Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Methods Cell Biol ; 182: 265-284, 2024.
Article in English | MEDLINE | ID: mdl-38359982

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomerase-independent and recombination-based mechanism used by approximately 15% of human cancers to maintain telomere length and to sustain proliferation. ALT-positive cells display unique features that could be exploited for tailored cancer therapies. A key limitation for the development of ALT-specific treatments is the lack of an assay to detect ALT-positive cells that is easy to perform and that can be scaled up. One of the most broadly used assays for ALT detection, CCA (C-circle assay), does not provide single-cell information and it is not amenable to High-Throughput Screening (HTS). To overcome these limitations, we developed Native-FISH (N-FISH) as an alternative method to visualize ALT-specific single-stranded telomeric DNA. N-FISH produces single-cell data, can be applied to fixed tissues, does not require DNA isolation or amplification steps, and it can be miniaturized in a 384-well format. This protocol details the steps to perform N-FISH protocol both in a low- and high-throughput format to analyze ALT. While low-throughput N-FISH is useful to assay the ALT state of cell lines, we expect that the miniaturized N-FISH assay coupled with high-throughput imaging will be useful in functional genomics and chemical screens to identify novel cellular factors that regulate ALT and potential ALT therapeutic targets for cancer therapies directed against ALT-positive tumors, respectively.


Subject(s)
High-Throughput Screening Assays , Neoplasms , Humans , Animals , DNA , Telomere/genetics , Fishes/genetics
2.
J Cell Sci ; 136(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-37129573

ABSTRACT

Restricting the localization of the evolutionarily conserved centromeric histone H3 variant CENP-A to centromeres prevents chromosomal instability (CIN). The mislocalization of CENP-A to non-centromeric regions contributes to CIN in yeasts, flies and human cells. Even though overexpression and mislocalization of CENP-A have been reported in cancers, the mechanisms responsible for its mislocalization remain poorly understood. Here, we used an imaging-based high-throughput RNAi screen to identify factors that prevent mislocalization of overexpressed YFP-tagged CENP-A (YFP-CENP-A) in HeLa cells. Among the top five candidates in the screen - the depletion of which showed increased nuclear YFP-CENP-A fluorescence - were the histone chaperones CHAF1B (or p60) and CHAF1A (or p150). Follow-up validation and characterization experiments showed that CHAF1B-depleted cells exhibited CENP-A mislocalization, CIN phenotypes and increased enrichment of CENP-A in chromatin fractions. The depletion of DAXX, a histone H3.3 chaperone, suppressed CENP-A mislocalization and CIN in CHAF1B-depleted cells. We propose that in CHAF1B-depleted cells, DAXX promotes mislocalization of the overexpressed CENP-A to non-centromeric regions, resulting in CIN. In summary, we identified regulators of CENP-A localization and defined a role for CHAF1B in preventing DAXX-dependent CENP-A mislocalization and CIN.


Subject(s)
Chromosomal Proteins, Non-Histone , Histones , Humans , Histones/genetics , Centromere Protein A/genetics , HeLa Cells , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromatin , Centromere/metabolism , Molecular Chaperones/metabolism , Chromosomal Instability , Autoantigens/genetics , Chromatin Assembly Factor-1/genetics
3.
Sci Adv ; 8(13): eabj8360, 2022 04.
Article in English | MEDLINE | ID: mdl-35353576

ABSTRACT

The cohesin complex is central to chromatin looping, but mechanisms by which these long-range chromatin interactions are formed and persist remain unclear. We demonstrate that interactions between a transcription factor (TF) and the cohesin loader NIPBL regulate enhancer-dependent gene activity. Using mass spectrometry, genome mapping, and single-molecule tracking methods, we demonstrate that the glucocorticoid (GC) receptor (GR) interacts with NIPBL and the cohesin complex at the chromatin level, promoting loop extrusion and long-range gene regulation. Real-time single-molecule experiments show that loss of cohesin markedly diminishes the concentration of TF molecules at specific nuclear confinement sites, increasing TF local concentration and promoting gene regulation. Last, patient-derived acute myeloid leukemia cells harboring cohesin mutations exhibit a reduced response to GCs, suggesting that the GR-NIPBL-cohesin interaction is defective in these patients, resulting in poor response to GC treatment.


Subject(s)
Chromosomal Proteins, Non-Histone , Receptors, Glucocorticoid , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Humans , Receptors, Glucocorticoid/genetics , Cohesins
4.
Cytometry A ; 97(12): 1248-1264, 2020 12.
Article in English | MEDLINE | ID: mdl-33141508

ABSTRACT

Deep learning is rapidly becoming the technique of choice for automated segmentation of nuclei in biological image analysis workflows. In order to evaluate the feasibility of training nuclear segmentation models on small, custom annotated image datasets that have been augmented, we have designed a computational pipeline to systematically compare different nuclear segmentation model architectures and model training strategies. Using this approach, we demonstrate that transfer learning and tuning of training parameters, such as the composition, size, and preprocessing of the training image dataset, can lead to robust nuclear segmentation models, which match, and often exceed, the performance of existing, off-the-shelf deep learning models pretrained on large image datasets. We envision a practical scenario where deep learning nuclear segmentation models trained in this way can be shared across a laboratory, facility, or institution, and continuously improved by training them on progressively larger and varied image datasets. Our work provides computational tools and a practical framework for deep learning-based biological image segmentation using small annotated image datasets. Published [2020]. This article is a U.S. Government work and is in the public domain in the USA.


Subject(s)
Deep Learning , Cell Nucleus , Image Processing, Computer-Assisted
5.
Genome Res ; 29(10): 1719-1732, 2019 10.
Article in English | MEDLINE | ID: mdl-31515286

ABSTRACT

One of the hallmarks of cancer is chromosome instability (CIN), which leads to aneuploidy, translocations, and other chromosome aberrations. However, in the vast majority of human tumors the molecular basis of CIN remains unknown, partly because not all genes controlling chromosome transmission have yet been identified. To address this question, we developed an experimental high-throughput imaging (HTI) siRNA assay that allows the identification of novel CIN genes. Our method uses a human artificial chromosome (HAC) expressing the GFP transgene. When this assay was applied to screen an siRNA library of protein kinases, we identified PINK1, TRIO, IRAK1, PNCK, and TAOK1 as potential novel genes whose knockdown induces various mitotic abnormalities and results in chromosome loss. The HAC-based assay can be applied for screening different siRNA libraries (cell cycle regulation, DNA damage response, epigenetics, and transcription factors) to identify additional genes involved in CIN. Identification of the complete spectrum of CIN genes will reveal new insights into mechanisms of chromosome segregation and may expedite the development of novel therapeutic strategies to target the CIN phenotype in cancer cells.


Subject(s)
Chromosomal Instability/genetics , Chromosomes, Human/genetics , Protein Kinases/genetics , RNA, Small Interfering/genetics , Aneuploidy , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Cell Line, Tumor , Chromosomes, Artificial, Human/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Mitosis/genetics , Protein Kinases/isolation & purification , Protein Serine-Threonine Kinases/genetics , RNA, Double-Stranded/genetics , Transgenes , Translocation, Genetic/genetics
6.
Cancer Cell ; 31(1): 50-63, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28073004

ABSTRACT

Given the paucity of druggable mutations in high-risk neuroblastoma (NB), we undertook chromatin-focused small interfering RNA and chemical screens to uncover epigenetic regulators critical for the differentiation block in high-risk NB. High-content Opera imaging identified 53 genes whose loss of expression led to a decrease in NB cell proliferation and 16 also induced differentiation. From these, the secondary chemical screen identified SETD8, the H4K20me1 methyltransferase, as a druggable NB target. Functional studies revealed that SETD8 ablation rescued the pro-apoptotic and cell-cycle arrest functions of p53 by decreasing p53K382me1, leading to activation of the p53 canonical pathway. In pre-clinical xenograft NB models, genetic or pharmacological (UNC0379) SETD8 inhibition conferred a significant survival advantage, providing evidence for SETD8 as a therapeutic target in NB.


Subject(s)
Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Neuroblastoma/drug therapy , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/physiology , Cell Differentiation , Cell Proliferation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/physiology , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Quinazolines/pharmacology , Tumor Suppressor Protein p53/analysis
7.
Cancer Immunol Res ; 2(11): 1080-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25080445

ABSTRACT

Despite the strides that immunotherapy has made in mediating tumor regression, the clinical effects are often transient, and therefore more durable responses are still needed. The temporary nature of the therapy-induced immune response can be attributed to tumor immune evasion mechanisms, mainly the effect of suppressive immune cells and, in particular, regulatory T cells (Treg). Although the depletion of Tregs has been shown to be effective in enhancing immune responses, selective depletion of these suppressive cells without affecting other immune cells has not been very successful, and new agents are sought. We found that PI3K-Akt pathway inhibitors selectively inhibit Tregs with minimal effect on conventional T cells (Tconv). Our results clearly show selective in vitro inhibition of activation (as represented by a decrease in downstream signaling) and proliferation of Tregs in comparison with Tconvs when treated with different Akt and PI3K inhibitors. This effect has been observed in both human and murine CD4 T cells. In vivo treatment with these inhibitors resulted in a significant and selective reduction in Tregs in both naïve and tumor-bearing mice. Furthermore, these PI3K-Akt inhibitors led to a significant therapeutic antitumor effect, which was shown to be Treg dependent. Here, we report the use of PI3K-Akt pathway inhibitors as potent agents for the selective depletion of suppressive Tregs. We show that these inhibitors are able to enhance the antitumor immune response and are therefore promising clinical reagents for Treg depletion.


Subject(s)
Lymphocyte Activation/immunology , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
8.
Cancer Immunol Res ; 2(9): 911-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24872025

ABSTRACT

Studies have shown that Listeria monocytogenes (Lm)-based vaccine expressing a fusion protein comprising truncated listeriolysin O (LLO) and human papilloma virus (HPV) E7 protein (Lm-LLO-E7) induces a decrease in regulatory T cells (Treg) and complete regression of established, transplanted HPV-TC-1 tumors in mice. However, how the Lm-based vaccine causes a decrease in Tregs remains unclear. Using a highly attenuated Lm dal dat ΔactA strain (LmddA)-based vaccine, we report here that the vector LmddA was sufficient to induce a decrease in the proportion of Tregs by preferentially expanding CD4(+)FoxP3(-) T cells and CD8(+) T cells by a mechanism dependent on and directly mediated by LLO. Episomal expression of a nonhemolytic truncated LLO in Lm (LmddA-LLO) significantly augmented the expansion, thus further decreasing Treg frequency. Although adoptive transfer of Tregs compromised the antitumor efficacy of the LmddA-LLO-E7 vaccine, a combination of LmddA-LLO and an Lm-based vaccine expressing E7 protein (Lm-E7) induced complete regression against established TC-1 tumors. An engineered LLO-minus Lm expressing perfringolysin O (PFO) that enables the recombinant bacteria to exit from the phagolysosome without LLO confirmed that the adjuvant effect was dependent on LLO. These results suggest that LLO may serve as a promising adjuvant by preferentially inducing the expansions of CD4(+)FoxP3(-) T cells and CD8(+) T cells, thus reducing the ratio of Tregs to CD4(+)FoxP3(-) T cells and to CD8(+) T cells favoring immune responses to eradicate tumor.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Listeria monocytogenes/genetics , Papillomavirus E7 Proteins/genetics , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , Female , Genetic Vectors , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/therapy , Plasmids/genetics , T-Lymphocytes, Cytotoxic/immunology
9.
J Immunol ; 189(5): 2338-47, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22837483

ABSTRACT

Programmed death receptor 1 (PD-1) is an important signaling molecule often involved in tumor-mediated suppression of activated immune cells. Binding of this receptor to its ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), attenuates T cell activation, reduces IL-2 and IFN-γ secretion, decreases proliferation and cytotoxicity, and induces apoptosis. B7-DC-Ig is a recombinant protein that binds and targets PD-1. It is composed of an extracellular domain of murine B7-DC fused to the Fc portion of murine IgG2a. In this study, we demonstrate that B7-DC-Ig can enhance the therapeutic efficacy of vaccine when combined with cyclophosphamide. We show that this combination significantly enhances Ag-specific immune responses and leads to complete eradication of established tumors in 60% of mice and that this effect is CD8 dependent. We identified a novel mechanism by which B7-DC-Ig exerts its therapeutic effect that is distinctly different from direct blocking of the PD-L1-PD-1 interaction. In this study, we demonstrate that there are significant differences between levels and timing of surface PD-1 expression on different T cell subsets. We found that these differences play critical roles in anti-tumor immune effect exhibited by B7-DC-Ig through inhibiting proliferation of PD-1(high) CD4 T cells, leading to a significant decrease in the level of these cells, which are enriched for regulatory T cells, within the tumor. In addition, it also leads to a decrease in PD-1(high) CD8 T cells, tipping the balance toward nonexhausted functional PD-1(low) CD8 T cells. We believe that the PD-1 expression level on T cells is a crucial factor that needs to be considered when designing PD-1-targeting immune therapies.


Subject(s)
Cancer Vaccines/immunology , Programmed Cell Death 1 Receptor/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Escape/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Animals , B7-H1 Antigen , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Drug Delivery Systems/methods , Female , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
10.
PLoS One ; 7(1): e29079, 2012.
Article in English | MEDLINE | ID: mdl-22272227

ABSTRACT

Identification of gene expression profiles of cancer stem cells may have significant implications in the understanding of tumor biology and for the design of novel treatments targeted toward these cells. Here we report a potential ovarian cancer stem cell gene expression profile from isolated side population of fresh ascites obtained from women with high-grade advanced stage papillary serous ovarian adenocarcinoma. Affymetrix U133 Plus 2.0 microarrays were used to interrogate the differentially expressed genes between side population (SP) and main population (MP), and the results were analyzed by paired T-test using BRB-ArrayTools. We identified 138 up-regulated and 302 down-regulated genes that were differentially expressed between all 10 SP/MP pairs. Microarray data was validated using qRT-PCR and17/19 (89.5%) genes showed robust correlations between microarray and qRT-PCR expression data. The Pathway Studio analysis identified several genes involved in cell survival, differentiation, proliferation, and apoptosis which are unique to SP cells and a mechanism for the activation of Notch signaling is identified. To validate these findings, we have identified and isolated SP cells enriched for cancer stem cells from human ovarian cancer cell lines. The SP populations were having a higher colony forming efficiency in comparison to its MP counterpart and also capable of sustained expansion and differentiation in to SP and MP phenotypes. 50,000 SP cells produced tumor in nude mice whereas the same number of MP cells failed to give any tumor at 8 weeks after injection. The SP cells demonstrated a dose dependent sensitivity to specific γ-secretase inhibitors implicating the role of Notch signaling pathway in SP cell survival. Further the generated SP gene list was found to be enriched in recurrent ovarian cancer tumors.


Subject(s)
Carcinoma, Papillary/genetics , Cystadenocarcinoma, Serous/genetics , Gene Expression Profiling , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cystadenocarcinoma, Serous/pathology , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Neoplasm Staging , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/genetics , Tumor Cells, Cultured
11.
PLoS One ; 6(7): e21121, 2011.
Article in English | MEDLINE | ID: mdl-21754983

ABSTRACT

Clear cell ovarian cancer is an epithelial ovarian cancer histotype that is less responsive to chemotherapy and carries poorer prognosis than serous and endometrioid histotypes. Despite this, patients with these tumors are treated in a similar fashion as all other ovarian cancers. Previous genomic analysis has suggested that clear cell cancers represent a unique tumor subtype. Here we generated the first whole genomic expression profiling using epithelial component of clear cell ovarian cancers and normal ovarian surface specimens isolated by laser capture microdissection. All the arrays were analyzed using BRB ArrayTools and PathwayStudio software to identify the signaling pathways. Identified pathways validated using serous, clear cell cancer cell lines and RNAi technology. In vivo validations carried out using an orthotopic mouse model and liposomal encapsulated siRNA. Patient-derived clear cell and serous ovarian tumors were grafted under the renal capsule of NOD-SCID mice to evaluate the therapeutic potential of the identified pathway. We identified major activated pathways in clear cells involving in hypoxic cell growth, angiogenesis, and glucose metabolism not seen in other histotypes. Knockdown of key genes in these pathways sensitized clear cell ovarian cancer cell lines to hypoxia/glucose deprivation. In vivo experiments using patient derived tumors demonstrate that clear cell tumors are exquisitely sensitive to antiangiogenesis therapy (i.e. sunitinib) compared with serous tumors. We generated a histotype specific, gene signature associated with clear cell ovarian cancer which identifies important activated pathways critical for their clinicopathologic characteristics. These results provide a rational basis for a radically different treatment for ovarian clear cell patients.


Subject(s)
Microdissection , Molecular Targeted Therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Animals , Cell Death/drug effects , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Glucose/deficiency , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indoles/pharmacology , Mice , Mice, Nude , Microvessels/drug effects , Microvessels/pathology , Necrosis , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/genetics , Pyrroles/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sunitinib , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
12.
Cancer Cell ; 18(2): 185-97, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20708159

ABSTRACT

Although VEGF-targeted therapies are showing promise, new angiogenesis targets are needed to make additional gains. Here, we show that increased Zeste homolog 2 (EZH2) expression in either tumor cells or in tumor vasculature is predictive of poor clinical outcome. The increase in endothelial EZH2 is a direct result of VEGF stimulation by a paracrine circuit that promotes angiogenesis by methylating and silencing vasohibin1 (vash1). Ezh2 silencing in the tumor-associated endothelial cells inhibited angiogenesis mediated by reactivation of VASH1, and reduced ovarian cancer growth, which is further enhanced in combination with ezh2 silencing in tumor cells. Collectively, these data support the potential for targeting ezh2 as an important therapeutic approach.


Subject(s)
Histone-Lysine N-Methyltransferase/physiology , Neovascularization, Pathologic/physiopathology , Ovarian Neoplasms/blood supply , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , DNA Methylation , DNA Primers , Enhancer of Zeste Homolog 2 Protein , Female , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Fluorescence , Ovarian Neoplasms/pathology , Polycomb Repressive Complex 2
13.
Cancer Cell ; 16(6): 521-32, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19962670

ABSTRACT

Advanced stage papillary serous tumors of the ovary are responsible for the majority of ovarian cancer deaths, yet the molecular determinants modulating patient survival are poorly characterized. Here, we identify and validate a prognostic gene expression signature correlating with survival in a series of microdissected serous ovarian tumors. Independent evaluation confirmed the association of a prognostic gene microfibril-associated glycoprotein 2 (MAGP2) with poor prognosis, whereas in vitro mechanistic analyses demonstrated its ability to prolong tumor cell survival and stimulate endothelial cell motility and survival via the alpha(V)beta(3) integrin receptor. Increased MAGP2 expression correlated with microvessel density suggesting a proangiogenic role in vivo. Thus, MAGP2 may serve as a survival-associated target.


Subject(s)
Contractile Proteins/genetics , Glycoproteins/genetics , Ovarian Neoplasms/genetics , Cell Adhesion , Cell Movement , Female , Humans , Intercellular Signaling Peptides and Proteins , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , Recombinant Proteins/genetics , Signal Transduction , Survival Analysis
14.
Am J Pathol ; 175(2): 592-604, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19608871

ABSTRACT

Matrilysin-1 (also called matrix metalloproteinase-7) is expressed in injured lung and in cancer but not in normal epithelia. Bronchiolization of the alveoli (BOA), a potential precursor of lung cancer, is a histologically distinct type of metaplasia that is composed of cells resembling airway epithelium in the alveolar compartment. We demonstrate that there is increased expression of matrilysin-1 in human lesions and BOA in the CC10-human achaete-scute homolog-1 transgenic mouse model. Forced expression of the matrilysin-1 gene in immortalized human normal airway epithelial BEAS-2B and HPLD1 cells, which do not normally express matrilysin-1, promoted cellular migration, suggesting a functional link for BOA formation via bronchiolar cell migration. In addition, matrilysin-1 stimulated proliferation and inhibited Fas-induced apoptosis, while a knockdown by RNA interference decreased cell growth, migration, and increased sensitivity to apoptosis. Western blotting demonstrated increased levels of phospho-p38 and phospho-Erk1/2 kinases after matrilysin-1 expression. Gene expression analysis uncovered several genes that were related to cell growth, migration/movement, and death, which could potentially facilitate bronchiolization. In vivo, the formation of BOA lesions was reduced when CC10-human achaete-scute homolog-1 mice were crossed with matrilysin-1 null mice and was correlated with reduced matrilysin-1 expression in BOA. We conclude that matrilysin-1 may play an important role in the bronchiolization of alveoli by promoting proliferation, migration, and attenuation of apoptosis involving multiple genes in the MAP kinase pathway.


Subject(s)
Bronchioles/pathology , Lung Neoplasms/pathology , Matrix Metalloproteinase 7/physiology , Precancerous Conditions/pathology , Pulmonary Alveoli/pathology , Animals , Bronchioles/enzymology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Humans , Lung Neoplasms/enzymology , Matrix Metalloproteinase 7/genetics , Mice , Mice, Transgenic , Precancerous Conditions/enzymology , Pulmonary Alveoli/enzymology
15.
Mol Cancer Ther ; 7(10): 3123-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18852115

ABSTRACT

We recently used RNA interference to show that a negative correlation of L-asparaginase (L-ASP) chemotherapeutic activity with asparagine synthetase (ASNS) expression in the ovarian subset of the NCI-60 cell line panel is causal. To determine whether that relationship would be sustained in a larger, more diverse set of ovarian cell lines, we have now measured ASNS mRNA expression using microarrays and a branched-DNA RNA assay, ASNS protein expression using an electrochemiluminescent immunoassay, and L-ASP activity using an MTS assay on 19 human ovarian cancer cell lines. Contrary to our previous findings, L-ASP activity was only weakly correlated with ASNS mRNA expression; Pearson's correlation coefficients were r = -0.21 for microarray data and r = -0.39 for the branched-DNA RNA assay, with just the latter being marginally statistically significant (P = 0.047, one-tailed). ASNS protein expression measured by liquid-phase immunoassay exhibited a much stronger correlation (r = -0.65; P = 0.0014, one-tailed). We conclude that ASNS protein expression measured by immunoassay is a strong univariate predictor of L-ASP activity in ovarian cancer cell lines. These findings provide rationale for evaluation of ASNS protein expression as a predictive biomarker of clinical L-ASP activity in ovarian cancer.


Subject(s)
Asparaginase/metabolism , Aspartate-Ammonia Ligase/metabolism , Biomarkers, Tumor/metabolism , Ovarian Neoplasms/enzymology , Aspartate-Ammonia Ligase/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , DNA Fingerprinting , Female , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Cancer Res ; 68(13): 5478-86, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18593951

ABSTRACT

Despite the existence of morphologically indistinguishable disease, patients with advanced ovarian tumors display a broad range of survival end points. We hypothesize that gene expression profiling can identify a prognostic signature accounting for these distinct clinical outcomes. To resolve survival-associated loci, gene expression profiling was completed for an extensive set of 185 (90 optimal/95 suboptimal) primary ovarian tumors using the Affymetrix human U133A microarray. Cox regression analysis identified probe sets associated with survival in optimally and suboptimally debulked tumor sets at a P value of <0.01. Leave-one-out cross-validation was applied to each tumor cohort and confirmed by a permutation test. External validation was conducted by applying the gene signature to a publicly available array database of expression profiles of advanced stage suboptimally debulked tumors. The prognostic signature successfully classified the tumors according to survival for suboptimally (P = 0.0179) but not optimally debulked (P = 0.144) patients. The suboptimal gene signature was validated using the independent set of tumors (odds ratio, 8.75; P = 0.0146). To elucidate signaling events amenable to therapeutic intervention in suboptimally debulked patients, pathway analysis was completed for the top 57 survival-associated probe sets. For suboptimally debulked patients, confirmation of the predictive gene signature supports the existence of a clinically relevant predictor, as well as the possibility of novel therapeutic opportunities. Ultimately, the prognostic classifier defined for suboptimally debulked tumors may aid in the classification and enhancement of patient outcome for this high-risk population.


Subject(s)
Carcinoma, Papillary/diagnosis , Carcinoma, Papillary/genetics , Gene Expression Profiling , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Biomarkers, Tumor/genetics , Carcinoma, Papillary/mortality , Carcinoma, Papillary/surgery , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Neoplasm, Residual , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/mortality , Ovarian Neoplasms/surgery , Prognosis , Survival Analysis , Treatment Failure
18.
Cell Res ; 18(5): 538-48, 2008 May.
Article in English | MEDLINE | ID: mdl-18427574

ABSTRACT

Ovarian cancer is a major health problem for women in the United States. Despite evidence of considerable heterogeneity, most cases of ovarian cancer are treated in a similar fashion. The molecular basis for the clinicopathologic characteristics of these tumors remains poorly defined. Whole genome expression profiling is a genomic tool, which can identify dysregulated genes and uncover unique sub-classes of tumors. The application of this technology to ovarian cancer has provided a solid molecular basis for differences in histology and grade of ovarian tumors. Differentially expressed genes identified pathways implicated in cell proliferation, invasion, motility, chromosomal instability, and gene silencing and provided new insights into the origin and potential treatment of these cancers. The added knowledge provided by global gene expression profiling should allow for a more rational treatment of ovarian cancers. These techniques are leading to a paradigm shift from empirical treatment to an individually tailored approach. This review summarizes the new genomic data on epithelial ovarian cancers of different histology and grade and the impact it will have on our understanding and treatment of this disease.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Ovarian Neoplasms/genetics , Animals , Epithelium/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Ovarian Neoplasms/pathology
19.
Endocrinology ; 149(6): 2888-98, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18325988

ABSTRACT

Adrenomedullin (AM) and proadrenomedullin N-terminal 20 peptide (PAMP) are secretory hormones, but it is not unusual to find them in intracellular compartments. Using yeast-2 hybrid technology, we found interactions between AM and several microtubule-associated proteins (MAPs), and between PAMP and tubulin. Expression of fluorescent-tagged AM and PAMP as well as immunofluorescence for the native peptides showed a complete decoration of the microtubules and colocalization with other MAPs. PAMP, but not AM, bound to tubulin in vitro and destabilized tubulin polymerization. Down-regulation of the gene coding for both AM and PAMP through small interfering RNA technology resulted in morphological changes, microtubule stabilization, increase in posttranslational modifications of tubulin such as acetylation and detyrosination, reduction in cell motility, and partial arrest at the G2 phase of the cell cycle, when compared with cells transfected with the same vector carrying a scrambled sequence. These results show that PAMP is a novel MAP, whereas AM may be exerting more subtle effects in regulating cytoskeleton function.


Subject(s)
Adrenomedullin/physiology , Cytoskeleton/physiology , Microtubules/physiology , Peptide Fragments/physiology , Protein Precursors/physiology , Adenocarcinoma , Adrenomedullin/chemistry , Adrenomedullin/genetics , Animals , CHO Cells , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Cricetulus , Haplorhini , Humans , Lung Neoplasms , Protein Precursors/chemistry , Protein Precursors/genetics
20.
Dis Markers ; 23(5-6): 367-76, 2007.
Article in English | MEDLINE | ID: mdl-18057520

ABSTRACT

Ovarian cancer is complex disease composed of different histological grades and types. However, the underlying molecular mechanisms involved in the development of different phenotypes remain largely unknown. Epidemiological studies identified multiple exogenous and endogenous risk factors for ovarian cancer development. Among them, an inflammatory stromal microenvironment seems to play a critical role in the initiation of the disease. The interaction between such a microenvironment, genetic polymorphisms, and different epithelial components such as endosalpingiosis, endometriosis, and ovarian inclusion cyst in the ovarian cortex may induce different genetic changes identified in the epithelial component of different histological types of ovarian tumors. Genetic studies on different histological grades and types provide insight into the pathogenetic pathways for the development of different disease phenotypes. However, the link between all these genetic changes and the etiological factors remains to be established.


Subject(s)
Carcinoma/etiology , Carcinoma/pathology , Ovarian Neoplasms/etiology , Ovarian Neoplasms/pathology , Carcinoma/genetics , Female , Humans , Ovarian Neoplasms/genetics , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...