Network


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

Hotspot


Dive into the research topics where Jacqueline J. Stevens is active.

Publication


Featured researches published by Jacqueline J. Stevens.


International Journal of Environmental Research and Public Health | 2010

Arsenic Trioxide Modulates DNA Synthesis and Apoptosis in Lung Carcinoma Cells

Alice M. Walker; Jacqueline J. Stevens; Kenneth Ndebele; Paul B. Tchounwou

Arsenic trioxide, the trade name Trisenox, is a drug used to treat acute promyleocytic leukemia (APL). Studies have demonstrated that arsenic trioxide slows cancer cells growth. Although arsenic influences numerous signal-transduction pathways, cell-cycle progression, and/or apoptosis, its apoptotic mechanisms are complex and not entirely delineated. The primary objective of this research was to evaluate the effects of arsenic trioxide on DNA synthesis and to determine whether arsenic-induced apoptosis is mediated via caspase activation, p38 mitogen–activated protein kinase (MAPK), and cell cycle arrest. To achieve this goal, lung cancer cells (A549) were exposed to various concentrations (0, 2, 4, 6, 8, and 10 μg/mL) of arsenic trioxide for 48 h. The effect of arsenic trioxide on DNA synthesis was determined by the [3H]thymidine incorporation assay. Apoptosis was determined by the caspase-3 fluorescein isothiocyanate (FITC) assay, p38 MAP kinase activity was determined by an immunoblot assay, and cell-cycle analysis was evaluated by the propidium iodide assay. The [3H]thymidine-incorporation assay revealed a dose-related cytotoxic response at high levels of exposure. Furthermore, arsenic trioxide modulated caspase 3 activity and induced p38 MAP kinase activation in A549 cells. However, cell-cycle studies showed no statistically significant differences in DNA content at subG1 check point between control and arsenic trioxide treated cells.


International Journal of Environmental Research and Public Health | 2010

The Effects of Arsenic Trioxide on DNA Synthesis and Genotoxicity in Human Colon Cancer Cells

Jacqueline J. Stevens; Barbara Graham; Alice M. Walker; Paul B. Tchounwou; Christian Rogers

Colon cancer is the third leading cause of cancer-related deaths worldwide. Recent studies in our laboratory have demonstrated that arsenic trioxide is cytotoxic in human colon cancer (HT-29), lung (A549) and breast (MCF-7) carcinoma cells. The purpose of the present study is to investigate the effects of arsenic trioxide on DNA synthesis and the possible genotoxic effects on human colon cancer cells. HT-29 cells were cultured according to standard protocol, followed by exposure to various doses (0, 2, 4, 6, 8, 10, and 12 μg/mL) of arsenic trioxide for 24 h. The proliferative response (DNA synthesis) to arsenic trioxide was assessed by [3H]thymidine incorporation. The genotoxic effects of arsenic-induced DNA damage in a human colon cancer cell line was evaluated by the alkaline single cell gel electrophoresis. Results indicated that arsenic trioxide affected DNA synthesis in HT-29 cells in a biphasic manner; showing a slight but not significant increase in cell proliferation at lower levels of exposure (2, 4 and 6 μg/mL) followed by a significant inhibition of cell proliferation at higher doses (i.e., 8 and 10 μg/mL). The study also confirmed that arsenic trioxide exposure caused genotoxicity as revealed by the significant increase in DNA damage, comet tail-lengths, and tail moment when compared to non-exposed cells. Results of the [3H]thymidine incorporation assay and comet assay revealed that exposure to arsenic trioxide affected DNA synthesis and exhibited genotoxic effects in human colon cancer cells.


Journal of Cancer Science & Therapy | 2016

Evaluation of Arsenic Trioxide Potential for Lung Cancer Treatment:Assessment of Apoptotic Mechanisms and Oxidative Damage

Alice M. Walker; Jacqueline J. Stevens; Kenneth Ndebele; Paul B. Tchounwou

Background Lung cancer is one of the most lethal and common cancers in the world, causing up to 3 million deaths annually. The chemotherapeutic drugs that have been used in treating lung cancer include cisplatin-pemetrexed, cisplastin-gencitabinoe, carboplatin-paclitaxel and crizotinib. Arsenic trioxide (ATO) has been used in the treatment of acute promyelocytic leukemia. However, its effects on lung cancer are not known. We hypothesize that ATO may also have a bioactivity against lung cancer, and its mechanisms of action may involve apoptosis, DNA damage and changes in stress-related proteins in lung cancer cells. Methods To test the above stated hypothesis, lung carcinoma (A549) cells were used as the test model. The effects of ATO were examined by performing 6-diamidine-2 phenylindole (DAPI) nuclear staining for morphological characterization of apoptosis, flow cytometry analysis for early apoptosis, and western blot analysis for stress-related proteins (Hsp70 and cfos) and apoptotic protein expressions. Also, the single cell gel electrophoresis (Comet) assay was used to evaluate the genotoxic effect. Results ATO-induced apoptosis was evidenced by chromatin condensation and formation of apoptotic bodies as revealed by DAPI nuclear staining. Cell shrinkage and membrane blebbing were observed at 4 and 6 µg/ml of ATO. Data from the western blot analysis revealed a significant dose-dependent increase (p < 0.05) in the Hsp 70, caspase 3 and p53 protein expression, and a significant (p < 0.05) decrease in the cfos, and bcl-2 protein expression at 4 and 6 µg/ml of ATO. There was a slight decrease in cytochrome c protein expression at 4 and 6 µg/ ml of ATO. Comet assay data revealed significant dose-dependent increases in the percentages of DNA damage, Comet tail lengths, and Comet tail moment. Conclusion Taken together our results indicate that ATO is cytotoxic to lung cancer cells and its bioactivity is associated with oxidative damage, changes in cellular morphology, and apoptosis.


Journal of Cancer Science & Therapy | 2017

Arsenic Trioxide Induces Apoptosis via Specific Signaling Pathways in HT-29 Colon Cancer Cells

Jacqueline J. Stevens; Barbara Graham; Erika Dugo; Bezawit Berhaneselassie-Sumner; Kenneth Ndebele; Paul B. Tchounwou

Background Arsenic trioxide (ATO) is highly effective in the treatment of patients with acute promyelocytic leukemia (APL). It is a chemotherapeutic agent that has been shown to induce apoptosis in several tumor cell lines. However, research into its effects on colon carcinoma cells is still very limited. We previously reported that ATO is cytotoxic and causes DNA damage in HT-29 human colorectal adenocarcinoma cells. In the present study, we further evaluated its effect on oxidative stress (OS), and examined its apoptotic mechanisms of action on HT-29 cells. Methods OS was assessed by spectrophotometric measurements of MDA levels while cell cycle analysis was evaluated by flow cytometry to determine whether ATO induces cell cycle arrest. Its effect on early apoptosis was also evaluated by flow cytometry using Annexin V-FITC/PI staining. Fluorescence microscopy was used to detect the morphological changes, and Western blotting was carried out to determine the expression of apoptosis-related proteins. Results The lipid peroxidation assay revealed a dose-dependent increase in MDA production. DAPI staining showed morphological changes in the cell’s nucleus due to apoptosis. Cell cycle analysis and Annexin V-FITC assay also demonstrated a dose-dependent effect of ATO in the accumulation of cells at the sub G1 phase, and the percentages of Annexin V-positive cells, respectively. Western blot data showed that ATO upregulated the expression of caspase 3, Bax, and cytochrome C, and down-regulated the expression of Bcl-2. Conclusion Taken together, our findings indicate that ATO induces OS and cytotoxicity in HT-29 cells through the mitochondria mediated intrinsic pathway of apoptosis.


International Journal of Environmental Research and Public Health | 2014

Enhancement of Arsenic Trioxide-Mediated Changes in Human Induced Pluripotent Stem Cells (IPS)

Barbara Graham; Jacqueline J. Stevens; Phatia Wells; Jennifer Sims; Christian Rogers; Sophia S. Leggett; Stephen I. N. Ekunwe; Kenneth Ndebele

Induced pluripotent stem cells (IPS) are an artificially derived type of pluripotent stem cell, showing many of the same characteristics as natural pluripotent stem cells. IPS are a hopeful therapeutic model; however there is a critical need to determine their response to environmental toxins. Effects of arsenic on cells have been studied extensively; however, its effect on IPS is yet to be elucidated. Arsenic trioxide (ATO) has been shown to inhibit cell proliferation, induce apoptosis and genotoxicity in many cells. Based on ATOs action in other cells, we hypothesize that it will induce alterations in morphology, inhibit cell viability and induce a genotoxic effect on IPS. Cells were treated for 24 hours with ATO (0–9 µg/mL). Cell morphology, viability and DNA damage were documented. Results indicated sufficient changes in morphology of cell colonies mainly in cell ability to maintain grouping and ability to remain adherent. Cell viability decreased in a dose dependent manner. There were significant increases in tail length and moment as well as destruction of intact DNA as concentration increased. Exposure to ATO resulted in a reproducible dose dependent sequence of events marked by changes in morphology, decrease of cell viability, and induction of genotoxicity in IPS.


Cancer Research | 2011

Abstract 4210: Oxidative stress, DNA damage, and apoptosis mediated by arsenic trioxide in lung carcinoma cells

Alice M. Walker; Jacqueline J. Stevens; Paul B. Tchounwou

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Lung cancer is one of the most lethal and common of cancers in the world, causing up to 3 million deaths annually. Arsenic trioxide (ATO) has been used in the treatment of relapsed/refractory acute promyelocytic leukemias. However, its effects on lung cancer are not known. We hypothesize that ATO may also have a bioactivity against lung cancer, and its mechanisms of action may involve apoptosis, DNA damage and changes in stress-related proteins in lung cancer cells. Using lung cancer (A549) cells as test model, mthe effects of ATO were examined by 6-diamidine-2 phenylindole (DAPI) nuclear staining for apoptosis, commet assay for genotoxicity ane western blotanalysis for stress protein (Hsp 70 and cfos) expression. Morphological changes of apoptosis (condensed chromatin and apoptotic bodies) were observed with DAPI and cell shrinkage and membrane blebbing without DAPI at 4 and 6µg/ml of ATO. The comet assay data revealed that DNA damage was significant as indicated by increased of the precent of DNA damage, tailmoment and tail length. There was significant increase in Hsp70 expression and significant decrease in cfos in ATO-treated cells compared to controls. ATO is cytotoxic to lung cancer cells and its toxicity is mediated via oxidative stress and DNA damage leading to apoptosis. However, further studies are warranted to understand its specific molecular mechanisms of toxicity. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4210. doi:10.1158/1538-7445.AM2011-4210


Cancer Research | 2010

Abstract 3455: Arsenic trioxide modulates DNA synthesis and apoptosis in lung carcinoma cells

Alice M. Walker; Jacqueline J. Stevens; Paul B. Tchounwou

Arsenic trioxide, the trade name Trisenox, is a drug used to treat acute promyleocytic leukemia (APL). Studies have demonstrated that arsenic trioxide slows the cancer cells growth. Although it has been reported that arsenic acts on cells by influencing numerous signal transduction pathways, cell cycle progression, and/or apoptosis, its apoptotic mechanisms are very complex. The primary objective of this research is to evaluate the effects of arsenic trioxide on DNA synthesis and to determine whether arsenic induced-apoptosis is mediated via caspase activation, p38 mitogen-activated protein kinase (MAPK), and cell cycle arrest. To achieve this goal, the lung cancer (A549) cells were cultured following standard protocol, and exposed to various doses (0, 2, 4, and 6 µg/ml) of arsenic trioxide for 48 h. The effect of arsenic trioxide on DNA synthesis was determined by [ 3 H]thymidine incorporation assay. Apoptosis was determined by caspase-3 fluorescein isothiocyanate (FITC) assay, and immunoblot analysis (p38 MAP kinase activity). The cell cycle analysis was evaluated propidium iodide assay. [ 3 H]Thymidine incorporation assay revealed dose-related cytotoxic response at higher levels of exposure. Our findings revealed that arsenic trioxide modulated caspase 3 activity and induced p38 map kinase activation in lung carcinoma (A549) cells. The data revealed no cell cycle arrest at the sub-G 0 in A549 cells when treated with arsenic trioxide. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3455.


Metal ions in biology and medicine : proceedings of the ... International Symposium on Metal Ions in Biology and Medicine held ... = Les ions metalliques en biologie et en medecine : ... Symposium international sur les ions metalliques ... | 2008

Cytotoxic Effect of Arsenic Trioxide in Adenocarcinoma Colorectal Cancer (HT-29) Cells.

Jacqueline J. Stevens; Barbara Graham-Evans; Alice M. Walker; Brinda Armstead; Paul B. Tchounwou


International Journal of Environmental Research and Public Health | 2005

Analysis of gene regulation in rabbit corneal epithelial cells induced by ultraviolet radiation.

Jacqueline J. Stevens; Christian Rogers; Carolyn B. Howard; Caronda Moore; Lai-Man Chan


Annals of Oncology | 2016

P-018Assessment of therapeutic potential of arsenic trioxide in the management of human hepatocellular carcinoma

E. Dugo; C. Yedjou; Jacqueline J. Stevens; Paul B. Tchounwou

Collaboration


Dive into the Jacqueline J. Stevens's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth Ndebele

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar

Barbara Graham

Jackson State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Yedjou

Jackson State University

View shared research outputs
Top Co-Authors

Avatar

E. Dugo

Jackson State University

View shared research outputs
Top Co-Authors

Avatar

Jennifer Sims

Jackson State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge