Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Anal Chem ; 88(16): 7899-903, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27436111

ABSTRACT

Currently used elution methods for strong cation exchange (SCX) chromatography are based on two principles: salt and pH gradient. In this paper, we report the first observation of peptide elution by acid gradient. The degree of peptide separation using C18-SCX StageTip was greatly improved by our acid and salt-based elution method compared with a salt-based elution method. This development enabled us to identify over 22 000 phosphopeptides from 2 mg of protein without labor-intensive sample preparation. Our method is simple, robust, scalable, and low-cost and can be easily implemented without any special equipment or techniques.


Subject(s)
Peptides/isolation & purification , Proteome , Cations/chemistry , Chromatography, Ion Exchange , HeLa Cells , Humans , Hydrogen-Ion Concentration , Peptides/chemistry , Salts/chemistry
2.
PLoS One ; 10(5): e0126483, 2015.
Article in English | MEDLINE | ID: mdl-25993439

ABSTRACT

Transforming growth factor-ß (TGF-ß) has a dual role in tumorigenesis, acting as either a tumor suppressor or as a pro-oncogenic factor in a context-dependent manner. Although TGF-ß antagonists have been proposed as anti-metastatic therapies for patients with advanced stage cancer, how TGF-ß mediates metastasis-promoting effects is poorly understood. Establishment of TGF-ß-related protein expression signatures at the metastatic site could provide new mechanistic information and potentially allow identification of novel biomarkers for clinical intervention to discriminate TGF-ß oncogenic effects from tumor suppressive effects. In the present study, we found that systemic administration of the TGF-ß receptor kinase inhibitor, SB-431542, significantly inhibited lung metastasis from transplanted 4T1 mammary tumors in Balb/c mice. The differentially expressed proteins in the comparison of lung metastases from SB-431542 treated and control vehicle-treated groups were analyzed by a quantitative LTQ Orbitrap Velos system coupled with stable isotope dimethyl labeling. A total of 36,239 peptides from 6,694 proteins were identified, out of which 4,531 proteins were characterized as differentially expressed. A subset of upregulated proteins in the control group was validated by western blotting and immunohistochemistry. The eukaryotic initiation factor (eIF) family members constituted the most enriched protein pathway in vehicle-treated compared with SB-43512-treated lung metastases, suggesting that increased protein expression of specific eIF family members, especially eIF4A1 and eEF2, is related to the metastatic phenotype of advanced breast cancer and can be down-regulated by TGF-ß pathway inhibitors. Thus our proteomic approach identified eIF pathway proteins as novel potential mediators of TGF-ß tumor-promoting activity.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Benzamides/pharmacology , Cell Line, Tumor , Dioxoles/pharmacology , Eukaryotic Initiation Factors/metabolism , Female , Humans , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Proteome/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors
3.
Cancer Res ; 74(21): 6139-49, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205100

ABSTRACT

Smad3, a major intracellular mediator of TGFß signaling, functions as both a positive and negative regulator in carcinogenesis. In response to TGFß, the TGFß receptor phosphorylates serine residues at the Smad3 C-tail. Cancer cells often contain high levels of the MAPK and CDK activities, which can lead to the Smad3 linker region becoming highly phosphorylated. Here, we report, for the first time, that mutation of the Smad3 linker phosphorylation sites markedly inhibited primary tumor growth, but significantly increased lung metastasis of breast cancer cell lines. In contrast, mutation of the Smad3 C-tail phosphorylation sites had the opposite effect. We show that mutation of the Smad3 linker phosphorylation sites greatly intensifies all TGFß-induced responses, including growth arrest, apoptosis, reduction in the size of putative cancer stem cell population, epithelial-mesenchymal transition, and invasive activity. Moreover, all TGFß responses were completely lost on mutation of the Smad3 C-tail phosphorylation sites. Our results demonstrate a critical role of the counterbalance between the Smad3 C-tail and linker phosphorylation in tumorigenesis and metastasis. Our findings have important implications for therapeutic intervention of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Smad3 Protein/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phosphorylation/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
4.
J Histochem Cytochem ; 62(12): 846-63, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25141865

ABSTRACT

Transforming growth factor-ß (TGF-ß) is an important regulator of cellular homeostasis and disease pathogenesis. Canonical TGF-ß signaling occurs through Smad2/3-Smad4 complexes; however, recent in vitro studies suggest that elevated levels of TGF-ß may activate a novel mixed Smad complex (Smad2/3-Smad1/5/9), which is required for some of the pro-oncogenic activities of TGF-ß. To determine if mixed Smad complexes are evident in vivo, we developed antibodies that can be used with a proximity ligation assay to detect either canonical or mixed Smad complexes in formalin-fixed paraffin-embedded sections. We demonstrate high expression of mixed Smad complexes in the tissues from mice genetically engineered to express high levels of TGF-ß1. Mixed Smad complexes were also prominent in 15-16 day gestation mouse embryos and in breast cancer xenografts, suggesting important roles in embryonic development and tumorigenesis. In contrast, mixed Smad complexes were expressed at extremely low levels in normal adult mouse tissue, where canonical complexes were correspondingly higher. We show that this methodology can be used in archival patient samples and tissue microarrays, and we have developed an algorithm to quantitate the brightfield read-out. These methods will allow quantitative analysis of cell type-specific Smad signaling pathways in physiological and pathological processes.


