Network


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

Hotspot


Dive into the research topics where Donghai Dai is active.

Publication


Featured researches published by Donghai Dai.


Obstetrics and Gynecology International | 2013

G Protein-Coupled Estrogen Receptor-Selective Ligands Modulate Endometrial Tumor Growth

Whitney K. Petrie; Megan K. Dennis; Chelin Hu; Donghai Dai; Jeffrey B. Arterburn; Harriet O. Smith; Helen J. Hathaway; Eric R. Prossnitz

Endometrial carcinoma is the most common cancer of the female reproductive tract. GPER/GPR30 is a 7-transmembrane spanning G protein-coupled receptor that has been identified as the third estrogen receptor, in addition to ERα and ERβ. High GPER expression is predictive of poor survival in endometrial and ovarian cancer, but despite this, the estrogen-mediated signaling pathways and specific estrogen receptors involved in endometrial cancer remain unclear. Here, employing ERα-negative Hec50 endometrial cancer cells, we demonstrate that GPER mediates estrogen-stimulated activation of ERK and PI3K via matrix metalloproteinase activation and subsequent transactivation of the EGFR and that ER-targeted therapeutic agents (4-hydroxytamoxifen, ICI182,780/fulvestrant, and Raloxifene), the phytoestrogen genistein, and the “ERα-selective” agonist propylpyrazole triol also function as GPER agonists. Furthermore, xenograft tumors of Hec50 cells yield enhanced growth with G-1 and estrogen, the latter being inhibited by GPER-selective pharmacologic antagonism with G36. These results have important implications with respect to the use of putatively ER-selective ligands and particularly for the widespread long-term use of “ER-targeted” therapeutics. Moreover, our findings shed light on the potential mechanisms of SERM/SERD side effects reported in many clinical studies. Finally, our results provide the first demonstration that pharmacological inhibition of GPER activity in vivo prevents estrogen-mediated tumor growth.


The Journal of Steroid Biochemistry and Molecular Biology | 2003

Progesterone regulation of activating protein-1 transcriptional activity: a possible mechanism of progesterone inhibition of endometrial cancer cell growth

Donghai Dai; Elizabeth S. Litman; Eric Schonteich; Kimberly K. Leslie

The uterine endometrium and cancers derived from it are classic models of hormone action: estrogen promotes growth and progesterone inhibits proliferation and results in differentiation. We have now identified a major pathway through which progesterone causes these growth-limiting effects. Ligand-bound progesterone receptors modulate the composition and transcriptional activity of members of the activating protein-1 (AP-1) family, and in particular, c-Jun. First, a dominant negative form of c-Jun inhibits the constitutive growth of Hec50co cells in a manner similar to the effects of progesterone through progesterone B receptors. Second, progesterone inhibits the transcriptional activity of the AP-1 complex in reporter gene assays. Third, the DNA binding of AP-1 and the composition of the individual AP-1 factors on DNA is regulated by progesterone on electrophoretic mobility shift assays. Fourth, progesterone strongly inhibits total AP-1 as well as c-Jun recruitment to the cyclin D1 promoter, but enhances AP-1 occupancy on the p53 and p21 promoters, as shown by chromatin immunoprecipitation assays. The effects of progesterone on AP-1 DNA binding are confirmed to result in altered transcription of these AP-1 target genes by RT-PCR. These studies establish that modulation of AP-1 activity is a potential pathway of progesterone-induced growth inhibition in endometrial cancer cells.


The Journal of Nuclear Medicine | 2008

Preclinical Development of a Neutral, Estrogen Receptor–Targeted, Tridentate 99mTc(I)-Estradiol-Pyridin-2-yl Hydrazine Derivative for Imaging of Breast and Endometrial Cancers

Tapan K. Nayak; Helen J. Hathaway; Chinnasamy Ramesh; Jeffrey B. Arterburn; Donghai Dai; Larry A. Sklar; Jeffrey P. Norenberg; Eric R. Prossnitz

Breast and endometrial cancers are the most common invasive malignancies in women, with more than 217,000 new diagnoses per year in the United States. These cancers are often classified into 2 subtypes based on the expression of the classical estrogen receptor. In this study, we describe a new structural class of neutral tridentate 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivatives for potential use in breast and endometrial cancer imaging. Methods: The 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative was synthesized via the Sonogashira cross-coupling reaction and radiolabeled via the tricarbonyl approach. Radiochemical purity was assessed by high-performance liquid chromatography. Cell-binding studies were performed with human breast adenocarcinoma MCF-7 cells. The in vivo biodistribution of the 99mTc(I) derivative was evaluated in virgin female C57BL/6 mice in defined phases of the estrous cycle. Biodistribution and SPECT/CT studies were performed with mice bearing MCF-7 and primary human endometrial tumors. Results: Radiochemical analysis demonstrated that the postpurification purity of the 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative was ≥95%, with a specific activity of 99mTc of 47.5 TBq/mmol. Cell-binding studies yielded a dissociation constant (mean ± SEM) of 11 ± 1.5 nM. In vivo studies revealed that receptor-mediated uptake was present in all phases of the estrous cycle in reproductive organs and mammary glands but was highest during the diestrous phase of the estrous cycle. Despite high nonspecific uptake in the liver, significant receptor-mediated uptake was observed in target tissues and estrogen receptor–expressing tumors (0.67% for MCF-7 tumors and 0.77% for endometrial tumors). Tumor uptake was reduced by approximately 50% on coinjection with 17β-estradiol. Conclusion: We have characterized a novel neutral tridentate 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative for potential use in breast and endometrial cancer imaging. This study represents the first step on a path toward the design of estrogen-based Tc-labeled tracers with improved targeting and SPECT imaging characteristics.


Biology of Reproduction | 2000

Regression of the Decidualized Mesometrium and Decidual Cell Apoptosis Are Associated with a Shift in Expression of Bcl2 Family Members

Donghai Dai; Bruce C. Moulton; Thomas F. Ogle

Abstract The purpose of this study was to determine whether regression of the decidua basalis (DB), which begins on Day 14 of pregnancy in the rat, results from an intrinsic program of apoptosis regulated by Bax and Bcl2. Expression of Bax and Bcl2 and the incidence of apoptosis were evaluated throughout gestation by Western blot analysis and detection of DNA fragments. Antiprogestin (RU486) was also administered during proliferation of DB to study progesterone regulation of Bax/Bcl2 balance. Bax, the pro-apoptotic protein, was expressed at a low level throughout pregnancy, whereas Bcl2, the pro-survival partner, was most abundantly expressed on Days 8 and 10, which are a time of proliferation and decidualization, and declined to barely detectable levels thereafter. These changes resulted in a 12-fold increase in the Bax:Bcl2 ratio on Day 17 as compared with Day 8 of pregnancy (P < 0.05). DNA laddering and in situ staining of DNA fragments first became visible on Day 14 and involved 2% of cells by Days 17 and 21 (P < 0.05). Treatment with RU486 on Day 9 enhanced Bax and suppressed Bcl2 within 6 h, increasing the Bax:Bcl2 ratio sixfold (P < 0.05). Apoptosis was minimal at 6 h and increased to 9% of cells by 24 h (P < 0.05). Thus, progesterone appears to regulate the apoptotic threshold of stromal cells by modulating Bax and Bcl2 expression.


Developmental Dynamics | 1999

VENTRALLY EMIGRATING NEURAL TUBE CELLS CONTRIBUTE TO THE FORMATION OF MECKEL'S AND QUADRATE CARTILAGE

G.S. Sohal; M.M. Ali; A.A. Ali; Donghai Dai

A population of multipotential neuroepithelial cells originating in the ventral portion of the hindbrain neural tube has been shown recently to emigrate at the site of attachment of the trigeminal nerve. These ventrally emigrating neural tube cells populate the mesenchyme of the first pharyngeal (branchial) arch. Because the Meckels and the quadrate cartilage develop from this mesenchyme, we sought to determine whether these ventrally emigrating neural tube cells contributed to their development. The ventral neural tube cells were tagged with a replication‐deficient retroviral vector containing the LacZ gene. This method permanently labels the descendents of the neural tube cells; thus, they can be subsequently tracked during development. The viral concentrate was microinjected into the lumen of the rostral hindbrain of chick embryos, after the emigration of neural crest is finished, on embryonic day 2 (stage 14). In control embryos, the virus was placed on top of the neural tube. Embryos were killed on days 3, 4, and 7 and processed for the detection of LacZ‐positive cells. By day 7, the Meckels and the quadrate cartilage can be easily recognized. LacZ‐positive cells were seen in both cartilages. They were located in perichondrium and in the cartilage. Immunostaining with the neural crest cell marker HNK‐1 indicated that the LacZ‐positive cells were HNK‐1 negative. The HNK‐1‐positive neural crest–derived cells were located in the cartilage but not in the perichondrium. These results indicate that the chondrocytes in the Meckels and the quadrate cartilage differentiate from two sources of cells; the ventrally emigrating neural tube cells and the neural crest. The developmental significance of differentiation of cartilage from the ventral neural tube cells and of the heterogeneous origin of chondrocytes in morphogenesis remains to be established. Dev Dyn 1999;216:37–44.


Cancer Research | 2005

A potential synergistic anticancer effect of paclitaxel and amifostine on endometrial cancer

Donghai Dai; Tan Nguyen; Suzy Davies; Daniel P. Theele; Claire F. Verschraegen; Kimberly K. Leslie

Although paclitaxel is one of the most effective chemotherapeutic agents, its usefulness is still limited in advanced and recurrent endometrial cancer. Amifostine protection of normal tissues against the side effects of chemotherapeutic agents has been clinically proven in cancer patients; however, its application in endometrial cancer has not been fully evaluated. We have investigated the use of paclitaxel and amifostine in controlling the growth of poorly differentiated endometrial cancer cells, Hec50co, in vitro and in vivo. Our studies show that amifostine had direct anticancer effects on endometrial cancer cells in vitro by arresting the cell cycle at the G1 phase and inducing apoptosis. Amifostine also inhibited s.c. tumor growth in athymic mice. Paclitaxel IC50 value was reduced from 14 to 2 nmol/L with pretreatment of a single dose of 178 micromol/L of amifostine for 72 hours. Amifostine also synergized with paclitaxel in the arrest of the cell cycle at the G2-M phase and in the induction of apoptosis. This two-drug regimen inhibited s.c. tumor growth as well as improved mouse survival significantly more than paclitaxel alone. Amifostine also significantly improved paclitaxel-induced cytotoxic effects on peripheral blood profiles. Our studies show that amifostine has direct anticancer effects on endometrial cancer. Our data have also shown a potential anticancer synergy between amifostine and paclitaxel in vitro and in vivo, whereas amifostine maintained a protective role in peripheral blood profiles. The dual specificity of amifostine action should be further investigated.


Journal of The Society for Gynecologic Investigation | 2004

Immunomodulatory and Transcriptional Effects of Progesterone Through Progesterone A and B Receptors in Hec50co Poorly Differentiated Endometrial Cancer Cells

Suzy Davies; Donghai Dai; Kimberly K. Leslie

Objective: Derivatives of progesterone, progestins, are used to treat endometrial cancer; however, the pathways activated by the hormone have not been fully investigated. Progesterone acts through two receptor isoforms, progesterone receptors A and B (PRA and PRB), transcription factors that control the expression of downstream genes leading to endometrial differentiation. The purpose of this study was to perform an expression analysis to identify the mechanisms underlying progesterones growth suppressive and immunomodulatory effects in endometrial cancer. Methods: To study the molecular effects of progesterone, PRs were introduced into Hec50co cells. Expression array analyses followed by confirmatory semiquantitative reverse-transcriptase polymerase chain reaction (RT-PCR) experiments were performed. Results: Expression analysis demonstrated a significant effect of progesterone after 12 hours of treatment on a number of genes, including cell signaling, DNA remodeling, apoptotic, tumor-suppressor, and transcription factors. Of particular interest was the consistent modulation of cytokines, which generally predicted for a powerful anti-inflammatory effect of progesterone through PR. Specifically, pro-inflammatory genes such as TNFα, IL-1β, and MCP-1/MCAF-1 were down-regulated and anti-inflammatory genes such as TRAP1 and SMAD4 were induced. Conclusion: We have discovered that progesterone has a modulatory effect on inflammation and many other important cellular functions. These effects likely underlie the inhibitory effects of progesterone on tumor growth and invasion.


Oncology Reports | 2011

Effects of bevacizumab in mouse model of endometrial cancer: Defining the molecular basis for resistance

Suzy Davies; Donghai Dai; Gavin Pickett; Kristina W. Thiel; Victoria P Korovkina; Kimberly K. Leslie

Endometrial cancer is the most frequent gynecologic cancer in women. Long-term outcomes for patients with advanced stage or recurrent disease are poor. Targeted molecular therapy against the vascular endothelial growth factor (VEGF) and its receptors constitute a new therapeutic option for these patients. The goal of our study was to assess the potential effectiveness of inhibition of VEGF/VEGFR signaling in a xenograft model of endometrial cancer using bevacizumab (Avastin, a humanized antibody against VEGFA). We also aimed to identify molecular markers of sensitivity or resistance to this agent. We show that bevacizumab retards tumor growth in athymic mice by inhibiting molecular components of signaling pathways that sustain cell survival and proliferation. We also demonstrate that resistance to bevacizumab may involve up-regulation of anti-apoptotic genes and certain proto-oncogenes. We propose that down-regulation of ARHGAP6 and MMP15 transcripts indicates that tumors are sensitive to bevacizumab whereas inhibition of PKCδ- or S6K-dependent signaling and up-regulation of TNFRS4 or MMP13 and MMP14 mark a developing resistance to bevacizumab therapy. Interestingly, the significant activation of c-Jun oncogene detected in bevacizumab-treated tumors suggests that, in endometrial cancers, the c-Jun-mediated pathway(s) contribute to bevacizumab resistance.


General Pharmacology-the Vascular System | 1999

Ventrally emigrating neural tube cells differentiate into vascular smooth muscle cells

A.A. Ali; M.M. Ali; Donghai Dai; G.S. Sohal

A multipotential cell population originating in the ventral part of the hindbrain neural tube, the ventrally emigrating neural tube cells (VENT cells), has recently been shown to migrate into the craniofacial mesenchyme. Because vascular smooth muscle cells develop from this mesenchyme, we sought to determine if the VENT cells contributed to their differentiation. VENT cells were tagged with replication-deficient retroviral vector with LacZ by microinjection into the lumen of the rostral hindbrain of chick embryos on day 2. Embryos were processed for the detection of LacZ positive cells on day 7. LacZ-positive cells were present in the wall of craniofacial arteries and veins. Immunostaining with the smooth muscle alpha-actin confirmed the labeled cells to be smooth muscle cells. It is concluded that some vascular smooth muscle cells differentiate from neural tube cells. the developmental and functional significance of which remains to be established.


International Journal of Oncology | 2015

TP53 oncomorphic mutations predict resistance to platinum- and taxane-based standard chemotherapy in patients diagnosed with advanced serous ovarian carcinoma

Pavla Brachova; Samuel R. Mueting; Matthew J. Carlson; Michael J. Goodheart; Anna Button; Sarah L. Mott; Donghai Dai; Kristina W. Thiel; Eric J. Devor; Kimberly K. Leslie

Individual mutations in the tumor suppressor TP53 alter p53 protein function. Some mutations create a non-functional protein, whereas others confer oncogenic activity, which we term ‘oncomorphic’. Since mutations in TP53 occur in nearly all ovarian tumors, the objective of this study was to determine the relationship of oncomorphic TP53 mutations with patient outcomes in advanced serous ovarian cancer patients. Clinical and molecular data from 264 high-grade serous ovarian cancer patients uniformly treated with standard platinum- and taxane-based adjuvant chemotherapy were downloaded from The Cancer Genome Atlas (TCGA) portal. Additionally, patient samples were obtained from the University of Iowa and individual mutations were analyzed in ovarian cancer cell lines. Mutations in the TP53 were annotated and categorized as oncomorphic, loss of function (LOF), or unclassified. Associations between mutation types, chemoresistance, recurrence, and progression-free survival (PFS) were calculated. Oncomorphic TP53 mutations were present in 21.3% of ovarian cancers in the TCGA dataset. Patients with oncomorphic TP53 mutations demonstrated significantly worse PFS, a 60% higher risk of recurrence (HR=1.60, 95% confidence intervals 1.09, 2.33, p=0.015), and higher rates of platinum resistance (χ2 test p=0.0024) when compared with single nucleotide mutations not categorized as oncomorphic. Furthermore, tumors containing oncomorphic TP53 mutations displayed unique protein expression profiles, and some mutations conferred increased clonogenic capacity in ovarian cancer cell models. Our study reveals that oncomorphic TP53 mutations are associated with worse patient outcome. These data suggest that future studies should take into consideration the functional consequences of TP53 mutations when determining treatment options.

Collaboration


Dive into the Donghai Dai's collaboration.

Top Co-Authors

Avatar

Kimberly K. Leslie

University of Iowa Hospitals and Clinics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael J. Goodheart

University of Iowa Hospitals and Clinics

View shared research outputs
Top Co-Authors

Avatar

Shujie Yang

University of Iowa Hospitals and Clinics

View shared research outputs
Top Co-Authors

Avatar

Suzy Davies

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas F. Ogle

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge