Network


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

Hotspot


Dive into the research topics where Jane Turbov is active.

Publication


Featured researches published by Jane Turbov.


Cancer Prevention Research | 2009

Ovarian Adenocarcinomas in the Laying Hen and Women Share Similar Alterations in p53, ras, and HER-2/neu

Hakim Aa; Catherine P. Barry; Barnes Hj; K. E. Anderson; James N. Petitte; Regina S. Whitaker; J.M. Lancaster; R. Wenham; Donna K. Carver; Jane Turbov; Andrew Berchuck; Levy Kopelovich; G. Rodriguez

We examined alterations in the p53 tumor suppressor gene and the ras and HER-2/neu oncogenes in chicken ovarian cancers to determine if these tumors have genetic alterations similar to those in human ovarian adenocarcinomas. Mutations in the p53 tumor suppressor gene and the H-ras and K-ras oncogenes were assessed by direct sequencing in 172 ovarian cancers obtained from 4-year-old birds enrolled at age 2 in two separate 2-year chemoprevention trials. Birds in trial B had approximately twice as many lifetime ovulations as those in trial A. Immunohistochemical staining for the HER-2/neu oncogene was done on a subset of avian ovarian and oviductal adenocarcinomas. Alterations in p53 were detected in 48% of chicken ovarian cancers. Incidence of p53 alterations varied according to the number of lifetime ovulations, ranging from 14% in trial A to 96% in trial B (P < 0.01). No mutations were seen in H-ras, and only 2 of 172 (1.2%) tumors had K-ras mutations. Significant HER-2/neu staining was noted in 10 of 19 ovarian adenocarcinomas but in only 1 of 17 oviductal adenocarcinomas. Similar to human ovarian cancers, p53 alterations are common in chicken ovarian adenocarcinomas and correlate with the number of lifetime ovulations. Ras mutations are rare, similar to high-grade human ovarian cancers. HER-2/neu overexpression is common and may represent a marker to exclude an oviductal origin in cancers involving both the ovary and oviduct.


Journal of Surgical Oncology | 1999

Tyrosine kinase inhibitors as antiproliferative agents against an estrogen‐dependent breast cancer cell line in vitro

George M. Twaddle; Jane Turbov; Naxin Liu; Satya M. Murthy

Receptor tyrosine kinase (RTK) activation is critical for growth factor‐mediated cell proliferation. Blockade of RTK activation inhibits growth factor‐induced cell proliferation. A panel of RTK inhibitors (tyrphostins) have been tested and compared for their antiproliferative effects on the hormone‐dependent human breast cancer cell line, MCF‐7, in vitro.


Cancer Prevention Research | 2013

Progesterone enhances calcitriol antitumor activity by upregulating vitamin D receptor expression and promoting apoptosis in endometrial cancer cells.

Laura R. Lee; P.N. Teng; Huyen Nguyen; Brian L. Hood; Leyla Kavandi; Guisong Wang; Jane Turbov; Larry G. Thaete; Chad A. Hamilton; George Larry Maxwell; Gustavo C. Rodriguez; Thomas P. Conrads; Viqar Syed

Human studies suggest that progesterone and calcitriol may prove beneficial in preventing or inhibiting oncogenesis, but the underlying mechanism is not fully understood. The current study investigates the effects of progesterone, calcitriol, and their combination on immortalized human endometrial epithelial cells and endometrial cancer cells and identifies their targets of action. Combination treatment with both agents enhanced vitamin D receptor expression and inhibited cell proliferation through caspase-3 activation and induction of G0–G1 cell-cycle arrest with associated downregulation of cyclins D1 and D3 and p27 induction. We used mass spectrometry–based proteomics to measure protein abundance differences between calcitriol-, progesterone-, or combination-exposed endometrial cells. A total of 117 proteins showed differential expression among these three treatments. Four proteins were then selected for validation studies: histone H1.4 (HIST1H1E), histidine triad nucleotide-binding protein 2 (HINT2), IFN-induced, double-stranded RNA-activated protein kinase (EIF2AK2), and Bcl-2–associated X protein (BAX). Abundance levels of selected candidates were low in endometrial cancer cell lines versus the immortalized endometrial epithelial cell line. All four proteins displayed elevated expression in cancer cells upon exposure to calcitriol, progesterone, or the combination. Further BAX analysis through gain- or loss-of-function experiments revealed that upregulation of BAX decreased cell proliferation by changing the BAX:BCL-2 ratio. Knockdown of BAX attenuated progesterone- and calcitriol-induced cell growth inhibition. Our results showed that progesterone and calcitriol upregulate the expression of BAX along with other apoptosis-related proteins, which induce inhibition of endometrial cancer cell growth by apoptosis and cell-cycle arrest. Cancer Prev Res; 6(7); 731–43. ©2013 AACR.


Cancer Epidemiology, Biomarkers & Prevention | 2008

Progestin Treatment Induces Apoptosis and Modulates Transforming Growth Factor-β in the Uterine Endometrium

Gustavo C. Rodriguez; B.J. Rimel; William Watkin; Jane Turbov; Cathy Barry; Hongyan Du; George Larry Maxwell; J.M. Cline

Background: Epidemiologic, animal, and human data suggest that progestins are potent endometrial cancer preventive agents. In the ovarian surface epithelium, progestins have been hypothesized to confer a cancer preventive effect via apoptosis and modulation of transforming growth factor-β (TGF-β). Given that the ovarian epithelium and endometrium share a common embryologic origin and similar reproductive and hormonal risk factors for malignancy, we tested the hypothesis that progestins confer biological effects in the endometrium similar to those in the ovary. Methods: Postmenopausal female macaques (n = 78) were randomized into four groups to receive a diet for 36 months containing no hormone versus conjugated equine estrogen (CEE), medroxyprogesterone acetate (MPA), or CEE + MPA. The endometrium was then examined immunohistochemically for treatment-specific changes using antibodies to activated caspase-3 (for apoptosis), Ki-67 (proliferation), and the TGF-β1, TGF-β2, and TGF-β3 isoforms. Results: Percentages of caspase-positive endometrial glandular cells were 3- to 5-fold higher in CEE + MPA–treated animals compared with all others (P < 0.05). Caspase-expressing cells were six times more numerous in the endometrial stroma of animals treated with MPA alone relative to other groups (P < 0.0001). Induction of endometrial glandular cell apoptosis in the CEE + MPA–treated group was associated with a dramatic increase in expression of TGF-β2 and TGF-β3 in the stromal compartment of the endometrium (P < 0.0001). Conclusion: Progestin treatment activates chemopreventive biological effects in the endometrium that are similar to those in the ovarian surface epithelium. These data may facilitate identification of a chemopreventive approach that dramatically lessens the risk of both uterine and ovarian cancer. (Cancer Epidemiol Biomarkers Prev 2008;17(3):578–84)


Cancer Prevention Research | 2013

Evidence of a Chemopreventive Effect of Progestin Unrelated to Ovulation on Reproductive Tract Cancers in the Egg-laying Hen

Gustavo C. Rodriguez; Barnes Hj; K. E. Anderson; Regina S. Whitaker; Andrew Berchuck; James N. Petitte; J.M. Lancaster; R. Wenham; Jane Turbov; Roger Day; George Larry Maxwell; Donna K. Carver

Epidemiologic, laboratory, and animal evidence suggests that progestins and vitamin D may be potent ovarian cancer preventives. Our objectives were to evaluate progestins as reproductive tract cancer chemopreventives in the chicken, determine whether restricted ovulation affected the incidence of reproductive tract tumors, and assess whether vitamin D would confer cancer protection either alone or in addition to progestin. A total of 2,400 two-year-old Single Comb White Leghorns were randomized into six groups (400 each) with hormonal and dietary manipulation for 2 years as follows: (i) no intervention, regular feed/caloric intake, (ii) control, (iii) vitamin D, (iv) the progestin levonorgestrel, (v) vitamin D plus levonorgestrel, and (vi) the progestin Provera (medroxyprogesterone acetate). Groups 2 to 6 were caloric restricted to inhibit ovulation. Our results indicated that caloric restriction decreased egg production by more than 60%, and was associated with a greater than 70% decrease in reproductive tract cancers. Ovulatory events did not differ among the caloric-restricted groups (groups 2–6), except for the group receiving levonorgestrel, which had fewer ovulatory events than controls (P = 0.046). After correcting for egg production, birds receiving progestins had significantly fewer reproductive tract cancers [OR, 0.61; confidence interval (CI), 0.39–0.95; P = 0.03], with similar proportionate reductions in tumors arising in either the ovary or oviduct. Vitamin D did not significantly affect cancer incidence overall, or add to the cancer preventive effect of progestins. This study suggests a protective effect of progestins against ovarian and oviductal cancers. These data support the concept that progestins provide a chemopreventive effect unrelated to ovulation. Cancer Prev Res; 6(12); 1283–92. ©2013 AACR.


Gynecologic Oncology | 2016

Progestins inhibit calcitriol-induced CYP24A1 and synergistically inhibit ovarian cancer cell viability: An opportunity for chemoprevention

Gustavo C. Rodriguez; Jane Turbov; R. Rosales; Jennifer Yoo; Jessica Hunn; Katherine J. Zappia; Kaarin Lund; Catherine P. Barry; Isabel V. Rodriguez; J. Wesley Pike; Thomas P. Conrads; Kathleen M. Darcy; George Larry Maxwell; Chad A. Hamilton; Viqar Syed; Larry G. Thaete

OBJECTIVES Previously we have shown in endometrial cells that progesterone (P4) and calcitriol (CAL, 1,25(OH)2D3) synergistically promote apoptosis and that progestins induce expression of the vitamin D receptor. In the current study we examined the progestin/vitamin D combination in ovarian cells and searched for other progestin-related effects on vitamin D metabolism that may underlie the novel interaction between progestins and vitamin D, including whether progestins inhibit CYP24A1, the enzyme that renders CAL inactive. METHODS We investigated the impact of P4 on CAL-induced CYP24A1 expression in cancer cell lines expressing progesterone receptors (PRs), [OVCAR-5, OVCAR-3-PGR (PR-transfected OVCAR-3 ovarian line), and T47D-WT, T47D-A and T47D-B (breast lines expressing PRs or individual PR isoforms)] or lines that do not express PRs (OVCAR-3 and T47D-Y). We examined CYP24A1 expression using RT-PCR and western blotting, and apoptosis by TUNEL. We also investigated P4 inhibition of Cyp24a1 in ovaries from CAL-treated mice. RESULTS CAL treatment induced CYP24A1 expression. When co-treated with P4, cell lines expressing PRs showed marked inhibition of CYP24A1 expression (p<0.001), along with increased apoptosis (p<0.01); cells not expressing PRs did not. Mouse ovaries showed a significant reduction in CAL-induced Cyp24a1 mRNA (p<0.001) and protein (p<0.01) in response to P4. CONCLUSIONS We show for the first time that progestins and vitamin D synergistically reduce cell viability and induce apoptosis in ovarian cells and that progestins PR-dependently inhibit CAL-induced CYP24A1, thus extending CAL activity. The combination of progestins and vitamin D deserves further consideration as a strategy for inhibiting ovarian carcinogenesis.


American Journal of Obstetrics and Gynecology | 2012

Nonsteroidal antiinflammatory drugs and progestins synergistically enhance cell death in ovarian epithelial cells

Gustavo C. Rodriguez; Jane Turbov; Andrew Berchuck; M. Sharon Stack; Jean A. Hurteau; Larry G. Thaete; Catherine P. Barry

OBJECTIVE There is growing evidence that progestins and nonsteroidal antiinflammatory drugs (NSAIDs) may prevent ovarian cancer. Because both induce apoptosis, we investigated the potential for synergistic impact of combined drug treatment on cell death. STUDY DESIGN Using normal and malignant human ovarian epithelial cells and an NSAID-sensitive human colon cancer cell line, we evaluated the effects of progestins and NSAIDs alone and in combination on apoptosis. RESULTS Both progestins and NSAIDs dose dependently inhibited cell growth (P < .0001). Doses of NSAIDs or progestins that independently reduced cell viability by less than 30% synergistically reduced cell viability by 70-95% when combined. Similarly, the NSAID/progestin combination conferred 4- to 18-fold (P < .05) increased apoptosis over either treatment alone. CONCLUSION Our results suggest it may be possible to combine progestins and NSAIDs to achieve ovarian cancer prevention at lower doses of each than are required for single administration, thereby lessening the risk of side effects posed by these agents.


Oncotarget | 2016

Progesterone potentiates the growth inhibitory effects of calcitriol in endometrial cancer via suppression of CYP24A1

Amber A. Bokhari; Laura R. Lee; Dewayne Raboteau; Jane Turbov; Isabel V. Rodriguez; John Wesley Pike; Chad A. Hamilton; George Larry Maxwell; Gustavo C. Rodriguez; Viqar Syed

Here, we evaluated the expression of CYP24A1, a protein that inactivates vitamin D in tissues. CYP24A1 expression was increased in advanced-stage endometrial tumors compared to normal tissues. Similarly, endometrial cancer cells expressed higher levels of CYP24A1 than immortalized endometrial epithelial cells. RT-PCR and Western blotting were used to examine CYP24A1 mRNA and protein levels in endometrial cancer cells after 8, 24, 72, and 120 h of exposure to progesterone, progestin derivatives and calcitriol, either alone or in combination. Progestins inhibited calcitriol-induced expression of CYP24A1 and splice variant CYP24SV mRNA and protein in cancer cells. Furthermore, actinomycin D, but not cycloheximide, blocked calcitriol-induced CYP24A1 splicing. siRNA-induced knockdown of CYP24A1 expression sensitized endometrial cancer cells to calcitriol-induced growth inhibition. These data suggest that CYP24A1 overexpression reduces the antitumor effects of calcitriol in cancer cells and that progestins may be beneficial for maintaining calcitriols anti-endometrial cancer activity.


Cancer Research | 2017

Abstract 1259: Stem cell property-suppressing microRNAs are stimulated and their target proteins inhibited by vitamin D and progesterone in ovarian cancer cells

R. Rosales; Jane Turbov; Jennifer Yoo; Gustavo C. Rodriguez; Larry G. Thaete

Vitamin D (1,25(OH)2D3) and progesterone (P4) have both been shown to reduce the incidence of ovarian cancer. Mechanisms involved in their actions are not fully known but each one can enhance the effect of the other. Six microRNAs (miRNAs), associated with suppression of cancer stem cell properties, were investigated first in a screening array and then with a more definitive RT-PCR investigation of the response of these miRNAs to 100 nM 1,25(OH)2D3 and 100 nM P4. These experiments were conducted in cells that do (OVCAR-3-PGR) and do not (SKOV3) express nuclear progesterone receptors (PGR). Let-7a and Let-7b are known to decrease expression of Ras and c-Myc and reduce stem cell self-renewal. Let-7b expression was increased (p miR-26a and miR-101 are known to downregulate EpCAM and EZH2, leading to decreased invasion and metastasis and to reduced proliferation. miR-101 increased (p miR-200b and miR-200c are known to suppress expression of ZEB1 which in turn decreases Bmi1 and Sox2 to increase E-cadherin (epithelial marker) and decrease vimentin (mesenchymal marker), thus inhibiting the epithelial-to-mesenchymal transition. Expression of miR-200b was increased (p These results point not only to differences among cancer cell lines but they also emphasize a possible mechanism by which P4 and 1,25(OH)2D3 may work together to prevent the occurrence of ovarian cancer, namely by suppressing stem cell renewal and the formation of neoplastic lesions. The results also reveal that P4 may act via other signaling pathways than its primary nuclear receptors to effect suppression of stem cell properties. Increased expression of cancer stem cell-suppressing miRNAs may be an important mechanism mediating the efficacy of a progestin-based ovarian cancer prevention strategy. Citation Format: Rebecca Rosales, Jane Turbov, Jennifer Yoo, Gustavo C. Rodriguez, Larry G. Thaete. Stem cell property-suppressing microRNAs are stimulated and their target proteins inhibited by vitamin D and progesterone in ovarian cancer cells [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 1259. doi:10.1158/1538-7445.AM2017-1259


Cancer Research | 2013

Abstract 1716: Calcitriol and progesterone synergistically inhibit growth in endometrial cancer cells.

Laura R. Lee; Brian L. Hood; Huyen Nguyen; Leyla Kavandi; Jane Turbov; Larry G. Thaete; Chad A. Hamilton; Gustavo C. Rodriguez; George L. Maxwell; Thomas P. Conrads; Viqar Syed

Human studies suggest that progesterone and calcitriol may prove beneficial in preventing or inhibiting oncogenesis, but the underlying mechanism is not fully understood. The current study investigates the effects of progesterone, calcitriol and their combination on immortalized human endometrial epithelial cells and endometrial cancer cells and identifies their targets of action. The combined cell treatment enhanced vitamin D receptor expression and synergistically inhibited cell proliferation through caspase-3 activation and induction of G0/G1 cell cycle arrest with associated down-regulation of cyclin D1, D3 and p27 induction. We used mass spectrometry-based proteomics to measure protein abundance differences between calcitriol-, progesterone-, or combination-exposed endometrial cells. A total of 117 proteins showed differential expression amongst these three treatments. Four proteins were then selected for validation studies: histone H1.4 (HIST1H1E), histidine triad nucleotide-binding protein 2 (HINT2), interferon-induced, double-stranded RNA-activated protein kinase (EIF2AK2), and Bcl-2-associated X protein (BAX). Abundance levels of selected candidates were low in endometrial cancer cell lines versus the immortalized endometrial epithelial cell line. All four proteins displayed elevated expression in cancer cells upon exposure to calcitriol, progesterone or the combination. Further BAX analysis through gain or loss of function experiments revealed that upregulation of BAX decreased cell proliferation by changing the BAX:Bcl2 ratio. Knock down of BAX attenuated progesterone- and calcitriol-induced cell growth inhibition. Our results showed that progesterone and calcitriol up-regulate the expression of BAX along with other apoptosis-related proteins, which induced inhibition of endometrial cancer cell growth by apoptosis and cell cycle arrest. Citation Format: Laura R. Lee, Brian L. Hood, Huyen Nguyen, Leyla Kavandi, Jane M. Turbov, Larry G. Thaete, Chad A. Hamilton, Gustavo C. Rodriguez, George L. Maxwell, Thomas P. Conrads, Viqar Syed. Calcitriol and progesterone synergistically inhibit growth in endometrial 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 1716. doi:10.1158/1538-7445.AM2013-1716

Collaboration


Dive into the Jane Turbov's collaboration.

Top Co-Authors

Avatar

Gustavo C. Rodriguez

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Larry G. Thaete

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Chad A. Hamilton

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

R. Rosales

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Viqar Syed

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George Larry Maxwell

Walter Reed Army Medical Center

View shared research outputs
Top Co-Authors

Avatar

Donna K. Carver

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

K. E. Anderson

North Carolina State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge