Network


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

Hotspot


Dive into the research topics where Demetrius M. Kokkinakis is active.

Publication


Featured researches published by Demetrius M. Kokkinakis.


Cancer Research | 2004

Modulation of gene expression in human central nervous system tumors under methionine deprivation-induced stress.

Demetrius M. Kokkinakis; Xiaoyan Liu; Sunil Chada; Mansoor M. Ahmed; Mohammed M. Shareef; Ujjal Singha; Sutin Yang; Jian-Hua Luo

Methionine deprivation imposes a metabolic stress, termed methionine stress, that inhibits mitosis and induces cell cycle arrest and apoptosis. The methionine-dependent central nervous system tumor cell lines DAOY (medulloblastoma), SWB61 (anaplastic oligodendroglioma), SWB40 (anaplastic astrocytoma), and SWB39 (glioblastoma multiforme) were compared with methionine-stress resistant SWB77 (glioblastoma multiforme). The cDNA-oligoarray analysis and reverse transcription-PCR verification indicated common changes in gene expression in methionine-dependent cell lines to include up-regulation/induction of cyclin D1, mitotic arrest deficient (MAD)1, p21, growth arrest and DNA-damage-inducible (GADD)45 α, GADD45 γ, GADD34, breast cancer (BRCA)1, 14-3-3σ, B-cell CLL/lymphoma (BCL)1, transforming growth factor (TGF)-β, TGF-β–induced early response (TIEG), SMAD5, SMAD7, SMAD2, insulin-like growth factor binding protein (IGFBP7), IGF-R2, vascular endothelial growth factor (VEGF), TNF-related apoptosis-inducing ligand (TRAIL), TNF-α converting enzyme (TACE), TRAIL receptor (TRAIL-R)2, TNFR-related death receptor (DR)6, TRAF interacting protein (I-TRAF), IL-6, MDA7, IL-1B convertase (ICE)-γ, δ and ε, IRF1, IRF5, IRF7, interferon (IFN)-γ and receptor components, ISG15, p65-NF-κB, JUN-B, positive cofactor (PC)4, C/ERB-β, inositol triphosphate receptor I, and methionine adenosyltransferase II. On the other hand, cyclins A1, A2, B1 and B2, cell division cycle (CDC)2 and its kinase, CDC25 A and B, budding uninhibited by benzimidazoles (BUB)1 and 3, MAD2, CDC28 protein kinase (CKS)1 and 2, neuroepithelial cell transforming gene (NET)1, activator of S-phase kinase (ASK), CDC14B phosphatase, BCL2, TGF-β activated kinase (TAK)1, TAB1, c-FOS, DNA topoisomerase II, DNA polymerase α, dihydrofolate reductase, thymidine kinase, stathmin, and MAP4 were down-regulated. In the methionine stress-resistant SWB77, only 20% of the above genes were affected, and then only to a lesser extent. In addition, some of the changes observed in SWB77 were opposite to those seen in methionine-dependent tumors, including expression of p21, TRAIL-R2, and TIEG. Despite similarities, differences between methionine-dependent tumors were substantial, especially in regard to regulation of cytokine expression. Western blot analysis confirmed that methionine stress caused the following: (a) a marked increase of GADD45α and γ in the wt-p53 cell lines SWB61 and 40; (b) an increase in GADD34 and p21 protein in all of the methionine-dependent lines; and (c) the induction of MDA7 and phospho-p38 in DAOY and SWB39, consistent with marked transcriptional activation of the former under methionine stress. It was additionally shown that methionine stress down-regulated the highly active phosphatidylinositol 3′-kinase pathway by reducing AKT phosphorylation, especially in DAOY and SWB77, and also reduced the levels of retinoblastoma (Rb) and pRb (P-ser780, P-ser795, and P-ser807/811), resulting in a shift in favor of unphosphorylated species in all of the methionine-dependent lines. Immunohistochemical analysis showed marked inhibition of nuclear translocation of nuclear factor κB under methionine stress in methionine-dependent lines. In this study we show for the first time that methionine stress mobilizes several defined cell cycle checkpoints and proapoptotic pathways while coordinately inhibiting prosurvival mechanisms in central nervous system tumors. It is clear that methionine stress-induced cytotoxicity is not restricted by the p53 mutational status.


Molecular Cancer Research | 2006

Mitotic Arrest, Apoptosis, and Sensitization to Chemotherapy of Melanomas by Methionine Deprivation Stress

Demetrius M. Kokkinakis; Anthony G. Brickner; John M. Kirkwood; Xiaoyan Liu; Jason E. Goldwasser; Anastasiya Kastrama; Cindy Sander; Dora Bocangel; Sunil Chada

Methionine deprivation stress (MDS) eliminates mitotic activity in melanoma cells regardless of stage, grade, or TP53 status, whereas it has a negligible effect on normal skin fibroblasts. In most cases, apoptosis accounts for the elimination of up to 90% of tumor cells from the culture within 72 hours after MDS, leaving a scattered population of multinucleated resistant cells. Loss of mitosis in tumor cells is associated with marked reduction of cyclin-dependent kinase (CDK) 1 transcription and/or loss of its active form (CDK1-P-Thr161), which is coincident with up-regulation of CDKN1A, CDKN1B, and CDKN1C (p21, p27, and p57). Expression of the proapoptotic LITAF, IFNGR, EREG, TNFSF/TNFRSF10 and TNFRSF12, FAS, and RNASEL is primarily up-regulated/induced in cells destined to undergo apoptosis. Loss of Aurora kinase B and BIRC5, which are required for histone H3 phosphorylation, is associated with the accumulation of surviving multinucleated cells. Nevertheless, noncycling survivors of MDS are sensitized to temozolomide, carmustin, and cisplatin to a much greater extent than normal skin fibroblasts possibly because of the suppression of MGMT/TOP1/POLB, MGMT/RAD52/RAD54, and cMET/RADD52, respectively. Sensitivity to these and additional genotoxic agents and radiation may also be acquired due to loss of cMET/OGG1, reduced glutathione reductase levels, and a G2-phase block that is a crucial step in the damage response associated with enhancement of drug toxicity. Although the genes controlling mitotic arrest and/or apoptosis in response to low extracellular methionine levels are unknown, it is likely that such control is exerted via the induction/up-regulation of tumor suppressors/growth inhibitor genes, such as TGFB, PTEN, GAS1, EGR3, BTG3, MDA7, and the proteoglycans (LUM, BGN, and DCN), as well as the down-regulation/loss of function of prosurvival genes, such as NFκB, MYC, and ERBB2. Although MDS targets several common genes in tumors, mutational variability among melanomas may decide which metabolic and signal transduction pathways will be activated or shutdown. (Mol Cancer Res 2006;4(8):575–89)


Chemico-Biological Interactions | 1991

Differences between pancreatropic nitrosamine carcinogens and N-nitrosodimethylamine in methylating DNA in various tissues of hamsters and rats.

Demetrius M. Kokkinakis

N-Nitrosobis(2-oxopropyl)amine (BOP) and N-nitroso(2-hydroxypropyl)(2-oxypropyl)amine (HPOP) induce pancreatic tumors in the Syrian hamster. BOP and HPOP target the kidneys, esophagus and upper respiratory system in rats, but the pancreas of this species is resistant to the above carcinogens. On the other hand, N-nitrosodimethylamine (DMN) induces hepatic and kidney tumors in the rat, and tumors of the liver and upper respiratory system in the hamster, but it is not known to affect the pancreas of either species. At equimolar doses, ratios of DMN versus BOP or HPOP mediated methylation in hamster liver DNA are 1.6 and 8.1, respectively. Respective ratios in the rat liver are 1.1 and 6.5. However, in both species equitoxic doses of BOP, HPOP and DMN induce similar levels of N7-methylguanine (N7-MeG) in hepatic DNA. At such doses methylation of kidney DNA is 24 and 14 times more extensive in BOP and HPOP than in DMN-treated hamsters. Similarly, ratios of N7-MeG in the pancreas of BOP and HPOP vs. DMN-treated hamsters are 10 and 5, respectively, while in the lung this ratio is 2.2 for both carcinogens. Levels of O6-methylguanine (O6-MeG) in the DNA of extrahepatic tissues are substantially greater in hamsters treated with BOP or HPOP than in those treated with an equitoxic dose of DMN. In rats, equitoxic doses of BOP and DMN induce similar levels of N7-MeG and O6-MeG in hepatic, kidney and lung DNA. However, levels of these adducts in pancreatic DNA are 2 times greater following BOP than DMN administration. Ratios of N7-MeG in pancreas, lung and kidney in HPOP vs. DMN-treated rats are 2.1, 2.7 and 2.1, respectively. Repair of O6-MeG is more effective in rat than in hamster liver, however in other tissues this is not always the case. Levels of O6-MeG in the pancreas of rats are reduced to half of their initial value between 40 and 50 h following the administration of 10, 50 or 20 mg/kg DMN, HPOP or BOP, respectively. However, half-lives for the repair of O6-MeG in hamster pancreas are 28, 62 and greater than 120 h at the respective doses of the above carcinogens. Since the above doses of DMN, HPOP and BOP induce 7, 19 and 41 nmol O6-MeG/mmol of guanine respectively in the hamster pancreas, it is suggested that the rate of repair could be a function of the initial concentration of this adduct. Differences between DMN and BOP or HPOP in methylating pancreatic DNA are sufficient to distinguish the latter two nitrosamines as pancreatic carcinogens for the hamster.


Cancer Biology & Therapy | 2004

Potential use of MDA-7 as a biological adjuvant for radiation therapy

Mohammed M. Shareef; Demetrius M. Kokkinakis; Mansoor M. Ahmed

Commentary to: MDA-7 Regulates Cell Growth and Radiosensitivity In Vitro of Primary (Non-Established) Human Glioma Cells A. Yacoub, C. Mitchell, Y. Hong, R.V. Gopalkrishnan, Z.-Z. Su, P. Gupta, M. Sauane, I.V. Lebedeva, D.T. Curiel, P.J. Mahasreshti, M.R. Rosenfeld, W.C. Broaddus, C.D. James, S.Grant, P.B. Fisher and P. Dent


Carcinogenesis | 2003

Allyl isothiocyanate, a constituent of cruciferous vegetables, inhibits growth of PC-3 human prostate cancer xenografts in vivo.

Sanjay K. Srivastava; Dong Xiao; Karen L. Lew; Pamela A. Hershberger; Demetrius M. Kokkinakis; Candace S. Johnson; Donald L. Trump


Clinical Cancer Research | 2001

Thresholds of O6-Alkylguanine-DNA Alkyltransferase which Confer Significant Resistance of Human Glial Tumor Xenografts to Treatment with 1,3-Bis(2-chloroethyl)-1-nitrosourea or Temozolomide

Demetrius M. Kokkinakis; Dora Bocangel; S. Clifford Schold; Robert C. Moschel; Anthony E. Pegg


Cancer Research | 1997

Role of O6-Methylguanine-DNA Methyltransferase in the Resistance of Pancreatic Tumors to DNA Alkylating Agents

Demetrius M. Kokkinakis; Mansoor M. Ahmed; Ruby Delgado; Mushdaq M. Fruitwala; Mohammed Mohiuddin; Jorge Albores-Saavedra


Carcinogenesis | 2004

High throughput screening of methylation status of genes in prostate cancer using an oligonucleotide methylation array

Yan Ping Yu; Shirish Paranjpe; Joel B. Nelson; Sydney D. Finkelstein; Baoguo Ren; Demetrius M. Kokkinakis; George K. Michalopoulos; Jian-Hua Luo


Cancer Research | 1996

Treatment of Human Brain Tumor Xenografts with O6-Benzyl-2′-deoxyguanosine and BCNU

S. Clifford Schold; Demetrius M. Kokkinakis; Jack L. Rudy; Robert C. Moschel; Anthony E. Pegg


Cancer Research | 1989

DNA Alkylation in the Hamster Induced by Two Pancreatic Carcinogens

Demetrius M. Kokkinakis; Dante G. Scarpelli

Collaboration


Dive into the Demetrius M. Kokkinakis's collaboration.

Top Co-Authors

Avatar

Anthony E. Pegg

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mansoor M. Ahmed

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

R. C. Moschel

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Robert C. Moschel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dora Bocangel

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian-Hua Luo

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge