatidylinositol 3-Kinases/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Nucleus/metabolism , Class Ia Phosphatidylinositol 3-Kinase , Enzyme Activation , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Transport
11.
Genome Res ; 22(11): 2120-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23028188

ABSTRACT

Endometrial cancer is the most common gynecological malignancy, with more than 280,000 cases occurring annually worldwide. Although previous studies have identified important common somatic mutations in endometrial cancer, they have primarily focused on a small set of known cancer genes and have thus provided a limited view of the molecular basis underlying this disease. Here we have developed an integrated systems-biology approach to identifying novel cancer genes contributing to endometrial tumorigenesis. We first performed whole-exome sequencing on 13 endometrial cancers and matched normal samples, systematically identifying somatic alterations with high precision and sensitivity. We then combined bioinformatics prioritization with high-throughput screening (including both shRNA-mediated knockdown and expression of wild-type and mutant constructs) in a highly sensitive cell viability assay. Our results revealed 12 potential driver cancer genes including 10 tumor-suppressor candidates (ARID1A, INHBA, KMO, TTLL5, GRM8, IGFBP3, AKTIP, PHKA2, TRPS1, and WNT11) and two oncogene candidates (ERBB3 and RPS6KC1). The results in the "sensor" cell line were recapitulated by siRNA-mediated knockdown in endometrial cancer cell lines. Focusing on ARID1A, we integrated mutation profiles with functional proteomics in 222 endometrial cancer samples, demonstrating that ARID1A mutations frequently co-occur with mutations in the phosphatidylinositol 3-kinase (PI3K) pathway and are associated with PI3K pathway activation. siRNA knockdown in endometrial cancer cell lines increased AKT phosphorylation supporting ARID1A as a novel regulator of PI3K pathway activity. Our study presents the first unbiased view of somatic coding mutations in endometrial cancer and provides functional evidence for diverse driver genes and mutations in this disease.


Subject(s)
Endometrial Neoplasms/genetics , Exome , Genes, Tumor Suppressor , Genomics , High-Throughput Nucleotide Sequencing , Oncogenes , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Female , Humans , Mutation , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , RNA, Small Interfering , Sequence Analysis, DNA , Systems Biology
12.
PLoS One ; 7(6): e39400, 2012.
Article in English | MEDLINE | ID: mdl-22745748

ABSTRACT

Autophagic cell death or abortive autophagy has been proposed to eliminate damaged as well as cancer cells, but there remains a critical gap in our knowledge in how this process is regulated. The goal of this study was to identify modulators of the autophagic cell death pathway and elucidate their effects on cellular signaling and function. The result of our siRNA library screenings show that an intact coatomer complex I (COPI) is obligatory for productive autophagy. Depletion of COPI complex members decreased cell survival and impaired productive autophagy which preceded endoplasmic reticulum stress. Further, abortive autophagy provoked by COPI depletion significantly altered growth factor signaling in multiple cancer cell lines. Finally, we show that COPI complex members are overexpressed in an array of cancer cell lines and several types of cancer tissues as compared to normal cell lines or tissues. In cancer tissues, overexpression of COPI members is associated with poor prognosis. Our results demonstrate that the coatomer complex is essential for productive autophagy and cellular survival, and thus inhibition of COPI members may promote cell death of cancer cells when apoptosis is compromised.


Subject(s)
Autophagy/physiology , Cell Death/physiology , Endoplasmic Reticulum Stress/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Autophagy/genetics , Blotting, Western , Cell Death/genetics , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum Stress/genetics , Female , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , RNA, Small Interfering
13.
Cancer Discov ; 1(2): 170-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21984976

ABSTRACT

We demonstrate that phosphatidylinositol 3-kinase (PI3K) pathway aberrations occur in >80% of endometrioid endometrial cancers, with coordinate mutations of multiple PI3K pathway members being more common than predicted by chance. PIK3R1 (p85α) mutations occur at a higher rate in endometrial cancer than in any other tumor lineage, and PIK3R2 (p85ß), not previously demonstrated to be a cancer gene, is also frequently mutated. The dominant activation event in the PI3K pathway appears to be PTEN protein loss. However, in tumors with retained PTEN protein, PI3K pathway mutations phenocopy PTEN loss, resulting in pathway activation. KRAS mutations are common in endometrioid tumors activating independent events from PI3K pathway aberrations. Multiple PIK3R1 and PIK3R2 mutations demonstrate gain of function, including disruption of a novel mechanism of pathway regulation wherein p85α dimers bind and stabilize PTEN. Taken together, the PI3K pathway represents a critical driver of endometrial cancer pathogenesis and a novel therapeutic target.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Endometrial Neoplasms/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Cell Line, Transformed , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Endometrial Neoplasms/metabolism , Female , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras) , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
14.
Mol Cancer ; 9: 140, 2010 Jun 09.
Article in English | MEDLINE | ID: mdl-20529378

ABSTRACT

BACKGROUND: Although the incidence of melanoma in the U.S. is rising faster than any other cancer, the FDA-approved chemotherapies lack efficacy for advanced disease, which results in poor overall survival. Lysophosphatidic acid (LPA), autotaxin (ATX), the enzyme that produces LPA, and the LPA receptors represent an emerging group of therapeutic targets in cancer, although it is not known which of these is most effective. RESULTS: Herein we demonstrate that thio-ccPA 18:1, a stabilized phosphonothionate analogue of carba cyclic phosphatidic acid, ATX inhibitor and LPA1/3 receptor antagonist, induced a marked reduction in the viability of B16F10 metastatic melanoma cells compared with PBS-treated control by 80-100%. Exogenous LPA 18:1 or D-sn-1-O-oleoyl-2-O-methylglyceryl-3-phosphothioate did not reverse the effect of thio-ccPA 18:1. The reduction in viability mediated by thio-ccPA 18:1 was also observed in A375 and MeWo melanoma cell lines, suggesting that the effects are generalizable. Interestingly, siRNA to LPA3 (siLPA3) but not other LPA receptors recapitulated the effects of thio-ccPA 18:1 on viability, suggesting that inhibition of the LPA3 receptor is an important dualistic function of the compound. In addition, siLPA3 reduced proliferation, plasma membrane integrity and altered morphology of A375 cells. Another experimental compound designed to antagonize the LPA1/3 receptors significantly reduced viability in MeWo cells, which predominantly express the LPA3 receptor. CONCLUSIONS: Thus the ability of thio-ccPA 18:1 to inhibit the LPA3 receptor and ATX are key to its molecular mechanism, particularly in melanoma cells that predominantly express the LPA3 receptor. These observations necessitate further exploration and exploitation of these targets in melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma, Experimental/drug therapy , Phosphatidic Acids/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Multienzyme Complexes/antagonists & inhibitors , Phosphodiesterase I/antagonists & inhibitors , Phosphoric Diester Hydrolases , Pyrophosphatases/antagonists & inhibitors , RNA, Small Interfering , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
15.
Cancer Cell ; 15(6): 539-50, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19477432

ABSTRACT

Lysophosphatidic acid (LPA) acts through high-affinity G protein-coupled receptors to mediate a plethora of physiological and pathological activities associated with tumorigenesis. LPA receptors and autotaxin (ATX/LysoPLD), the primary enzyme producing LPA, are aberrantly expressed in multiple cancer lineages. However, the role of ATX and LPA receptors in the initiation and progression of breast cancer has not been evaluated. We demonstrate that expression of ATX or each edg family LPA receptor in mammary epithelium of transgenic mice is sufficient to induce a high frequency of late-onset, estrogen receptor (ER)-positive, invasive, and metastatic mammary cancer. Thus, ATX and LPA receptors can contribute to the initiation and progression of breast cancer.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Adenosquamous/metabolism , Mammary Neoplasms, Experimental/metabolism , Multienzyme Complexes/metabolism , Phosphodiesterase I/metabolism , Pyrophosphatases/metabolism , Receptors, Lysophosphatidic Acid/physiology , Adenocarcinoma/secondary , Animals , Carcinoma, Adenosquamous/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cloning, Molecular , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Multienzyme Complexes/genetics , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Phosphodiesterase I/genetics , Phosphoric Diester Hydrolases , Pyrophosphatases/genetics , Receptors, Estrogen/metabolism , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction/physiology
16.
PLoS One ; 4(5): e5583, 2009.
Article in English | MEDLINE | ID: mdl-19440550

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) governs a number of physiologic and pathophysiological processes. Malignant ascites fluid is rich in LPA, and LPA receptors are aberrantly expressed by ovarian cancer cells, implicating LPA in the initiation and progression of ovarian cancer. However, there is an absence of systematic data critically analyzing the transcriptional changes induced by LPA in ovarian cancer. METHODOLOGY AND PRINCIPAL FINDINGS: In this study, gene expression profiling was used to examine LPA-mediated transcription by exogenously adding LPA to human epithelial ovarian cancer cells for 24 h to mimic long-term stimulation in the tumor microenvironment. The resultant transcriptional profile comprised a 39-gene signature that closely correlated to serous epithelial ovarian carcinoma. Hierarchical clustering of ovarian cancer patient specimens demonstrated that the signature is associated with worsened prognosis. Patients with LPA-signature-positive ovarian tumors have reduced disease-specific and progression-free survival times. They have a higher frequency of stage IIIc serous carcinoma and a greater proportion is deceased. Among the 39-gene signature, a group of seven genes associated with cell adhesion recapitulated the results. Out of those seven, claudin-1, an adhesion molecule and phenotypic epithelial marker, is the only independent biomarker of serous epithelial ovarian carcinoma. Knockdown of claudin-1 expression in ovarian cancer cells reduces LPA-mediated cellular adhesion, enhances suspended cells and reduces LPA-mediated migration. CONCLUSIONS: The data suggest that transcriptional events mediated by LPA in the tumor microenvironment influence tumor progression through modulation of cell adhesion molecules like claudin-1 and, for the first time, report an LPA-mediated expression signature in ovarian cancer that predicts a worse prognosis.


Subject(s)
Lysophospholipids/pharmacology , Ovarian Neoplasms/metabolism , Transcription, Genetic/drug effects , Blotting, Western , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Claudin-1 , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Membrane Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Transcription, Genetic/genetics
17.
Mol Oncol ; 2(1): 54-69, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19081821

ABSTRACT

Prostate cancer remains the most frequently diagnosed malignancy and the second leading cause of cancer mortality among men in the United States. Hormone refractory, metastatic disease has no molecular therapeutics to date and survival is poor. Lysophosphatidic acid (LPA) is a bioactive lipid exhibiting motility, invasive, growth, proliferative and survival effects in multiple cancer cell lineages. Cells express different combinations of LPA-specific G protein-coupled receptors, LPA(1), LPA(2) LPA(3), and LPA(4) as well as other LPA receptors, which bind LPA and thereby regulate lipid signaling. The role of specific LPA receptors in functional outcomes of lysolipid signaling remains to be fully elucidated in prostate cancer. We hypothesized that LPA can initiate cell migration through specific LPA receptors by activating actin-associating proteins involved in motility, including the vasodilator-stimulated phosphoprotein (VASP). In the present study, we demonstrate that LPA-induced lamellipodia formation in cells is dependent on LPA receptor-mediated phosphorylation of VASP, demonstrating a previously unknown regulation by LPA. LPA induces phosphorylation of VASP at Ser(157), through protein kinase A (PKA) since the stimulation was abrogated by PKA inhibition. In addition, we found the effects of LPA-induced lamellipodia formation and migration were reduced by knockdown of either VASP or LPA receptor expression, suggesting that LPA receptor-induced VASP phosphorylation is a critical mediator of migration initiation. Thus the LPA(2) and LPA(3) receptors, in addition to the previously implicated LPA(1) receptor, play a role in cellular motility potentially contributing to invasion and metastases. Emerging drugs targeting the LPA pathway may be beneficial for the treatment of metastatic progression in prostate cancer.


Subject(s)
Cell Movement/drug effects , Lysophospholipids/pharmacology , Phosphoproteins/metabolism , Prostatic Neoplasms/pathology , Pseudopodia/pathology , Receptors, Lysophosphatidic Acid/physiology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Humans , Male , Microfilament Proteins/metabolism , Prostatic Neoplasms/ultrastructure , Pseudopodia/drug effects
18.
J Natl Cancer Inst ; 100(22): 1630-42, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-19001604

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) acts through the cell surface G protein-coupled receptors, LPA1, LPA2, or LPA3, to elicit a wide range of cellular responses. It is present at high levels in intraperitoneal effusions of human ovarian cancer increasing cell survival, proliferation, and motility as well as stimulating production of neovascularizing factors. LPA2 and LPA3 and enzymes regulating the production and degradation of LPA are aberrantly expressed by ovarian cancer cells, but the consequences of these expression changes in ovarian cancer cells were unknown. METHODS: Expression of LPA1, LPA2, or LPA3 was inhibited or increased in ovarian cancer cells using small interfering RNAs (siRNAs) and lentivirus constructs, respectively. We measured the effects of changes in LPA receptor expression on cell proliferation (by crystal violet staining), cell motility and invasion (using Boyden chambers), and cytokines (interleukin 6 [IL-6], interleukin 8 [IL-8], and vascular endothelial growth factor [VEGF]) production by enzyme-linked immunosorbent assay. The role of LPA receptors in tumor growth, ascites formation, and cytokine production was assessed in a mouse xenograft model. All statistical tests were two-sided. RESULTS: SKOV-3 cells with increased expression of LPA receptors showed increased invasiveness, whereas siRNA knockdown inhibited both migration (P < .001, Student t test) and invasion. Knockdown of the LPA2 or LPA3 receptors inhibited the production of IL-6, IL-8, and VEGF in SKOV-3 and OVCAR-3 cells. SKOV-3 xenografts expressing LPA receptors formed primary tumors of increased size and increased ascites volume. Invasive tumors in the peritoneal cavity occurred in 75% (n = 4) of mice injected with LPA1 expressing SKOV-3 and 80% (n = 5) of mice injected with LPA2 or LPA3 expressing SKOV-3 cells. Metastatic tumors expressing LPA1, LPA2, and LPA3 were identified in the liver, kidney, and pancreas; tumors expressing LPA2 and LPA3 were detected in skeletal muscle; and tumors expressing LPA2 were also found in the cervical lymph node and heart. The percent survival of mice with tumors expressing LPA2 or LPA3 was reduced in comparison with animals with tumors expressing beta-galactosidase. CONCLUSIONS: Expression of LPA2 or LPA3 during ovarian carcinogenesis contributes to ovarian cancer aggressiveness, suggesting that the targeting of LPA production and action may have potential for the treatment of ovarian cancer.


Subject(s)
Gene Knockdown Techniques , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Lysophosphatidic Acid/metabolism , Animals , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Cell Survival , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Lentivirus , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
19.
Proc Natl Acad Sci U S A ; 105(13): 5248-53, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18367674

ABSTRACT

In primary mammalian cells, expression of oncogenes such as activated Ras induces premature senescence rather than transformation. We show that homozygous deletion of glycogen synthase kinase (GSK) 3beta (GSK3beta-/-) bypasses senescence induced by mutant Ras(V12) allowing primary mouse embryo fibroblasts (MEFs) as well as immortalized MEFs to exhibit a transformed phenotype in vitro and in vivo. Both catalytic activity and Axin-binding of GSK3beta are required to optimally suppress Ras transformation. The expression of Ras(V12) in GSK3beta-/-, but not in GSK3beta+/+ MEFs results in translocation of beta-catenin to the nucleus with concomitant up-regulation of cyclin D1. siRNA-mediated knockdown of beta-catenin decreases both cyclin D1 expression and anchorage-independent growth of transformed cells indicating a causal role for beta-catenin. Thus Ras(V12) and the lack of GSK3beta act in concert to activate the beta-catenin pathway, which may underlie the bypass of senescence and tumorigenic transformation by Ras.


Subject(s)
Cellular Senescence/physiology , Gene Deletion , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/metabolism , Homozygote , Transgenes/genetics , ras Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Catalysis , Cells, Cultured , Cyclin D1/metabolism , Fibroblasts , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Mice , Mice, Knockout , beta Catenin/metabolism , ras Proteins/genetics
20.
Clin Cancer Res ; 13(24): 7421-31, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18094426

ABSTRACT

PURPOSE: It is critical to develop methods to quantify the early pharmacodynamic effects of targeted therapeutics in vivo to make drug development more efficient and ensure biologically relevant dosing. Furthermore, an ability to identify patients likely to respond to targeted therapeutics would decrease the size, duration, and cost of clinical trials, resulting in more efficient translation to improved patient outcomes. Recent studies suggest that perifosine inhibits the phosphatidylinositol-3'-kinase (PI3K) pathway by preventing cell membrane recruitment of the AKT pleckstrin homology domain. EXPERIMENTAL DESIGN: A novel functional proteomics technology, reverse phase protein array, was used to establish and quantify pharmacodynamic markers of perifosine efficacy. RESULTS: Perifosine selectively prevents AKT recruitment to the membrane and blocks activation of downstream effectors. Perifosine inhibited breast, ovarian, and prostate cancer models. Growth inhibition was associated with apoptosis. Activation of AKT as a consequence of genomic aberrations predicted perifosine efficacy. In cell lines and xenografts, there was a highly statistically significant correlation between the degree of antitumor efficacy of different perifosine doses and quantified down-regulation of phosphorylation of AKT and of its downstream targets, particularly S6. CONCLUSIONS: Because of a strong correlation between proportional modulation of PI3K pathway biomarkers and quantified perifosine efficacy, it is likely that early measurement of such pharmacodynamic biomarkers with reverse phase protein array will optimize selection of responding patients and guide perifosine dosing. Furthermore, PI3K pathway activation status may allow baseline selection of patients most likely to respond to perifosine alone or in combination with other therapies.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Phosphorylcholine/analogs & derivatives , Protein Array Analysis/methods , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Humans , Immunoblotting , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/drug effects , Phosphorylation , Phosphorylcholine/pharmacokinetics , Proto-Oncogene Proteins c-akt/drug effects , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...