Subject(s)
Breast Neoplasms/metabolism , Mice/embryology , Signal Transduction , Smad Proteins/analysis , Smad Proteins/metabolism , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Antibodies/analysis , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Histocytological Preparation Techniques , Humans , Immunohistochemistry/methods , Mice, Transgenic , Molecular Sequence Data , Protein Interaction Mapping/methods , Sequence Alignment , Transforming Growth Factor beta/genetics , Up-Regulation
5.
Breast Cancer Res ; 16(3): R57, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24890385

ABSTRACT

INTRODUCTION: Transforming growth factor-ßs (TGF-ßs) play a dual role in breast cancer, with context-dependent tumor-suppressive or pro-oncogenic effects. TGF-ß antagonists are showing promise in early-phase clinical oncology trials to neutralize the pro-oncogenic effects. However, there is currently no way to determine whether the tumor-suppressive effects of TGF-ß are still active in human breast tumors at the time of surgery and treatment, a situation that could lead to adverse therapeutic responses. METHODS: Using a breast cancer progression model that exemplifies the dual role of TGF-ß, promoter-wide chromatin immunoprecipitation and transcriptomic approaches were applied to identify a core set of TGF-ß-regulated genes that specifically reflect only the tumor-suppressor arm of the pathway. The clinical significance of this signature and the underlying biology were investigated using bioinformatic analyses in clinical breast cancer datasets, and knockdown validation approaches in tumor xenografts. RESULTS: TGF-ß-driven tumor suppression was highly dependent on Smad3, and Smad3 target genes that were specifically enriched for involvement in tumor suppression were identified. Patterns of Smad3 binding reflected the preexisting active chromatin landscape, and target genes were frequently regulated in opposite directions in vitro and in vivo, highlighting the strong contextuality of TGF-ß action. An in vivo-weighted TGF-ß/Smad3 tumor-suppressor signature was associated with good outcome in estrogen receptor-positive breast cancer cohorts. TGF-ß/Smad3 effects on cell proliferation, differentiation and ephrin signaling contributed to the observed tumor suppression. CONCLUSIONS: Tumor-suppressive effects of TGF-ß persist in some breast cancer patients at the time of surgery and affect clinical outcome. Carefully tailored in vitro/in vivo genomic approaches can identify such patients for exclusion from treatment with TGF-ß antagonists.


Subject(s)
Breast Neoplasms/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Tumor Suppressor Proteins/genetics , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Ephrins/metabolism , Female , Humans , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering , Receptor, EphA2/metabolism , Smad2 Protein/genetics , Smad3 Protein/biosynthesis , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/biosynthesis , Tumor Suppressor Proteins/antagonists & inhibitors
6.
J Cell Sci ; 126(Pt 20): 4721-31, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23902688

ABSTRACT

Keratin filaments form cytoskeletal networks in epithelial cells. Dynamic rearrangement of keratin filament networks is required for epithelial cells to perform cellular processes such as cell migration and polarization; however, the mechanism governing keratin filament rearrangement remains unclear. Here, we describe a novel mechanism of keratin cytoskeleton organization mediated by casein kinase Iα (CK-1α) and a newly identified keratin-associated protein, FAM83H. Knockdown of FAM83H induces keratin filament bundling, whereas overexpression of FAM83H disassembles keratin filaments, suggesting that FAM83H regulates the filamentous state of keratins. Intriguingly, keratin filament bundling is concomitant with the dissociation of CK-1α from keratin filaments, whereas aberrant speckle-like localization of CK-1α is observed concomitantly with keratin filament disassembly. Furthermore, CK-1α inhibition, similar to FAM83H knockdown, causes keratin filament bundling and reverses keratin filament disassembly induced by FAM83H overexpression, suggesting that CK-1α mediates FAM83H-dependent reorganization of keratin filaments. Because the N-terminal region of FAM83H interacts with CK-1α and the C-terminal region interacts with keratins, FAM83H might tether CK-1α to keratins. Colorectal cancer tissue also shows keratin filament disassembly accompanied with FAM83H overexpression and aberrant CK-1α localization, and FAM83H-overexpressing cancer cells exhibit loss or alteration of epithelial cell polarity. Importantly, knockdown of FAM83H inhibits cell migration accompanied by keratin cytoskeleton rearrangement in colorectal cancer cells. These results suggest that keratin cytoskeleton organization is regulated by FAM83H-mediated recruitment of CK-1α to keratins, and that keratin filament disassembly caused by overexpression of FAM83H and aberrant localization of CK-1α could contribute to the progression of colorectal cancer.


Subject(s)
Casein Kinase Ialpha/metabolism , Colorectal Neoplasms/metabolism , Cytoskeleton/metabolism , Keratins/metabolism , Proteins/metabolism , Casein Kinase Ialpha/genetics , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
7.
PLoS One ; 8(3): e58859, 2013.
Article in English | MEDLINE | ID: mdl-23527039

ABSTRACT

Cancer cells alter their migratory properties during tumor progression to invade surrounding tissues and metastasize to distant sites. However, it remains unclear how migratory behaviors differ between tumor cells of different malignancy and whether these migratory behaviors can be utilized to assess the malignant potential of tumor cells. Here, we analyzed the migratory behaviors of cell lines representing different stages of breast cancer progression using conventional migration assays or time-lapse imaging and particle image velocimetry (PIV) to capture migration dynamics. We find that the number of migrating cells in transwell assays, and the distance and speed of migration in unconstrained 2D assays, show no correlation with malignant potential. However, the directionality of cell motion during 2D migration nicely distinguishes benign and tumorigenic cell lines, with tumorigenic cell lines harboring less directed, more random motion. Furthermore, the migratory behaviors of epithelial sheets observed under basal conditions and in response to stimulation with epidermal growth factor (EGF) or lysophosphatitic acid (LPA) are distinct for each cell line with regard to cell speed, directionality, and spatiotemporal motion patterns. Surprisingly, treatment with LPA promotes a more cohesive, directional sheet movement in lung colony forming MCF10CA1a cells compared to basal conditions or EGF stimulation, implying that the LPA signaling pathway may alter the invasive potential of MCF10CA1a cells. Together, our findings identify cell directionality as a promising indicator for assessing the tumorigenic potential of breast cancer cell lines and show that LPA induces more cohesive motility in a subset of metastatic breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cell Line, Tumor , Cell Migration Assays , Cell Movement/drug effects , Disease Progression , Epidermal Growth Factor/pharmacology , Female , Humans , Lysophospholipids/pharmacology , Neoplasm Metastasis , Phenotype , Tumor Stem Cell Assay
8.
J Lipid Res ; 53(12): 2698-707, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23034213

ABSTRACT

Transforming growth factor-ß (TGFß) is activated as a result of liver injury, such as cholestasis. However, its influence on endogenous metabolism is not known. This study demonstrated that TGFß regulates hepatic phospholipid and bile acid homeostasis through MAD homolog 3 (SMAD3) activation as revealed by lithocholic acid-induced experimental intrahepatic cholestasis. Lithocholic acid (LCA) induced expression of TGFB1 and the receptors TGFBR1 and TGFBR2 in the liver. In addition, immunohistochemistry revealed higher TGFß expression around the portal vein after LCA exposure and diminished SMAD3 phosphorylation in hepatocytes from Smad3-null mice. Serum metabolomics indicated increased bile acids and decreased lysophosphatidylcholine (LPC) after LCA exposure. Interestingly, in Smad3-null mice, the metabolic alteration was attenuated. LCA-induced lysophosphatidylcholine acyltransferase 4 (LPCAT4) and organic solute transporter ß (OSTß) expression were markedly decreased in Smad3-null mice, whereas TGFß induced LPCAT4 and OSTß expression in primary mouse hepatocytes. In addition, introduction of SMAD3 enhanced the TGFß-induced LPCAT4 and OSTß expression in the human hepatocellular carcinoma cell line HepG2. In conclusion, considering that Smad3-null mice showed attenuated serum ALP activity, a diagnostic indicator of cholangiocyte injury, these results strongly support the view that TGFß-SMAD3 signaling mediates an alteration in phospholipid and bile acid metabolism following hepatic inflammation with the biliary injury.


Subject(s)
Bile Acids and Salts/metabolism , Liver/metabolism , Phospholipids/metabolism , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Hep G2 Cells , Hepatocytes/chemistry , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lithocholic Acid , Liver/drug effects , Liver/injuries , Male , Mice , Mice, Inbred C57BL , Mice, Nude
9.
J Atheroscler Thromb ; 19(12): 1093-101, 2012.
Article in English | MEDLINE | ID: mdl-22878697

ABSTRACT

AIM: Ezetimibe selectively blocks intestinal cholesterol absorption by inhibiting Niemann-Pick C1-like 1 (NPC1L1) and reducing LDL cholesterol (LDL-C). In animals, ezetimibe reversed diet-induced obesity, liver steatosis, and insulin resistance. In humans, its potential effects on liver steatosis and insulin resistance have been suggested. We investigated the effects of ezetimibe on postprandial hyperlipidaemia and hyperglycaemia in obese subjects with dyslipidaemia in a double-blind randomized crossover trial. METHODS: Twenty obese men with hypertriglyceridaemia were assigned randomly to an ezetimibe- or a placebo-precedence-treated group. Subjects in the ezetimibe group were treated with ezetimibe (10 mg/day) for the first 4 weeks, followed by a 4-week interval and then treated with placebo for another 4 weeks. The placebo group received these treatments in reverse order. Subjects were requested to fast for at least 12 hours and then received a standard meal. Blood samples were collected at 0, 30, 60, 120, 240, 360 and 480 minutes after the meal on Days 0, 28, 56 and 84 and were used to measure the lipid and glucose metabolism markers. RESULTS: Ezetimibe significantly decreased the postprandial serum triglyceride excursion (p=0.01) and fasting serum LDL-C, remnant-like particles(RLP) and ApoB48 levels (p<0.05). Postprandial glucose excursion, serum insulin levels, serum glucose-dependent insulinotropic polypeptide (GIP) and active glucagon-like peptide-1 (GLP-1) were not significantly affected by ezetimibe treatment. CONCLUSION: Ezetimibe restored the postprandial dysregulation of lipid but did not affect glucose metabolism in a double-blind randomized crossover trial.


Subject(s)
Anticholesteremic Agents/therapeutic use , Azetidines/therapeutic use , Hyperglycemia/drug therapy , Hyperlipidemias/drug therapy , Adult , Apolipoprotein B-48/metabolism , Blood Glucose/metabolism , Cholesterol/metabolism , Cross-Over Studies , Double-Blind Method , Ezetimibe , Glucose/metabolism , Humans , Intestinal Mucosa/metabolism , Lipid Metabolism , Male , Membrane Proteins/metabolism , Membrane Transport Proteins , Middle Aged , Obesity/complications , Placebos , Postprandial Period , Prospective Studies , Time Factors , Triglycerides/metabolism
10.
J Proteome Res ; 11(8): 4201-10, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22716024

ABSTRACT

Since LC-MS-based quantitative proteomics has become increasingly applied to a wide range of biological applications over the past decade, numerous studies have performed relative and/or absolute abundance determinations across large sets of proteins. In this study, we discovered prognostic biomarker candidates from limited breast cancer tissue samples using discovery-through-verification strategy combining iTRAQ method followed by selected reaction monitoring/multiple reaction monitoring analysis (SRM/MRM). We identified and quantified 5122 proteins with high confidence in 18 patient tissue samples (pooled high-risk (n=9) or low-risk (n=9)). A total of 2480 proteins (48.4%) of them were annotated as membrane proteins, 16.1% were plasma membrane and 6.6% were extracellular space proteins by Gene Ontology analysis. Forty-nine proteins with >2-fold differences in two groups were chosen for further analysis and verified in 16 individual tissue samples (high-risk (n=9) or low-risk (n=7)) using SRM/MRM. Twenty-three proteins were differentially expressed among two groups of which MFAP4 and GP2 were further confirmed by Western blotting in 17 tissue samples (high-risk (n=9) or low-risk (n=8)) and Immunohistochemistry (IHC) in 24 tissue samples (high-risk (n=12) or low-risk (n=12)). These results indicate that the combination of iTRAQ and SRM/MRM proteomics will be a powerful tool for identification and verification of candidate protein biomarkers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Extracellular Matrix Proteins/metabolism , GPI-Linked Proteins/metabolism , Glycoproteins/metabolism , Amino Acid Sequence , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/isolation & purification , Breast Neoplasms/diagnosis , Carrier Proteins/chemistry , Carrier Proteins/isolation & purification , Chromatography, Ion Exchange , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/isolation & purification , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/isolation & purification , Glycoproteins/chemistry , Glycoproteins/isolation & purification , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Prognosis , Staining and Labeling , Statistics, Nonparametric , Tandem Mass Spectrometry
11.
Breast Cancer Res ; 12(5): R83, 2010.
Article in English | MEDLINE | ID: mdl-20942910

ABSTRACT

INTRODUCTION: Molecular dissection of the signaling pathways that underlie complex biological responses in the mammary epithelium is limited by the difficulty of propagating large numbers of mouse mammary epithelial cells, and by the inability of ribonucleic acid interference (RNAi)-based knockdown approaches to fully ablate gene function. Here we describe a method for the generation of conditionally immortalized mammary epithelial cells with defined genetic defects, and we show how such cells can be used to investigate complex signal transduction processes using the transforming growth factor beta (TGFß/Smad pathway as an example. METHODS: We intercrossed the previously described H-2Kb-tsA58 transgenic mouse (Immortomouse) which expresses a temperature-sensitive mutant of the simian virus-40 large T-antigen (tsTAg), with mice of differing Smad genotypes. A panel of conditionally immortalized mammary epithelial cell (IMEC) cultures were derived from the virgin mammary glands of offspring of these crosses and used to assess the Smad dependency of different biological responses to TGFß. RESULTS: IMECs could be propagated indefinitely at permissive temperatures and had a stable epithelial phenotype, resembling primary mammary epithelial cells with respect to several criteria, including responsiveness to TGFß. Using this panel of cells, we demonstrated that Smad3, but not Smad2, is necessary for TGFß-induced apoptotic, growth inhibitory and EMT responses, whereas either Smad can support TGFß-induced invasion as long as a threshold level of total Smad is exceeded. CONCLUSIONS: This work demonstrates the practicality and utility of generating conditionally immortalized mammary epithelial cell lines from genetically modified Immortomice for detailed investigation of complex signaling pathways in the mammary epithelium.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/genetics , Cell Differentiation , Cell Movement , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockout Techniques , Mammary Glands, Animal/cytology , Mice , Mice, Transgenic , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics
12.
PLoS One ; 5(2): e9201, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20169162

ABSTRACT

To gain insight into the role of genomic alterations in breast cancer progression, we conducted a comprehensive genetic characterization of a series of four cell lines derived from MCF10A. MCF10A is an immortalized mammary epithelial cell line (MEC); MCF10AT is a premalignant cell line generated from MCF10A by transformation with an activated HRAS gene; MCF10CA1h and MCF10CA1a, both derived from MCF10AT xenografts, form well-differentiated and poorly-differentiated malignant tumors in the xenograft models, respectively. We analyzed DNA copy number variation using the Affymetrix 500 K SNP arrays with the goal of identifying gene-specific amplification and deletion events. In addition to a previously noted deletion in the CDKN2A locus, our studies identified MYC amplification in all four cell lines. Additionally, we found intragenic deletions in several genes, including LRP1B in MCF10CA1h and MCF10CA1a, FHIT and CDH13 in MCF10CA1h, and RUNX1 in MCF10CA1a. We confirmed the deletion of RUNX1 in MCF10CA1a by DNA and RNA analyses, as well as the absence of the RUNX1 protein in that cell line. Furthermore, we found that RUNX1 expression was reduced in high-grade primary breast tumors compared to low/mid-grade tumors. Mutational analysis identified an activating PIK3CA mutation, H1047R, in MCF10CA1h and MCF10CA1a, which correlates with an increase of AKT1 phosphorylation at Ser473 and Thr308. Furthermore, we showed increased expression levels for genes located in the genomic regions with copy number gain. Thus, our genetic analyses have uncovered sequential molecular events that delineate breast tumor progression. These events include CDKN2A deletion and MYC amplification in immortalization, HRAS activation in transformation, PIK3CA activation in the formation of malignant tumors, and RUNX1 deletion associated with poorly-differentiated malignant tumors.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Chromosome Aberrations , Class I Phosphatidylinositol 3-Kinases , Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Gene Amplification , Gene Deletion , Gene Dosage , Genome-Wide Association Study , Humans , Mutation , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-myc/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Cancer Res ; 69(18): 7357-65, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19706770

ABSTRACT

To identify genetic events that characterize cancer progression, we conducted a comprehensive genetic evaluation of 161 primary breast tumors. Similar to the "mountain-and-hill" view of mutations, gene amplification also shows high- and low-frequency alterations in breast cancers. The frequently amplified genes include the well-known oncogenes ERBB2, FGFR1, MYC, CCND1, and PIK3CA, whereas other known oncogenes that are amplified, although less frequently, include CCND2, EGFR, FGFR2, and NOTCH3. More importantly, by honing in on minimally amplified regions containing three or fewer genes, we identified six new amplified genes: POLD3, IRAK4, IRX2, TBL1XR1, ASPH, and BRD4. We found that both the IRX2 and TBL1XR1 proteins showed higher expression in the malignant cell lines MCF10CA1h and MCF10CA1a than in their precursor, MCF10A, a normal immortalized mammary epithelial cell line. To study oncogenic roles of TBL1XR1, we performed knockdown experiments using a short hairpin RNA approach and found that depletion of TBL1XR1 in MCF10CA1h cells resulted in reduction of cell migration and invasion as well as suppression of tumorigenesis in mouse xenografts. Intriguingly, our mutation analysis showed the presence of activation mutations in the PIK3CA gene in a subset of tumors that also had DNA copy number increases in the PIK3CA locus, suggesting an additive effect of coexisting activating amino acid substitution and dosage increase from amplification. Our gene amplification and somatic mutation analysis of breast primary tumors provides a coherent picture of genetic events, both corroborating and novel, offering insight into the genetic underpinnings of breast cancer progression.


Subject(s)
Breast Neoplasms/genetics , Gene Amplification , Phosphatidylinositol 3-Kinases/genetics , Base Sequence , Breast Neoplasms/enzymology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , Enzyme Activation , Gene Dosage , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oncogenes , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Polymorphism, Single Nucleotide , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
14.
Cancer Res ; 68(18): 7278-82, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794114

ABSTRACT

We investigated the influence of acute wounding on tumor growth in a syngeneic mouse breast cancer model. Metastatic mouse breast cancer cells (4T1) were orthotopically injected into the mammary fat pads of BALB/c mice, and animals were wounded locally by full thickness dermal incisions above the mammary fat pads or remotely above the scapula 9 days later. Local, but not remote, wounding increased tumor size when compared with sham treatment. Injection of wound fluid close to the tumor site increased tumor growth, whereas in vitro wound fluid compared with serum increased the proliferation rate of 4T1 cells. Our results show that wound stroma can unfavorably influence growth of nearby tumors. This effect is T cell-dependent, as local wounding had no effect on tumor growth in nu/nu mice. The effect of wounding on tumor growth can be mimicked by acellular wound fluid, suggesting that T cells secrete or mediate secretion of cytokines or growth factors that then accelerate tumor growth. Here, we define an experimental model of wound-promoted tumor growth that will enable us to identify mechanisms and therapeutic targets to reduce the negative effect of tissue repair on residual tumors.


Subject(s)
Cell Transformation, Neoplastic/pathology , Mammary Glands, Animal/injuries , Mammary Neoplasms, Experimental/pathology , T-Lymphocytes/pathology , Wounds and Injuries/pathology , Animals , Cell Growth Processes/physiology , Disease Models, Animal , Female , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Wounds and Injuries/immunology
15.
Acta Trop ; 105(3): 269-73, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18243149

ABSTRACT

In the Philippines, insufficient consideration has been given to the implementation of systematic control measures against major abortifacient infectious agents in livestock. To elucidate the epidemiology of abortifacient infectious agents in livestock, the prevalence of four abortifacient agents was assessed. Initially, a total of 96 cattle including 17 cows with history of abortion were examined in a herd in Luzon at the request of the farm owner. Six (35.3%) of the 17 aborting cows were found to be serologically positive for Neospora caninum (N. caninum), whereas the seroprevalence in non-aborting cows was 15.9% (10/63). Four of the 6 serologically positive aborting cows were also RT-PCR-positive for bovine viral diarrhea virus (BVDV). Two (12.5%) of the 16 bulls examined were also found to be infected with BVDV, suggesting a putative risk factor of transmission via semen. Based on sequence analysis, the isolates detected belong to BVDV type 1b group. Furthermore, an epidemiological survey of abortifacient infectious agents was conducted with various species of livestock from herds located in Luzon. Out of the 105 water buffalo samples collected, 4 (3.8%) were indicated positive to N. caninum, 2 (1.9%) to Toxoplasma gondii (T. gondii) and 2 (1.9%) to Trypanosoma evansi (T. evansi). The overall seroprevalence of N. caninum in goat and sheep were 23.6% (21/89) and 26.3% (10/38), respectively. BVDV was not detected in these herds. The findings of this exploratory study indicate a relationship between infection and bovine abortion and that a lager study is required to statistically confirm this relationship.


Subject(s)
Abortion, Veterinary/epidemiology , Cattle Diseases/epidemiology , Diarrhea Viruses, Bovine Viral/isolation & purification , Eukaryota/isolation & purification , Pregnancy Complications, Infectious/veterinary , Pregnancy Complications, Parasitic/veterinary , Abortion, Veterinary/parasitology , Abortion, Veterinary/virology , Animals , Animals, Domestic , Antibodies, Protozoan/blood , Antibodies, Viral/blood , Cattle , Cattle Diseases/parasitology , Cattle Diseases/virology , Diarrhea Viruses, Bovine Viral/immunology , Eukaryota/immunology , Female , Neospora/isolation & purification , Philippines/epidemiology , Pregnancy , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Parasitic/epidemiology , Reverse Transcriptase Polymerase Chain Reaction , Seroepidemiologic Studies , Toxoplasma/isolation & purification , Trypanosoma/isolation & purification
16.
Dev Biol ; 312(2): 572-81, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17997399

ABSTRACT

Mutations in the TRPS1 gene lead to the tricho-rhino-phalangeal syndrome, which is characterized by skeletal defects and abnormal hair development. The TRPS1 gene encodes an atypical member of the GATA-type family of transcription factors. Here we show that mice with a disrupted Trps1 gene develop a chondrodysplasia characterized by diminished chondrocyte proliferation and decreased apoptosis in growth plates. Our analyses revealed that Trps1 is a repressor of Stat3 expression, which in turn controls chondrocyte proliferation and survival by regulating the expression of cyclin D1 and Bcl2. Our conclusion is supported (i) by siRNA-mediated depletion of Stat3 in Trps1-deficient chondrocytes, which normalized the expression of cyclin D1 and Bcl2, (ii) by overexpression of Trps1 in ATDC5 chondrocytes, which diminished Stat3 levels and increased proliferation and apoptosis, and (iii) by mutational analysis of the GATA-binding sites in the Stat3 gene, which revealed that their integrity is critical for the direct association with Trps1 and for Trps1-mediated repression of Stat3. Altogether our findings identify Trps1 as a novel regulator of chondrocytes proliferation and survival through the control of Stat3 expression.


Subject(s)
Chondrocytes/metabolism , GATA Transcription Factors/physiology , Repressor Proteins/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Blastocyst/metabolism , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Female , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/genetics
17.
Am J Pathol ; 166(5): 1393-403, 2005 May.
Article in English | MEDLINE | ID: mdl-15855640

ABSTRACT

We evaluated the therapeutic efficacy of topical administration of SN50, an inhibitor of nuclear factor-kappaB, in a corneal alkali burn model in mice. An alkali burn was produced with 1 N NaOH in the cornea of C57BL/6 mice under general anesthesia. SN50 (10 microg/microl) or vehicle was topically administered daily for up to 12 days. The eyes were processed for histological or immunohistochemical examination after bromodeoxyuridine labeling or for semi-quantification of cytokine mRNA. Topical SN50 suppressed nuclear factor-kappaB activation in local cells and reduced the incidence of epithelial defects/ulceration in healing corneas. Myofibroblast generation, macrophage invasion, activity of matrix metalloproteinases, basement membrane destruction, and expression of cytokines were all decreased in treated corneas compared with controls. To elucidate the role of tumor necrosis factor (TNF)-alpha in epithelial cell proliferation, we performed organ culture of mouse eyes with TNF-alpha, SN50, or an inhibitor of c-Jun N-terminal kinase (JNK) and examined cell proliferation in healing corneal epithelium in TNF-alpha-/- mice treated with SN50. An acceleration of epithelial cell proliferation by SN50 treatment was found to depend on TNF-alpha/JNK signaling. In conclusion, topical application of SN50 is effective in treating corneal alkali burns in mice.


Subject(s)
Alkalies , Burns, Chemical/drug therapy , Corneal Injuries , Eye Burns/drug therapy , NF-kappa B/antagonists & inhibitors , Peptides/administration & dosage , Administration, Topical , Animals , Basement Membrane/metabolism , Burns, Chemical/pathology , Burns, Chemical/physiopathology , Cornea/pathology , Cornea/physiopathology , Corneal Stroma/pathology , Cytokines/metabolism , Eye Burns/pathology , Eye Burns/physiopathology , Fibroblasts/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/pathology , Male , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Monocytes/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Peptides/therapeutic use , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Wound Healing
18.
Am J Pathol ; 166(5): 1405-18, 2005 May.
Article in English | MEDLINE | ID: mdl-15855641

ABSTRACT

Damage to the cornea from chemical burns is a serious clinical problem that often leads to permanent visual impairment. Because transforming growth factor (TGF)-beta has been implicated in the response to corneal injury, we evaluated the effects of altered TGF-beta signaling in a corneal alkali burn model using mice treated topically with an adenovirus (Ad) expressing inhibitory Smad7 and mice with a targeted deletion of the TGF-beta/activin signaling mediator Smad3. Expression of exogenous Smad7 in burned corneal tissue resulted in reduced activation of Smad signaling and nuclear factor-kappaB signaling via RelA/p65. Resurfacing of the burned cornea by conjunctival epithelium and its differentiation to cornea-like epithelium were both accelerated in Smad7-Ad-treated corneas with suppressed stromal ulceration, opacification, and neovascularization 20 days after injury. Introduction of the Smad7 gene suppressed invasion of monocytes/macrophages and expression of monocyte/macrophage chemotactic protein-1, TGF-beta1, TGF-beta2, vascular endothelial growth factor, matrix metalloproteinase-9, and tissue inhibitors of metalloproteinase-2 and abolished the generation of myofibroblasts. Although acceleration of healing of the burned cornea was also observed in mice lacking Smad3, the effects on epithelial and stromal healing were less pronounced than those in corneas treated with Smad7. Together these data suggest that overexpression of Smad7 may have effects beyond those of simply blocking Smad3/TGF-beta signaling and may represent an effective new strategy for treatment of ocular burns.


Subject(s)
Alkalies , Burns, Chemical/physiopathology , Corneal Injuries , DNA-Binding Proteins/pharmacology , Eye Burns/physiopathology , Eye/physiopathology , Trans-Activators/pharmacology , Wound Healing/drug effects , Adenoviridae/genetics , Animals , Burns, Chemical/metabolism , Burns, Chemical/pathology , Cell Division , Cornea/pathology , Cornea/physiopathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eye/metabolism , Eye Burns/metabolism , Eye Burns/pathology , Gene Transfer Techniques , Genetic Vectors , Growth Substances/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction/drug effects , Smad3 Protein , Smad7 Protein , Time Factors , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/metabolism
19.
Lab Invest ; 84(10): 1245-58, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15273699

ABSTRACT

Retinal pigment epithelial (RPE) cells dedifferentiate and undergo epithelial-mesenchymal transition (EMT) following retinal detachment, playing a central role in formation of fibrous tissue on the detached retina and vitreous retraction (proliferative vitreoretinopathy (PVR)). We have developed a mouse model of subretinal fibrosis with implications for PVR in which retinal detachment is induced without direct damage to the RPE cells. Transforming growth factor-beta (TGF-beta) has long been implicated both in EMT of RPEs and the development of PVR. Using mice null for Smad3, a key signaling intermediate downstream of TGF-beta and activin receptors, we show that Smad3 is essential for EMT of RPE cells induced by retinal detachment. De novo accumulation of fibrous tissue derived from multilayered RPE cells was seen following experimental retinal detachment in eyes of wild type, but not Smad3-null mice. Expression of alpha-smooth muscle actin, a hallmark of EMT in this cell type, and extracellular matrix components, lumican and collagen VI, were also not observed in eyes of Smad3-null mice. Our data show that induction of PDGF-BB by Smad3-dependent TGF-beta signaling is likely an important secondary proliferative component of the disease process. The results suggest that blocking the Smad3 pathway might be beneficial in prevention/treatment of PVR.


Subject(s)
DNA-Binding Proteins/physiology , Pigment Epithelium of Eye/pathology , Retinal Detachment/pathology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Vitreoretinopathy, Proliferative/pathology , Actins/metabolism , Animals , Becaplermin , Biomarkers/analysis , Cell Differentiation/physiology , Cell Line , Cell Movement/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Collagen Type VI/metabolism , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Humans , Keratan Sulfate/metabolism , Lumican , Mice , Mice, Inbred Strains , Mice, Knockout , Pigment Epithelium of Eye/metabolism , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Retinal Detachment/metabolism , Signal Transduction/physiology , Smad3 Protein , Swine , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta2 , Vitreoretinopathy, Proliferative/metabolism
20.
Lab Invest ; 84(10): 1259-70, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15258599

ABSTRACT

We examined the effect of adenovirus-mediated transient expression of Smad7, an inhibitory Smad in TGFbeta/activin signaling, on injury-induced epithelial-mesenchymal transition (EMT) of lens epithelium in mice. A volume of 3 microl of adenoviral solution was injected into the right lens of adult male C57BL/6 mice (n=56) at the time of capsular injury made using a hypodermic needle under general anesthesia. A mixture of recombinant adenovirus carrying CAG promoter-driven Cre (Cre adv) and mouse Smad7 complementary DNA (Smad7 adv) was administered to induce Smad7 expression, while control lenses were treated with Cre adv alone. After healing intervals of 2, 3, 5, and 10 days, animals were killed 2 h after labeling with bromodeoxyuridine (BrdU) and eyes were processed for histology. During healing, marked expression of Smad7 was observed in lens epithelial cells in the Smad7 adv group with loss of nuclear translocation of Smads2/3, while little Smad7 and abundant nuclear Smads2/3 were seen in cells in the Cre adv group. Lens epithelial cells in the Cre adv control group exhibited a fibroblastic appearance at days 5 and 10 and the capsular break was sealed with fibrous tissue, while Smad7 adv-treated cells around the capsular break retained their epithelial morphology and the break was not sealed. Expression of snail mRNA, and alpha-smooth muscle actin, lumican, and collagen VI proteins, markers of EMT, was observed in control-treated eyes, but not in cells of the Smad7 adv group at day 5 with minimal expression at day 10. Additionally, cell proliferation increased in epithelium infected with Smad7 adv consistent with suppression of injury-induced upregulation of TGFbeta1 in epithelium. We conclude that gene transfer of Smad7 in mice prevents injury-induced EMT of lens epithelial cells and sealing of the capsular break with fibrous tissue.


Subject(s)
Adenoviridae/genetics , DNA-Binding Proteins/genetics , Epithelial Cells/pathology , Eye Injuries/prevention & control , Lens Capsule, Crystalline/injuries , Lens, Crystalline/pathology , Trans-Activators/genetics , Transduction, Genetic , Actins/metabolism , Animals , Cell Division , Chondroitin Sulfate Proteoglycans/metabolism , Collagen Type VI/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Gene Expression Regulation , Genetic Therapy , Genetic Vectors/genetics , Keratan Sulfate/metabolism , Lens Capsule, Crystalline/metabolism , Lens Capsule, Crystalline/pathology , Lens, Crystalline/metabolism , Lumican , Male , Mesoderm/pathology , Mesoderm/physiology , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Smad7 Protein , Trans-Activators/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...