Network


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

Hotspot


Dive into the research topics where Karen Creswell is active.

Publication


Featured researches published by Karen Creswell.


Diabetes | 1992

Poly I:C Induces Development of Diabetes Mellitus in BB Rat

Douglas O. Sobel; Joseph T. Newsome; Cynthia H Ewel; Joseph A. Bellanti; Val Abbassi; Karen Creswell; Owen C. Blair

Polyinosinic polycytidilic acid (poly I:C), an inducer of α-interferon, accelerates the development of diabetes in diabetes-prone (DP) BioBreeding (BB) rats. This study investigates the effect of administering poly I:C to a diabetes-resistant (DR) strain of BB rats. We compared the incidence of diabetes, the degree of insulitis, the number of NK cells, helper-inducer cells, cytotoxic-suppressor cells, Ia+ T cells, RT6.1+ T cells, and NK cell bioactivity in DR rats treated with saline and with a 5 micrograms/g body wt (poly-5) dose and a 10 micrograms/g body wt (poly-10) dose of poly I:C. The incidence of diabetes was also compared with that of DP rats receiving poly-5. We found that both doses of poly I:C significantly induce the development of diabetes in the DR BB rat. However, treatment of DR rats with the higher dose induces a greater rate of development of diabetes and earlier onset of diabetes than the lower poly-5 dose. The rate of diabetes development and the mean age of onset were similar in poly-10–treated DR and poly-5–treated DP rats. A significant degree of insulitis occurred in all the poly I:C–treated DR rats, even those not developing diabetes. Peripheral blood NK cell number was greater in poly I:C than in saline-treated rats, after 2 wk of treatment and when killed. The percentage of OX19+ peripheral blood mononuclear cells expressing RT6.1 allotype or Ia antigen were similar in poly I:C– and saline-treated rats. of diabetes in poly-10–treated DR rats and poly-5–treated DP rats is consistent with a similar mechanism of poly I:C action in the DR and DP BB rats. Although the specific mechanism is not defined, NK cell numbers are elevated with poly I:C treatment. Alterations in RT6.1+ and la+ T cells do not appear to play a role.


Cancer Research | 2005

Tachyplesin Activates the Classic Complement Pathway to Kill Tumor Cells

Jinguo Chen; Xue-Ming Xu; Charles B. Underhill; Shanmin Yang; Luping Wang; Yixin Chen; Shuigen Hong; Karen Creswell; Lurong Zhang

Tachyplesin is a small, cationic peptide that possesses antitumor properties. However, little is known about its action mechanism. We used phage display to identify a protein that interacted with tachyplesin and isolated a sequence corresponding to the collagen-like domain of C1q, a key component in the complement pathway. Their interaction was subsequently confirmed by both ELISA and affinity precipitation. Tachyplesin seemed to activate the classic complement cascade because it triggered several downstream events, including the cleavage and deposition of C4 and C3 and the formation of C5b-9. When TSU tumor cells were treated with tachyplesin in the presence of serum, activated C4b and C3b could be detected on tumor cells by flow cytometry, Western blotting, and confocal microscopy. However, this effect was blocked when the tumor cells were treated with hyaluronidase or a large excess of hyaluronan, indicating that hyaluronan or related glycosaminoglycans were involved in this process. Treatment of cells with tachyplesin and serum increased in membrane permeability as indicated by the ability of FITC-dextran to enter the cytoplasm. Finally, the combination of tachyplesin and human serum markedly inhibited the proliferation and caused death of TSU cells, and these effects were attenuated if the serum was heat-inactivated or if hyaluronidase was added. Taken together, these observations suggest that tachyplesin binds to both hyaluronan on the cell surface and C1q in the serum and activates the classic complement cascade, which damages the integrity of the membranes of the tumor cells resulting in their death.


Cancer Research | 2013

C-Src modulates estrogen-induced stress and apoptosis in estrogen-deprived breast cancer cells

Ping Fan; Obi L. Griffith; Fadeke A. Agboke; Pavana Anur; Xiaojun Zou; Russell E. McDaniel; Karen Creswell; Sung Hoon Kim; John A. Katzenellenbogen; Joe W. Gray; V. Craig Jordan

The emergence of anti-estrogen resistance in breast cancer is an important clinical phenomenon affecting long-term survival in this disease. Identifying factors that convey cell survival in this setting may guide improvements in treatment. Estrogen (E2) can induce apoptosis in breast cancer cells that have been selected for survival after E2 deprivation for long periods (MCF-7:5C cells), but the mechanisms underlying E2-induced stress in this setting have not been elucidated. Here, we report that the c-Src kinase functions as a key adapter protein for the estrogen receptor (ER, ESR1) in its activation of stress responses induced by E2 in MCF-7:5C cells. E2 elevated phosphorylation of c-Src, which was blocked by 4-hydroxytamoxifen (4-OHT), suggesting that E2 activated c-Src through the ER. We found that E2 activated the sensors of the unfolded protein response (UPR), IRE1α (ERN1) and PERK kinase (EIF2AK3), the latter of which phosphorylates eukaryotic translation initiation factor-2α (eIF2α). E2 also dramatically increased reactive oxygen species production and upregulated expression of heme oxygenase HO-1 (HMOX1), an indicator of oxidative stress, along with the central energy sensor kinase AMPK (PRKAA2). Pharmacologic or RNA interference-mediated inhibition of c-Src abolished the phosphorylation of eIF2α and AMPK, blocked E2-induced ROS production, and inhibited E2-induced apoptosis. Together, our results establish that c-Src kinase mediates stresses generated by E2 in long-term E2-deprived cells that trigger apoptosis. This work offers a mechanistic rationale for a new approach in the treatment of endocrine-resistant breast cancer.


European Journal of Cancer | 2014

Inhibition of c-Src blocks oestrogen-induced apoptosis and restores oestrogen-stimulated growth in long-term oestrogen-deprived breast cancer cells

Ping Fan; Fadeke A. Agboke; Russell E. McDaniel; Elizabeth E. Sweeney; Xiaojun Zou; Karen Creswell; V. Craig Jordan

PURPOSE Our publications demonstrate that physiological concentrations of oestrogen (E2) induce endoplasmic reticulum and oxidative stress which finally result in apoptosis in E2-deprived breast cancer cells, MCF-7:5C. c-Src is involved in the process of E2-induced stress. To mimic the clinical administration of c-Src inhibitors, we treated cells with either E2, a c-Src inhibitor PP2, or the combination for 8 weeks to further explore the apoptotic potential of the c-Src inhibitor and E2 on MCF-7:5C cells. METHODS Protein levels of receptors and signalling pathways were examined by immunoblotting. Expression of mRNA was detected through real-time polymerase chain reaction (PCR). Cell cycles were analysed by flow cytometry. RESULTS Long-term treatment with PP2 alone or E2 alone decreased cell growth. In contrast, a combination of PP2 and E2 blocked apoptosis and the resulting cell line (MCF-7:PF) was unique, as they grew vigorously in culture with physiological levels of E2, which could be blocked by the pure antioestrogen ICI182,780. One major change was that PP2 collaborated with E2 to increase the level of insulin-like growth factor-1 receptor beta (IGF-1Rβ). Blockade of IGF-1Rβ completely abolished E2-stimulated growth in MCF-7:PF cells. Furthermore, combination treatment up-regulated transcription factors, Twist1 and Snail, and repressed E-cadherin expression which made MCF-7:PF cells display a characteristic phenotype of epithelial-mesenchymal transition (EMT). CONCLUSIONS These data illustrate the role of the c-Src inhibitor to block E2-induced apoptosis and enhance E2-stimulated growth. Caution must be exercised when considering c-Src inhibitors in clinical trials following the development of acquired resistance to aromatase inhibitors, especially in the presence of the patients own oestrogen.


Journal of Autoimmunity | 1995

The role of NK cell activity in the pathogenesis of poly I:C accelerated and spontaneous diabetes in the diabetes prone BB rat

Douglas O. Sobel; Norio Azumi; Karen Creswell; Daniel A. Holterman; Owen C. Blair; Joseph A. Bellanti; Val Abbassi; John C. Hiserodt

The development of insulin dependent diabetes mellitus (IDDM) and diabetes in the diabetes prone (DP) BB rat animal model of IDDM is thought to be due to an autoimmune process. Natural killer (NK) cells have been implicated but not proven to play a pathogenetic role in BB rats due to the increased NK cell number and activity found in these animals. We have recently reported that poly I:C, an inducer of cytokines and a potent enhancer of NK cell function, accelerates the development of diabetes in DP BB rats and induces diabetes in diabetes resistant (DR) BB rats. Since we have further demonstrated that poly I:C administration to BB rats increases NK cell number and levels of inducers of NK cell activity, interferon-alpha and IL-6 which is described therein, we tested the hypothesis that NK cell activity plays an important role in poly I:C accelerated disease. The role of NK cells in poly I:C accelerated diabetes and spontaneous diabetes was examined by determining whether selective depletion of NK cells using a rat NK cell specific antibody (anti-NKR-P1 antibody) alters the development of diabetes. The treatment of BB rats with anti-NKR-P1 antibody resulted in a significantly lower mean NK cell activity of splenic mononuclear cells than that found in control animals. However, the development of diabetes and degree of insulitis was not significantly different between treatment groups. BB rats administered anti-NKR-P1 antibody with poly I:C had a lower mean splenocyte NK cell activity and lower mean NK cell number within the peripheral blood and inflamed islets than rats administered poly I:C alone. However, anti-NKR-P1 antibody administration did not alter the accelerated development of diabetes or the degree of insulitis in poly I:C treated animals. These data document that NK cells do not play a major role in the pathogenesis of poly I:C accelerated diabetes or spontaneous diabetes in the DP BB rat.


Chemical Research in Toxicology | 2012

Detection of acrolein-derived cyclic DNA adducts in human cells by monoclonal antibodies

Jishen Pan; Bisola Awoyemi; Zhuoli Xuan; Priya Vohra; Hsiang-Tsui Wang; Marcin Dyba; Emily Greenspan; Ying Fu; Karen Creswell; Lihua Zhang; Deborah L. Berry; Moon-shong Tang; Fung-Lung Chung

Acrolein (Acr) is a ubiquitous environmental pollutant found in cigarette smoke and automobile exhaust. It can also be produced endogenously by oxidation of polyunsaturated fatty acids. The Acr-derived 1,N(2)-propanodeoxyguanosine (Acr-dG) adducts in DNA are mutagenic lesions that are potentially involved in human cancers. In this study, monoclonal antibodies were raised against Acr-dG adducts and characterized using ELISA. They showed strong reactivity and specificity toward Acr-dG, weaker reactivity toward crotonaldehyde- and trans-4-hydroxy-2-nonenal-derived 1,N(2)-propanodeoxyguanosines, and weak or no reactivity toward 1,N(6)-ethenodeoxyadenosine and 8-oxo-deoxyguanosine. Using these antibodies, we developed assays to detect Acr-dG in vivo: first, a simple and quick FACS-based assay for detecting these adducts directly in cells; second, a highly sensitive direct ELISA assay for measuring Acr-dG in cells and tissues using only 1 μg of DNA without DNA digestion and sample enrichment; and third, a competitive ELISA for better quantitative measurement of Acr-dG levels in DNA samples. The assays were validated using Acr-treated HT29 cell DNA samples or calf thymus DNA, and the results were confirmed by LC-MS/MS-MRM. An immunohistochemical assay was also developed to detect and visualize Acr-dG in HT29 cells as well as in human oral cells. These antibody-based methods provide useful tools for the studies of Acr-dG as a cancer biomarker and of the molecular mechanisms by which cells respond to Acr-dG as a ubiquitous DNA lesion.


Mutation Research | 2016

Nucleotide excision repair deficiency increases levels of acrolein-derived cyclic DNA adduct and sensitizes cells to apoptosis induced by docosahexaenoic acid and acrolein.

Jishen Pan; Elizabeth Sinclair; Zhuoli Xuan; Marcin Dyba; Ying Fu; Supti Sen; Deborah L. Berry; Karen Creswell; Jiaxi Hu; Rabindra Roy; Fung-Lung Chung

The acrolein derived cyclic 1,N(2)-propanodeoxyguanosine adduct (Acr-dG), formed primarily from ω-3 polyunsaturated fatty acids such as docosahexaenoic acid (DHA) under oxidative conditions, while proven to be mutagenic, is potentially involved in DHA-induced apoptosis. The latter may contribute to the chemopreventive effects of DHA. Previous studies have shown that the levels of Acr-dG are correlated with apoptosis induction in HT29 cells treated with DHA. Because Acr-dG is shown to be repaired by the nucleotide excision repair (NER) pathway, to further investigate the role of Acr-dG in apoptosis, in this study, NER-deficient XPA and its isogenic NER-proficient XAN1 cells were treated with DHA. The Acr-dG levels and apoptosis were sharply increased in XPA cells, but not in XAN1 cells when treated with 125μM of DHA. Because DHA can induce formation of various DNA damage, to specifically investigate the role of Acr-dG in apoptosis induction, we treated XPA knockdown HCT116+ch3 cells with acrolein. The levels of both Acr-dG and apoptosis induction increased significantly in the XPA knockdown cells. These results clearly demonstrate that NER deficiency induces higher levels of Acr-dG in cells treated with DHA or acrolein and sensitizes cells to undergo apoptosis in a correlative manner. Collectively, these results support that Acr-dG, a ubiquitously formed mutagenic oxidative DNA adduct, plays a role in DHA-induced apoptosis and suggest that it could serve as a biomarker for the cancer preventive effects of DHA.


Cancer Research | 2013

Abstract 120: Detection of acrolein-derived cyclic DNA adducts in human cells by monoclonal antibodies.

Jishen Pan; Bisola Awoyemi; Zhuoli Xuan; Priya Vohra; Hsiang-Tsui Wang; Marcin Dyba; Emily Greenspan; Ying Fu; Karen Creswell; Lihua Zhang; Deborah L. Berry; Moon-shong Tang; Fung-Lung Chung

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Acrolein (Acr) is a ubiquitous environmental pollutant found in cigarette smoke and automobile exhaust. It can also be produced endogenously by oxidation of polyunsaturated fatty acids. The Acr-derived 1,N2-propanodeoxyguanosine (Acr-dG) adducts in DNA are mutagenic lesions that are potentially involved in human cancers. In this study, monoclonal antibodies were raised against Acr-dG adducts and characterized using ELISA. They showed strong reactivity and specificity towards Acr-dG, weaker reactivity towards crotonaldehyde- and trans-4-hydroxy-2-nonenal-derived 1,N2-propanodeoxyguanosines, and weak or no reactivity towards 1,N6-ethenodeoxyadenosine, 8-oxo-deoxyguanosine, and benzo(a)pyrene- and malondialdehdye-derived adducts. Using these novel antibodies, we developed assays to detect Acr-dG in vivo: First, a simple and quick FACS-based assay for detecting these adducts directly in cells; Second, a highly sensitive direct ELISA assay for measuring Acr-dG in cells and tissues using only one μg DNA; And third, a competitive ELISA for better quantitative measurement of Acr-dG levels in DNA. The assays were validated using Acr-treated HT29 cell DNA samples or calf thymus DNA and the results were confirmed by LC-MS/MS-MRM. An immunohistochemical assay was also developed to detect and visualize Acr-dG in HT29 cells as well as in human oral cells. These antibody-based methods provide useful tools for the studies of Acr-dG as a cancer biomarker and of the molecular mechanisms by which cells respond to Acr-dG as a ubiquitous DNA lesion. (This work was supported by NCI grant CA134892) Citation Format: Jishen Pan, Bisola Awoyemi, Zhuoli Xuan, Priya Vohra, Hsiang-Tsui Wang, Marcin Dyba, Emily Greenspan, Ying Fu, Karen Creswell, Lihua Zhang, Deborah Berry, Moon-Shong Tang, Fung-Lung Chung. Detection of acrolein-derived cyclic DNA adducts in human cells by monoclonal antibodies. [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 120. doi:10.1158/1538-7445.AM2013-120


Cancer Research | 2013

Abstract 830: Transcriptional modulation of estrogen-induced apoptosis through activation of c-Fos/c-Jun in long-term estrogen deprived breast cancer cells.

Ping Fan; Fadeke A. Agboke; Obi L. Griffith; Russell E. McDaniel; Xiaojun Zou; Karen Creswell; Joe W. Gray; Virgil Craig Jordan

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Recent clinical trials have demonstrated that estrogen (E2) alone reduces breast cancer incidence in postmenopausal women and has therapeutic effects on aromatase inhibitor resistant patients which both are related with the effect of E2-induced apoptosis. We have shown that E2 induces apoptosis in long-term E2 deprived breast cancer cells (MCF-7:5C) through stress responses, but the molecular mechanism underlying E2-induced stress remains to be elucidated. Here, we report that E2 activated the sensors of unfolded protein response (UPR) inositol-requiring protein 1 alpha (IRE1α) and PRK-like endoplasmic reticulum kinase (PERK) within 24 hours. Knockdown of PERK and IRE-1α through small interferon RNAs (siRNA) partially prevented E2-induced apoptosis, which suggested that endoplasmic reticulum stress was involved in the E2-induced apoptosis. Further examination showed E2 activated both classical estrogen responsive element (ERE) pathway and nonclassical activating protein-1 (AP-1) pathway in MCF-7:5C cells. Classical ERE regulated genes were not directly involved in E2-induced apoptosis. However, the transcription factor c-Fos acted as a critical trigger involved in stress responses induced by E2 in MCF-7:5C cells. E2 immediately elevated c-Fos expression in MCF-7:5C cells but not in wild-type breast cancer cells and 4-hydroxytamoxifen blocked this stimulation which suggested that E2 activated c-Fos through estrogen receptor (ER). E2 increased phosphorylation of c-Jun after 24 hours treatment but did not significantly enhance the abundance of c-Jun as c-Fos. c-Fos protein forms stable heterodimers with c-Jun which preferentially bind to phorbol 12-O-tetradecanoate-13-acetate (TPA)-responsive element (TRE). Interestingly, low doses of TPA which activated TRE activity could induce apoptosis and activated apoptosis-related genes similar to E2 in MCF-7:5C cells. Knockdown of c-Fos and c-Jun with specific siRNAs blocked E2-induced apoptosis, which confirmed the activation of AP-1 played a critical role in the process of apoptosis induced by E2. Although c-Fos closely associates with c-Jun to form a stable heterodimer, they had differential functions in modulation of UPR and other nuclear transcription factors. Knockdown of c-Fos but not c-Jun could inhibit the activation of UPR by E2 in MCF-7:5C cells. Furthermore, downregulation of c-Fos abolished the expression of multiple nuclear transcription factors such as SRC-3, NF-κB, and p300 etc in MCF-7:5C cells. Overall, these data illustrate that c-Jun might provide a dimerizing site for c-Fos, whereas c-Fos was identified as a pivotal regulatory target of E2 in MCF-7:5C cells to trigger the apoptotic cascades. This study provides an important rationale for further exploration of E2-induced apoptosis in endocrine resistant breast cancer to improve clinical benefit. Citation Format: Ping Fan, Fadeke Agboke, Obi L. Griffith, Russell E. McDaniel, Xiaojun Zou, Karen Creswell, Joe W. Gray, Virgil Craig Jordan. Transcriptional modulation of estrogen-induced apoptosis through activation of c-Fos/c-Jun in long-term estrogen deprived breast cancer cells. [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 830. doi:10.1158/1538-7445.AM2013-830


Cancer Research | 2001

RGD-Tachyplesin Inhibits Tumor Growth

Yixin Chen; Xue-Ming Xu; Shuigen Hong; Jinguo Chen; Ningfei Liu; Charles B. Underhill; Karen Creswell; Lurong Zhang

Collaboration


Dive into the Karen Creswell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ping Fan

Georgetown University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge