Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Daniel G. Rosen is active.

Publication


Featured researches published by Daniel G. Rosen.


Clinical Cancer Research | 2004

Selection of Potential Markers for Epithelial Ovarian Cancer with Gene Expression Arrays and Recursive Descent Partition Analysis

Karen H. Lu; Andrea P. Patterson; Lin Wang; Rebecca T. Marquez; Edward N. Atkinson; Keith A. Baggerly; Lance R. Ramoth; Daniel G. Rosen; Jinsong Liu; Ingegerd Hellström; David I. Smith; Lynn C. Hartmann; David A. Fishman; Andrew Berchuck; Rosemarie Schmandt; Regina S. Whitaker; David M. Gershenson; Gordon B. Mills; Robert C. Bast

Purpose: Advanced-stage epithelial ovarian cancer has a poor prognosis with long-term survival in less than 30% of patients. When the disease is detected in stage I, more than 90% of patients can be cured by conventional therapy. Screening for early-stage disease with individual serum tumor markers, such as CA125, is limited by the fact that no single marker is up-regulated and shed in adequate amounts by all ovarian cancers. Consequently, use of multiple markers in combination might detect a larger fraction of early-stage ovarian cancers. Experimental Design: To identify potential candidates for novel markers, we have used Affymetrix human genome arrays (U95 series) to analyze differences in gene expression of 41,441 known genes and expressed sequence tags between five pools of normal ovarian surface epithelial cells (OSE) and 42 epithelial ovarian cancers of different stages, grades, and histotypes. Recursive descent partition analysis (RDPA) was performed with 102 probe sets representing 86 genes that were up-regulated at least 3-fold in epithelial ovarian cancers when compared with normal OSE. In addition, a panel of 11 genes known to encode potential tumor markers [mucin 1, transmembrane (MUC1), mucin 16 (CA125), mesothelin, WAP four-disulfide core domain 2 (HE4), kallikrein 6, kallikrein 10, matrix metalloproteinase 2, prostasin, osteopontin, tetranectin, and inhibin] were similarly analyzed. Results: The 3-fold up-regulated genes were examined and four genes [Notch homologue 3 (NOTCH3), E2F transcription factor 3 (E2F3), GTPase activating protein (RACGAP1), and hematological and neurological expressed 1 (HN1)] distinguished all tumor samples from normal OSE. The 3-fold up-regulated genes were analyzed using RDPA, and the combination of elevated claudin 3 (CLDN3) and elevated vascular endothelial growth factor (VEGF) distinguished the cancers from normal OSE. The 11 known markers were analyzed using RDPA, and a combination of HE4, CA125, and MUC1 expression could distinguish tumor from normal specimens. Expression at the mRNA level in the candidate markers was examined via semiquantitative reverse transcription-PCR and was found to correlate well with the array data. Immunohistochemistry was performed to identify expression of the genes at the protein level in 158 ovarian cancers of different histotypes. A combination of CLDN3, CA125, and MUC1 stained 157 (99.4%) of 158 cancers, and all of the tumors were detected with a combination of CLDN3, CA125, MUC1, and VEGF. Conclusions: Our data are consistent with the possibility that a limited number of markers in combination might identify >99% of epithelial ovarian cancers despite the heterogeneity of the disease.


Proceedings of the National Academy of Sciences of the United States of America | 2006

The chemokine growth-regulated oncogene 1 (Gro-1) links RAS signaling to the senescence of stromal fibroblasts and ovarian tumorigenesis

Gong Yang; Daniel G. Rosen; Zhihong Zhang; Robert C. Bast; Gordon B. Mills; Justin A. Colacino; Imelda Mercado-Uribe; Jinsong Liu

Epithelial–stromal interactions play a critical role in tumor initiation and progression; cancer-associated stroma, but not normal stroma, is known to be tumor-promoting. However, the molecular signal used by epithelial cancer cells to reprogram normal stroma to a tumorigenic stroma is not known. Here, we present evidence to suggest that the chemokine growth-regulated oncogene 1 (Gro-1) may be one such signaling molecule. We showed that the expression of Gro-1 is activated by RAS and is vital for cell survival and the malignant transformation of ovarian epithelial cells. Surprisingly, we found that Gro-1 is a potent inducer of senescence in stromal fibroblasts and that this effect depends on functional p53. Senescent fibroblasts induced by Gro-1 can promote tumor growth whereas abrogation of senescence through immortalization results in loss of such tumor promoting activity. We also demonstrated that stromal fibroblasts adjacent to epithelial cancer cells are senescent in human ovarian cancer specimens and in heterografts from RAS-transformed human ovarian epithelial cells and ovarian cancer cells. Moreover, Gro-1 was expressed at significantly higher amounts in ovarian cancer than in normal tissues and was higher in serum samples from women with ovarian cancer than in serum from women without ovarian cancer. These findings provide strong evidence that RAS-induced Gro-1 can reprogram the stromal microenvironment through the induction of senescence of fibroblasts and thus can promote tumorigenesis. Therefore, Gro-1 may be a therapeutic target as well as a diagnostic marker in ovarian cancer.


Nature Medicine | 2005

Lineage infidelity of epithelial ovarian cancers is controlled by HOX genes that specify regional identity in the reproductive tract

Wenjun Cheng; Jinsong Liu; Hiroyuki Yoshida; Daniel G. Rosen; Honami Naora

Although epithelial ovarian cancers (EOCs) have been thought to arise from the simple epithelium lining the ovarian surface or inclusion cysts, the major subtypes of EOCs show morphologic features that resemble those of the müllerian duct–derived epithelia of the reproductive tract. We found that HOX genes, which normally regulate müllerian duct differentiation, are not expressed in normal ovarian surface epithelium (OSE), but are expressed in different EOC subtypes according to the pattern of müllerian-like differentiation of these cancers. Ectopic expression of Hoxa9 in tumorigenic mouse OSE cells gave rise to papillary tumors resembling serous EOCs. In contrast, Hoxa10 and Hoxa11 induced morphogenesis of endometrioid-like and mucinous-like EOCs, respectively. Hoxa7 showed no lineage specificity, but promoted the abilities of Hoxa9, Hoxa10 and Hoxa11 to induce differentiation along their respective pathways. Therefore, inappropriate activation of a molecular program that controls patterning of the reproductive tract could explain the morphologic heterogeneity of EOCs and their assumption of müllerian-like features.


Journal of Biological Chemistry | 2005

Mitochondrial manganese-superoxide dismutase expression in ovarian cancer: Role in cell proliferation and response to oxidative stress

Yumin Hu; Daniel G. Rosen; Yan Zhou; Li Feng; Gong Yang; Jinsong Liu; Peng Huang

Superoxide dismutases (SODs) are important antioxidant enzymes responsible for the elimination of superoxide radical (\batchmode \documentclass[fleqn,10pt,legalpaper]{article} \usepackage{amssymb} \usepackage{amsfonts} \usepackage{amsmath} \pagestyle{empty} \begin{document} \(\mathrm{O}_{2}^{-}\) \end{document}). The manganese-containing SOD (Mn-SOD) has been suggested to have tumor suppressor function and is located in the mitochondria where the majority of \batchmode \documentclass[fleqn,10pt,legalpaper]{article} \usepackage{amssymb} \usepackage{amsfonts} \usepackage{amsmath} \pagestyle{empty} \begin{document} \(\mathrm{O}_{2}^{-}\) \end{document} is generated during respiration. Although increased reactive oxygen species (ROS) in cancer cells has long been recognized, the expression of Mn-SOD in cancer and its role in cancer development remain elusive. The present study used a human tissue microarray to analyze Mn-SOD expression in primary ovarian cancer tissues, benign ovarian lesions, and normal ovary epithelium. Significantly higher levels of Mn-SOD protein expression were detected in the malignant tissues compared with normal tissues (p < 0.05). In experimental systems, suppression of Mn-SOD expression by small interfering RNA caused a 70% increase of superoxide in ovarian cancer cells, leading to stimulation of cell proliferation in vitro and more aggressive tumor growth in vivo. Furthermore, stimulation of mitochondrial \batchmode \documentclass[fleqn,10pt,legalpaper]{article} \usepackage{amssymb} \usepackage{amsfonts} \usepackage{amsmath} \pagestyle{empty} \begin{document} \(\mathrm{O}_{2}^{-}\) \end{document} production induced an increase of Mn-SOD expression. Our findings suggest that the increase in Mn-SOD expression in ovarian cancer is a cellular response to intrinsic ROS stress and that scavenging of superoxide by SOD may alleviate the ROS stress and thus reduce the simulating effect of ROS on cell growth.


Nature Chemical Biology | 2009

Identification of a chemical probe for NAADP by virtual screening

Edmund Naylor; Abdelilah Arredouani; Sridhar R. Vasudevan; Alexander M. Lewis; Raman Parkesh; Akiko Mizote; Daniel G. Rosen; Justyn M. Thomas; Minoru Izumi; A. Ganesan; A Galione; Grant C. Churchill

Research into the biological role of the Ca2+-releasing second messenger NAADP (nicotinic acid adenine dinucleotide phosphate) has been hampered by a lack of chemical probes. To find new chemical probes for exploring NAADP signaling, we turned to virtual screening, which can evaluate millions of molecules rapidly and inexpensively. We used NAADP as the query ligand to screen the chemical library ZINC for compounds with 3D-shape and electrostatic similarity. We tested the top-ranking hits in a sea urchin egg bioassay and found that one hit, Ned-19, blocks NAADP signaling at nanomolar concentrations. In intact cells, Ned-19 blocked NAADP signaling and fluorescently labeled NAADP receptors. Moreover, we show the utility of Ned-19 as a chemical probe by using it to demonstrate that NAADP is a key causal link between glucose sensing and Ca2+ increases in mouse pancreatic beta cells.


International Journal of Cancer | 2007

Expression of multiple human endogenous retrovirus surface envelope proteins in ovarian cancer

Feng Wang-Johanning; Jinsong Liu; Kiera Rycaj; Miao Huang; Kate Tsai; Daniel G. Rosen; Dung Tsa Chen; Danielle W. Lu; Kirstin F. Barnhart; Gary L. Johanning

Individual classes of human endogenous retrovirus (HERV) genes and proteins are expressed in cancer, but expression of more than one type of HERV is rare. We report here the expression of multiple HERV genes and proteins in ovarian cell lines and tissues. Expression of HERV‐K env mRNA was greater in ovarian epithelial tumors than in normal ovarian tissues (N = 254). The expression of this protein on the surface and in the cytoplasm of ovarian cancer cells was confirmed using anti‐HERV‐K specific antibody by flow cytometric analysis. The frequency of expression of HERV‐K env protein in multitissue microarrays (N = 641) was determined by immunohistochemistry and a significant correlation with tumor histotype was found. A significantly increased expression of HERV‐K was observed in tumors with low malignant potential and low grade, relative to expression in normal ovarian tissues. The increase in expression of HERV‐K env protein took place in a stepwise fashion in serous papillary adenocarcinoma. Interestingly, we found that other classes of HERV env mRNAs, including ERV3 and HERV‐E, are expressed in the same ovarian cancer tissues that expressed HERV‐K. Furthermore, anti‐HERV antibodies including anti‐ERV3 (30%), anti‐HERV‐E (40%) and anti‐HERV‐K (55%) were detected in patients with ovarian cancer, but not in normal female controls. HERV env proteins are frequently transcribed and translated in ovarian epithelial tumors, and multiple HERV families are detectable in ovarian cancer. HERV env proteins, and especially those expressed on the cell surface, may serve as novel tumor targets for detection, diagnosis and immunotherapy of ovarian cancer.


Modern Pathology | 2009

ALDH1 expression correlates with favorable prognosis in ovarian cancers

Bin Chang; Guangzhi Liu; Fengxia Xue; Daniel G. Rosen; Lianchun Xiao; Xuemei Wang; Jinsong Liu

Aldehyde dehydrogenase 1 (ALDH1), a detoxifying enzyme responsible for the oxidation of intracellular aldehydes, was shown to have a function in the early differentiation of stem cells, through its function in oxidizing retinol to retinoic acid. It has been shown that ALDH1 is a predictor of poor clinical outcome in breast cancer. The authors hypothesized that the level of ALDH1 expression may be correlated with the clinical outcome of patients with ovarian cancer. Immunohistochemical staining of ALDH1 expression was analyzed in 442 primary ovarian carcinomas using tissue microarray. The associations between the expression of the ALDH1 and clinical factors (diagnosis, tumor grade, stage, and clinical response to chemotherapy), as well as overall and disease-free survival, were analyzed. Expression of ALDH1 was found in 48.9% of the samples. Fishers exact test suggested that high expression of ALDH1 was significantly associated with endometrioid adenocarcinoma (P<0.0001), early-stage disease (P=0.006), complete response to chemotherapy (P<0.05), and a low serum level of CA125 (P=0.02). High percentage of cells expressing ALDH1 was associated with a longer overall survival time (P=0.01) and disease-free survival time (P=0.006) by log-rank test. In contrast to its function in breast cancer, ALDH1 was a favorable prognostic factor in ovarian carcinoma. ALDH1 therefore may have a different function in ovarian cancer than it does in breast cancer.


Modern Pathology | 2004

Validation of tissue microarray technology in ovarian carcinoma

Daniel G. Rosen; Xuelin Huang; Michael T. Deavers; Anais Malpica; Elvio G. Silva; Jinsong Liu

High-throughput tissue microarray allows many clinical specimens to be analyzed simultaneously on a single slide. One potential limitation of tissue microarray is the correct representation of each tumor with the small tissue core. Because tumors from different organs have different levels of heterogeneity, it requires a validation study for each one of them. We compared immunostaining of Ki-67, estrogen receptors, and p53 in whole sections of 45 cases of high-grade serous ovarian carcinoma with six core samples from those sections with regard to the number of tissue cores needed to reliably represent a whole section. Staining for Ki-67 was graded high or low by automated image analysis of 10 high-power fields; staining for estrogen receptor and p53 was scored on a 0-to-3 scale. Correlation coefficients for whole-section vs core stains were 0.86 for Ki-67, 0.93 for estrogen receptors, and 0.82 for p53. A total of 54 (6.6%) of the cores were inadequate for scoring. The probability that results from one core would correctly represent all three markers in the whole section was 91%; that for two cores was 96%; and that for three cores was 98%. Our results show that analysis of a single readable core matched the staining pattern of a whole section more than 90% of the time, and analysis of two cores increased that value to more than 95%, demonstrating that ovarian carcinoma tissue microarray is a reliable technique to analyze the expression of markers.


Cancer | 2006

The role of constitutively active signal transducer and activator of transcription 3 in ovarian tumorigenesis and prognosis

Daniel G. Rosen; Imelda Mercado-Uribe; Gong Yang; Robert C. Bast; Hesham M. Amin; Raymond Lai; Jinsong Liu

Signal transducer and activator of transcription 3 (Stat3), which is a latent transcription factor that participates in the transcriptional activation of apoptosis and cell cycle progression, has been implicated as an oncogene in several neoplastic diseases. However, the specific role of Stat3 in ovarian carcinogenesis remains poorly understood. The objectives of the current study were to examine the effect of Stat3 activation on the phenotypic transformation of an immortalized, nontumorigenic ovarian epithelial cell line and to evaluate the expression of tyrosine‐activated Stat3 (pStat3) in tissue microarrays from 303 ovarian carcinomas to determine its prognostic relevance and to correlate its expression with several upstream oncogenes of Stat3 and with the oncogenes involved in apoptosis and proliferation.


Clinical Cancer Research | 2010

CXCR2 Promotes Ovarian Cancer Growth through Dysregulated Cell Cycle, Diminished Apoptosis, and Enhanced Angiogenesis

Gong Yang; Daniel G. Rosen; Guangzhi Liu; Fan Yang; Xiaoqing Guo; Xue Xiao; Fengxia Xue; Imelda Mercado-Uribe; Jiaoti Huang; Sue-Hwa Lin; Gordon B. Mills; Jinsong Liu

Purpose: Chemokine receptor CXCR2 is associated with malignancy in several cancer models; however, the mechanisms involved in CXCR2-mediated tumor growth remain elusive. Here, we investigated the role of CXCR2 in human ovarian cancer. Experimental Design: CXCR2 expression was silenced by stable small hairpin RNA in ovarian cancer cell lines T29Gro-1, T29H, and SKOV3. Western blotting, immunofluorescence, enzyme-linked immunosorbent assay, flow cytometry, electrophoretic mobility shift assay, and mouse assay were used to detect CXCR2, interleukin-8, Gro-1, cell cycle, apoptosis, DNA binding of NF-κB, and tumor growth. Immunohistochemical staining of CXCR2 was done in 240 high-grade serous ovarian carcinoma samples. Results: Knockdown of CXCR2 expression by small hairpin RNA reduced tumorigenesis of ovarian cancer cells in nude mice. CXCR2 promoted cell cycle progression by modulating cell cycle regulatory proteins, including p21 (waf1/cip1), cyclin D1, CDK6, CDK4, cyclin A, and cyclin B1. CXCR2 inhibited cellular apoptosis by suppressing phosphorylated p53, Puma, and Bcl-xS; suppressing poly(ADP-ribose) polymerase cleavage; and activating Bcl-xL and Bcl-2. CXCR2 stimulated angiogenesis by increasing levels of vascular endothelial growth factor and decreasing levels of thrombospondin-1, a process likely involving mitogen-activated protein kinase, and NF-κB. Overexpression of CXCR2 in high-grade serous ovarian carcinomas was an independent prognostic factor of poor overall survival (P < 0.001) and of early relapse (P = 0.003) in the univariate analysis. Conclusions: Our data provide strong evidence that CXCR2 regulates the cell cycle, apoptosis, and angiogenesis through multiple signaling pathways, including mitogen-activated protein kinase and NF-κB, in ovarian cancer. CXCR2 thus has potential as a therapeutic target and for use in ovarian cancer diagnosis and prognosis. Clin Cancer Res; 16(15); 3875–86. ©2010 AACR.

Collaboration


Dive into the Daniel G. Rosen's collaboration.

Top Co-Authors

Avatar

Jinsong Liu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gong Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Robert C. Bast

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Imelda Mercado-Uribe

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Aysegul A. Sahin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gordon B. Mills

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Karen H. Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Constance Albarracin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zhihong Zhang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bin Chang

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge