Network


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

Hotspot


Dive into the research topics where Zhikun Ma is active.

Publication


Featured researches published by Zhikun Ma.


PLOS ONE | 2012

p53 inactivation upregulates p73 expression through E2F-1 mediated transcription.

Chaitali Tophkhane; Shihe Yang; Yunbo Jiang; Zhikun Ma; Dharmalingam Subramaniam; Shrikant Anant; Shingo Yogosawa; Toshiyuki Sakai; Wanguo Liu; Susan M. Edgerton; Ann D. Thor; Xiaohe Yang

While p73 overexpression has been associated with increased apoptosis in cancer tissues, p73 overexpressing tumors appear to be of high grade malignancy. Why this putative tumor suppressor is overexpressed in cancer cells and what the function of overexpressed p73 is in breast cancers are critical questions to be addressed. By investigating the effect of p53 inactivation on p73 expression, we found that both protein and mRNA levels of TAp73 were increased in MCF-7/p53siRNA cells, MCF-7/p53mt135 cells and HCT-116/p53−/− cells, as compared to wild type control, suggesting that p53 inactivation by various forms upregulates p73. We showed that p53 knockdown induced p73 was mainly regulated at the transcriptional level. However, although p53 has a putative binding site in the TAp73 promoter, deletion of this binding site did not affect p53 knockdown mediated activation of TAp73 promoter. Chromatin immuno-precipitation (ChIP) data demonstrated that loss of p53 results in enhanced occupancy of E2F-1 in the TAp73 promoter. The responsive sequence of p53 inactivation mediated p73 upregulation was mapped to the proximal promoter region of the TAp73 gene. To test the role of E2F-1 in p53 inactivation mediated regulation of p73 transcription, we found that p53 knockdown enhanced E2F-1 dependent p73 transcription, and mutations in E2F-1 binding sites in the TAp73 promoter abrogated p53 knockdown mediated activation of TAp73 promoter. Moreover, we demonstrated that p21 is a mediator of p53-E2F crosstalk in the regulation of p73 transcription. We concluded that p53 knockdown/inactivation may upregulate TAp73 expression through E2F-1 mediated transcriptional regulation. p53 inactivation mediated upregulation of p73 suggests an intrinsic rescuing mechanism in response to p53 mutation/inactivation. These findings support further analysis of the correlation between p53 status and p73 expression and its prognostic/predictive significance in human cancers.


Oncotarget | 2017

Phenformin inhibits growth and epithelial-mesenchymal transition of ErbB2-overexpressing breast cancer cells through targeting the IGF1R pathway

Zhiying Guo; Ming Zhao; Erin W. Howard; Qingxia Zhao; Amanda B. Parris; Zhikun Ma; Xiaohe Yang

Reports suggest that metformin, a popular anti-diabetes drug, prevents breast cancer through various systemic effects, including insulin-like growth factor receptor (IGFR) regulation. Although the anti-cancer properties of metformin have been well-studied, reports on a more bioavailable/potent biguanide, phenformin, remain sparse. Phenformin exerts similar functional activity to metformin and has been reported to impede mammary carcinogenesis in rats. Since the effects of phenformin on specific breast cancer subtypes have not been fully explored, we used ErbB2-overexpressing breast cancer cell and animal models to test the anti-cancer potential of phenformin. We report that phenformin (25-75 μM) decreased cell proliferation and impaired cell cycle progression in SKBR3 and 78617 breast cancer cells. Reduced tumor size after phenformin treatment (30 mg/kg/day) was demonstrated in an MMTV-ErbB2 transgenic mouse syngeneic tumor model. Phenformin also blocked epithelial-mesenchymal transition, decreased the invasive phenotype, and suppressed receptor tyrosine kinase signaling, including insulin receptor substrate 1 and IGF1R, in ErbB2-overexpressing breast cancer cells and mouse mammary tumor-derived tissues. Moreover, phenformin suppressed IGF1-stimulated proliferation, receptor tyrosine kinase signaling, and epithelial-mesenchymal transition markers in vitro. Together, our study implicates phenformin-mediated IGF1/IGF1R regulation as a potential anti-cancer mechanism and supports the development of phenformin and other biguanides as breast cancer therapeutics.Reports suggest that metformin, a popular anti-diabetes drug, prevents breast cancer through various systemic effects, including insulin-like growth factor receptor (IGFR) regulation. Although the anti-cancer properties of metformin have been well-studied, reports on a more bioavailable/potent biguanide, phenformin, remain sparse. Phenformin exerts similar functional activity to metformin and has been reported to impede mammary carcinogenesis in rats. Since the effects of phenformin on specific breast cancer subtypes have not been fully explored, we used ErbB2-overexpressing breast cancer cell and animal models to test the anti-cancer potential of phenformin. We report that phenformin (25–75 μM) decreased cell proliferation and impaired cell cycle progression in SKBR3 and 78617 breast cancer cells. Reduced tumor size after phenformin treatment (30 mg/kg/day) was demonstrated in an MMTV-ErbB2 transgenic mouse syngeneic tumor model. Phenformin also blocked epithelial-mesenchymal transition, decreased the invasive phenotype, and suppressed receptor tyrosine kinase signaling, including insulin receptor substrate 1 and IGF1R, in ErbB2-overexpressing breast cancer cells and mouse mammary tumor-derived tissues. Moreover, phenformin suppressed IGF1-stimulated proliferation, receptor tyrosine kinase signaling, and epithelial-mesenchymal transition markers in vitro. Together, our study implicates phenformin-mediated IGF1/IGF1R regulation as a potential anti-cancer mechanism and supports the development of phenformin and other biguanides as breast cancer therapeutics.


Oncotarget | 2017

Activation of cancerous inhibitor of PP2A (CIP2A) contributes to lapatinib resistance through induction of CIP2A-Akt feedback loop in ErbB2-positive breast cancer cells

Ming Zhao; Erin W. Howard; Amanda B. Parris; Zhiying Guo; Qingxia Zhao; Zhikun Ma; Ying Xing; Bolin Liu; Susan M. Edgerton; Ann D. Thor; Xiaohe Yang

Lapatinib, a small molecule ErbB2/EGFR inhibitor, is FDA-approved for the treatment of metastatic ErbB2-overexpressing breast cancer; however, lapatinib resistance is an emerging clinical challenge. Understanding the molecular mechanisms of lapatinib-mediated anti-cancer activities and identifying relevant resistance factors are of pivotal significance. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that is overexpressed in breast cancer. Our study investigated the role of CIP2A in the anti-cancer efficacy of lapatinib in ErbB2-overexpressing breast cancer cells. We found that lapatinib concurrently downregulated CIP2A and receptor tyrosine kinase signaling in ErbB2-overexpressing SKBR3 and 78617 cells; however, these effects were attenuated in lapatinib-resistant (LR) cells. CIP2A overexpression rendered SKBR3 and 78617 cells resistant to lapatinib-induced apoptosis and growth inhibition. Conversely, CIP2A knockdown via lentiviral shRNA enhanced cell sensitivity to lapatinib-induced growth inhibition and apoptosis. Results also suggested that lapatinib downregulated CIP2A through regulation of protein stability. We further demonstrated that lapatinib-induced CIP2A downregulation can be recapitulated by LY294002, suggesting that Akt mediates CIP2A upregulation. Importantly, lapatinib induced differential CIP2A downregulation between parental BT474 and BT474/LR cell lines. Moreover, CIP2A shRNA knockdown significantly sensitized the BT474/LR cells to lapatinib. Collectively, our results demonstrate that CIP2A is a molecular target and resistance factor of lapatinib with a critical role in lapatinib-induced cellular responses, including the inhibition of the CIP2A-Akt feedback loop. Further investigation of lapatinib-mediated CIP2A regulation will advance our understanding of lapatinib-associated anti-tumor activities and drug resistance.Lapatinib, a small molecule ErbB2/EGFR inhibitor, is FDA-approved for the treatment of metastatic ErbB2-overexpressing breast cancer; however, lapatinib resistance is an emerging clinical challenge. Understanding the molecular mechanisms of lapatinib-mediated anti-cancer activities and identifying relevant resistance factors are of pivotal significance. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that is overexpressed in breast cancer. Our study investigated the role of CIP2A in the anti-cancer efficacy of lapatinib in ErbB2-overexpressing breast cancer cells. We found that lapatinib concurrently downregulated CIP2A and receptor tyrosine kinase signaling in ErbB2-overexpressing SKBR3 and 78617 cells; however, these effects were attenuated in lapatinib-resistant (LR) cells. CIP2A overexpression rendered SKBR3 and 78617 cells resistant to lapatinib-induced apoptosis and growth inhibition. Conversely, CIP2A knockdown via lentiviral shRNA enhanced cell sensitivity to lapatinib-induced growth inhibition and apoptosis. Results also suggested that lapatinib downregulated CIP2A through regulation of protein stability. We further demonstrated that lapatinib-induced CIP2A downregulation can be recapitulated by LY294002, suggesting that Akt mediates CIP2A upregulation. Importantly, lapatinib induced differential CIP2A downregulation between parental BT474 and BT474/LR cell lines. Moreover, CIP2A shRNA knockdown significantly sensitized the BT474/LR cells to lapatinib. Collectively, our results demonstrate that CIP2A is a molecular target and resistance factor of lapatinib with a critical role in lapatinib-induced cellular responses, including the inhibition of the CIP2A-Akt feedback loop. Further investigation of lapatinib-mediated CIP2A regulation will advance our understanding of lapatinib-associated anti-tumor activities and drug resistance.


Scientific Reports | 2018

Ganetespib targets multiple levels of the receptor tyrosine kinase signaling cascade and preferentially inhibits ErbB2-overexpressing breast cancer cells

Harry Lee; Nipun Saini; Erin W. Howard; Amanda B. Parris; Zhikun Ma; Qingxia Zhao; Ming Zhao; Bolin Liu; Susan M. Edgerton; Ann D. Thor; Xiaohe Yang

Although ErbB2-targeted therapeutics have significantly improved ErbB2+ breast cancer patient outcomes, therapeutic resistance remains a significant challenge. Therefore, the development of novel ErbB2-targeting strategies is necessary. Importantly, ErbB2 is a sensitive client protein of heat shock protein 90 (HSP90), which regulates client protein folding, maturation, and stabilization. HSP90 inhibition provides an alternative therapeutic strategy for ErbB2-targeted degradation. In particular, ganetespib, a novel HSP90 inhibitor, is a promising agent for ErbB2+ cancers. Nevertheless, the anti-cancer efficacy and clinical application of ganetespib for ErbB2+ breast cancer is largely unknown. In our study, we examined the anti-cancer effects of ganetespib on ErbB2+ BT474 and SKBR3 breast cancer cells, and isogenic paired cancer cell lines with lentivirus-mediated ErbB2 overexpression. Ganetespib potently inhibited cell proliferation, cell cycle progression, survival, and activation/phosphorylation of ErbB2 and key downstream effectors in ErbB2+ breast cancer cells. Moreover, ganetespib decreased the total protein levels of HSP90 client proteins and reduced ErbB2 protein half-life. ErbB2-overexpressing cancer cells were also more sensitive to ganetespib-mediated growth inhibition than parental cells. Ganetespib also strikingly potentiated the inhibitory effects of lapatinib in BT474 and SKBR3 cells. Ultimately, our results support the application of ganetespib-mediated HSP90 inhibition as a promising therapeutic strategy for ErbB2+ breast cancer.


Carcinogenesis | 2018

Caloric restriction inhibits mammary tumorigenesis in MMTV-ErbB2 transgenic mice through the suppression of ER and ErbB2 pathways and inhibition of epithelial cell stemness in premalignant mammary tissues

Zhikun Ma; Amanda B. Parris; Erin W. Howard; Yujie Shi; Shihe Yang; Yunbo Jiang; Lingfei Kong; Xiaohe Yang

Caloric intake influences the onset of many diseases, including cancer. In particular, caloric restriction (CR) has been reported to suppress mammary tumorigenesis in various models. However, the underlying cancer preventive mechanisms have not been fully explored. To this end, we aimed to characterize the anticancer mechanisms of CR using MMTV-ErbB2 transgenic mice, a well-established spontaneous ErbB2-overexpressing mammary tumor model, by focusing on cellular and molecular changes in premalignant tissues. In MMTV-ErbB2 mice with 30% CR beginning at 8 weeks of age, mammary tumor development was dramatically inhibited, as exhibited by reduced tumor incidence and increased tumor latency. Morphogenic mammary gland analyses in 15- and 20-week-old mice indicated that CR significantly decreased mammary epithelial cell (MEC) density and proliferative index. To understand the underlying mechanisms, we analyzed the effects of CR on mammary stem/progenitor cells. Results from fluorescence-activated cell sorting analyses showed that CR modified mammary tissue hierarchy dynamics, as evidenced by decreased luminal cells (CD24highCD49flow), putative mammary reconstituting unit subpopulation (CD24highCD49fhigh) and luminal progenitor cells (CD61highCD49fhigh). Mammosphere and colony-forming cell assays demonstrated that CR significantly inhibited mammary stem cell self-renewal and progenitor cell numbers. Molecular analyses indicated that CR concurrently inhibited estrogen receptor (ER) and ErbB2 signaling. These molecular changes were accompanied by decreased mRNA levels of ER-targeted genes and epidermal growth factor receptor/ErbB2 family members and ligands, suggesting ER-ErbB2 signaling cross-talk. Collectively, our data demonstrate that CR significantly impacts ER and ErbB2 signaling, which induces profound changes in MEC reprogramming, and mammary stem/progenitor cell inhibition is a critical mechanism of CR-mediated breast cancer prevention.


Cancer Research | 2017

Abstract 1908: FGFR inhibitor, AZD4547, impedes the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice

Qingxia Zhao; Amanda B. Parris; Erin W. Howard; Ming Zhao; Ying Xing; Zhikun Ma; Xiaohe Yang

The fibroblast growth factor receptor (FGFR) family (FGFR1-4) of receptor tyrosine kinases (RTKs) regulates signaling pathways involved in cell proliferation and differentiation. In particular, FGFR1 and FGFR2, which are found in the terminal end buds of developing mammary ducts, play a role in mammary development and glandular morphogenesis involving the regulation of mammary stem cells (MaSCs) in mice. As such, a number of FGFR inhibitors are being tested in preclinical studies and clinical trials for anti-tumor properties. Nevertheless, reports on FGFR inhibitor-mediated breast cancer prevention are sparse. In this study, we aimed to investigate the anti-cancer benefits of AZD4547, a small molecule inhibitor of FGFR1-3, on ErbB2-overexpressing breast cancer models. We particularly focus on the effects of AZD4547 on MaSCs and tumor-initiating cells (TICs) in the premalignant tissues of MMTV-ErbB2 transgenic mice. We first demonstrated the anti-proliferative effects of AZD4547 (1-5 µM) on human ErbB2-overexpressing breast cancer cell lines. We further showed that AZD4547 confers potent inhibition of the stemness of these breast cancer cells, as indicated by significant depletion of ALDH+ cells and impaired tumorsphere formation. To study the in vivo effects of AZD4547 on the stemness of mammary epithelial cells (MECs), MMTV-ErbB2 transgenic mice were administered AZD4547 (2-6 mg/kg/day) for 10 weeks (weeks 8-18 of age) during the ‘risk window’ for mammary tumor development. Histopathological analysis indicated that AZD4547 significantly inhibited ductal branching and MEC proliferation. To examine the effect of AZD4547 on MEC subpopulations and tissue hierarchy dynamics in the premalignant mammary tissues of this model, we performed flow cytometry analyses on the primary MECs using CD24/CD49f and CD61/CD49f cell surface markers. The results showed that AZD4547 treatment substantially reduced MaSC-derived luminal and myoepithelial cell populations. AZD4547 also selectively suppressed the CD61highCD49fhigh cell population, which is enriched with luminal progenitor cells that give rise to TICs during MMTV-ErbB2 mammary tumor development. Mammosphere and colony-forming cell (CFC) assays on primary MECs demonstrated that the stemness of these cells was also blocked by AZD4547 prior to malignant transformation. Consistently, AZD4547 inhibited the anchorage-independent growth of cells from spontaneous tumors. Moreover, we demonstrated that AZD4547 downregulated multiple pathways, including the inactivation of FGFR, EGFR, and Wnt/β-catenin signaling. Collectively, the morphogenic, MaSC/TIC, and signaling regulation associated with AZD4547 treatment provides critical evidence for AZD4547 as a breast cancer preventative and therapeutic agent, which ultimately reveals clues for more effective eradication of refractory mammary tumors. Citation Format: Qingxia Zhao, Amanda B. Parris, Erin W. Howard, Ming Zhao, Ying Xing, Zhikun Ma, Xiaohe Yang. FGFR inhibitor, AZD4547, impedes the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1908. doi:10.1158/1538-7445.AM2017-1908


Cancer Research | 2013

Abstract 4790: Adult exposure to a soy-rich diet promotes mammary tumor development in MMTV-erbB-2 transgenic mice through induction of ER-erbB-2 crosstalk.

Xiaohe Yang; Zhikun Ma

The impact of soy and soy isoflavones, such as genistein, on breast cancer risk remains controversial. Although meta-analysis of multiple studies suggests that soy intake may be associated with a small reduction in breast cancer risk, increasing reports show that soy/genistein may be deleterious to breast cancer under certain conditions. Therefore, it is urgent to identify the factors that may affect the safety and risk of soy/genistein on breast cancer. We tackled this clinically relevant problem by examining the effect of adult exposure to soy/genistein on a subgroup of breast cancers that are positive for both estrogen receptor (ER) and erbB-2. erbB-2 is a receptor tyrosine kinase that is frequently overexpressed in breast cancer. ER+/erbB-2+ breast cancer accounts for ∼15-20% of breast cancer. Available data suggest that low dose soy/genistein stimulates ER+ breast cancer cells. On the other hand, genistein, a well known tyrosine kinase inhibitor, may inhibit erbB-2 overexpressing cells. The effect soy/genistein on ER+/erbB-2+ breast cancer is a dilemmatic question to be addressed. In this study MMTV-erbB-2 transgenic mice fed an AIN-93G diet were switched to a soy-rich diet (Purina 5001) at 20 weeks of age. Mammary tumor development in these mice was compared to that in control mice on a lifelong AIN-93G diet. We found that the adult switch to a soy diet accelerated mammary tumor development in this model. Average tumor latency for the control and soy-exposed groups were 36 and 32 weeks, respectively. Whole mount analysis indicated that the soy-diet-switch induced proliferation in the mammary glands as indicated by increased BrdU incorporation and epithelial density. We also found that changes in signaling of ER and erbB-2 pathways were modest at 24 weeks but more evident at 32 weeks. The soy-diet-switch induced ERα expression and phosphorylation which led to increased expression of ER target genes, including c-myc, cyclin D1, and Bcl-2. These changes were accompanied by increased phosphorylation/activation of EGFR, erbB-2, erbB-3, AKT1, and ERK½. Expression of EGFR and erbB-3 was also increased. Although the mammary tumors developed in this model were ER negative, mammary epithelial cells in the premalignant tissues were ER+/erbB-2+. These results provide in vivo evidence that the soy-rich diet stimulates the proliferation of ER+/erbB-2+ mammary epithelial cells, suggesting that soy/genistein-associated estrogenic activity overrides its tyrosine kinase inhibitor activity, possibly through ER-erbB-2 crosstalk. Our data suggests that ER+/erbB-2+ breast cancer might be particularly vulnerable to the risk associated with adult exposure to soy/genistein, and therefore patients with ER+/erbB-2+ tumors should be alerted. Citation Format: XiaoHe Yang, Zhikun Ma. Adult exposure to a soy-rich diet promotes mammary tumor development in MMTV-erbB-2 transgenic mice through induction of ER-erbB-2 crosstalk. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4790. doi:10.1158/1538-7445.AM2013-4790


Cancer Research | 2013

Abstract 4874: Bisphenol A exposure negates tamoxifen-mediated chemoprevention in MMTV-erbB2 transgenic mice.

Xiaohe Yang; Zhikun Ma; Yunbo Jiang

Bisphenol A (BPA), the building block of polycarbonate plastics with estrogenic activity, is one of the most common chemicals people are exposed to in daily life. Increasing reports suggest a link between BPA exposure and increased breast cancer risk. Although the effect of early BPA exposure and breast cancer risk later in life has been extensively studied, whether this xenoestrogen interferes with hormonal therapeutic agents, such as tamoxifen, has hardly been documented. In this study, we tested the effect of BPA exposure on mammary tumor development and tamoxifen-mediated chemoprevention in MMTV-erbB2 transgenic mice. BPA (500 ng/kg in sesame oil) was administered daily by subcutaneous injection for 8 weeks, starting from 8 weeks of age. Citrate tamoxifen at a dose of 1 mg/kg/day was administered by oral gavage alone or in combination with BPA. Based on tumor latency, we found that BPA alone accelerated mammary tumor development whereas tamoxifen treatment profoundly delayed the tumorigenesis. A combination of tamoxifen-treatment and BPA exposure, however, significantly (p Citation Format: XiaoHe Yang, Zhikun Ma, Yunbo Jiang. Bisphenol A exposure negates tamoxifen-mediated chemoprevention in MMTV-erbB2 transgenic mice. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4874. doi:10.1158/1538-7445.AM2013-4874


Cancer Research | 2012

Abstract 5452: In utero exposure to bisphenol A promotes mammary tumor development in MMTV-erbB-2 transgenic mice

Zhikun Ma; Xia Cao; Xiaohe Yang

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Increasing evidence indicates that environmental exposure plays a critical role in breast cancer etiology. Bisphenol A (BPA), the building block of polycarbonate plastics with estrogenic activity, is one of the most common chemicals exposed in daily life. Recent studies suggest a link between BPA exposure and increased breast cancer risk. However, specific conditions and the underlying mechanisms of BPA exposure associated breast cancer risk remain unclear. This study aimed to investigate the effects of in utero exposure to BPA on mammary tumor development in MMTV-erbB-2 transgenic mice, which exemplifies a scenario of gene-environmental interaction. Since erbB-2 is amplified/overexpressed in approximately 30% of breast cancer, which has been associated with poor prognosis and therapeutic resistance, use of this clinically relevant mammary tumor model is of high translational value. In this study, pregnant MMTV-erbB-2 mice were subcutaneously injected with 0, 25 ng, 250 ng and 250 µg/Kg body weight BPA daily between day 13 and day 19 of gestation. We found that in utero exposed mice in the low dose (25 and 250 ng) groups, but not the high dose group (250 µg), displayed earlier vaginal opening and prolonged estrous phase, suggesting a dose dependent pro-estrogenic effect of in utero exposure to BPA. Whole mount analysis showed that mammary glands of mice with in utero exposure to low dose BPA had a longer ductal extension at 6 weeks and increased lateral branching/alveolar structures at 10 weeks, which was more significant in the 250 ng group but less evident in the high dose group. Molecular analysis of mammary tissues at 10 weeks indicated that in utero exposure to BPA increased phosphorylation/activation of erbB-2, EGFR, erbB-3 Erk1/2 and Akt, and expression of erbB-3 and EGFR. Signaling of estrogen receptor (ER) pathway was upregulated concomitantly, as reflected by increased expression and phosphorylation of ERα and upregulation of cyclin D1 and c-myc. More importantly, mice in the 250 ng group but not the high dose group, developed tumor earlier than the mice in the control group, suggesting that dose is a critical factor affecting the outcomes. These data demonstrated that in utero exposure to BPA may promote mammary tumor development in erbB-2 transgenic mice in a dose dependent manner. Concomitant activation of the erbB-2 and ER pathways in the premalignant tissues suggests that induction of ER-erbB-2 crosstalk may play a critical role in mammary tumorigenesis promoted by in utero exposure to BPA, which underscores the significance of gene-environment interaction in early exposure to BPA associated breast cancer risk. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5452. doi:1538-7445.AM2012-5452


Cancer Research | 2012

Abstract 548: Erythropoietin promotes mammary tumor development in MMTV-erbB-2 transgenic mice

Zhikun Ma; Shihe Yang; Xia Cao; Xiaoxi Zhang; Wanming Zhao; Weiqun Ding; Zhizhuang Joe Zhao; Xiaohe Yang

Erythropoietin (EPO), a cytokine that stimulates erythropoiesis, is frequently used to treat anemia in cancer patients. Recent clinical studies indicate that EPO administration might be associated with tumor progression and reduced overall survival rate. However, the mechanisms of EPO associated risk in cancer patients remain poorly understood. In particular, a spontaneous tumor model that allows us to test the effects of EPO on tumor development/progression and provide mechanistic insights into multifactoral interactions in a physiological context has not been established. In this study, we used the MMTV-erbB-2 transgenic mouse model to test the specific effect of EPO on erbB-2 overexpressing breast cancers and aimed to establish a spontaneous tumor model for relevant studies. Female erbB-2 transgenic mice were injected with EPO? at 2000 U/Kg body weight, twice a week for three weeks (between week 20 and 22). We found that EPO treatment during the premalignant risk window not only increased hemoglobin levels but also significantly promoted mammary tumor development. In contrast to a 36-week mean latency of tumor development in control group, the mean latency for EPO exposed mice was 30 weeks. Moreover, EPO exposure resulted in increased pulmonary metastasis. Whole mount analysis of mammary gland at 23 weeks showed that EPO exposure induced ductal branching and alveolar outgrowth, which was consistent with increased BrdU incorporation in EPO exposed mammary tissues. Molecular analysis of mammary tissues at 23 weeks showed that the protein levels of p-Jak2, p-erbB-2, p-Erk and p-Akt1 were significantly increased, which was accompanied by increased expression of EGFR, erbB-3, IGFR, Bcl-2, cyclin D1 and VEGF. These data suggest that EPO exposure induced the activation of both EPOR/Jak2 and erbB-2/EGFR/erbB-3 pathways, and had a broad impact on signal transduction and gene expression in mammary tissues. Although the difference in signal transduction and gene expression of tumor tissues between control and EPO exposed groups was less significant, tumors from EPO treated mice showed increased microvessel density as compared to controls, suggesting EPO exposure induced a long lasting proangiogenic effect. Consistently, conditioned medium extracted from EPO treated mammary tissues at 23 weeks significantly increased tube formation of endothelial cells. Taken together, our results demonstrate that EPO treatment promotes the development and metastasis of erbB-2 overexpressing mammary tumors and established the MMTV-erbB-2 mouse model for the studies of EPO associated cancer risk. The underlying mechanisms involve enhanced activation of erbB-2 pathway through EPOR/Jak2 activation and increased proangiogenic activities in EPO exposed mammary tissues. Since erbB-2 is overexpressed in about 30% of human breast cancers, these data will have significant impact on safe use of EPO in breast cancer patients with anemia. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 548. doi:1538-7445.AM2012-548

Collaboration


Dive into the Zhikun Ma's collaboration.

Top Co-Authors

Avatar

Xiaohe Yang

University of Oklahoma

View shared research outputs
Top Co-Authors

Avatar

Amanda B. Parris

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar

Erin W. Howard

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar

Ming Zhao

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shihe Yang

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Yunbo Jiang

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Zhiying Guo

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ann D. Thor

Anschutz Medical Campus

View shared research outputs
Researchain Logo
Decentralizing Knowledge