Network


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

Hotspot


Dive into the research topics where Elena V. Efimova is active.

Publication


Featured researches published by Elena V. Efimova.


Cancer Research | 2007

Signal Transducer and Activator of Transcription 1 Regulates Both Cytotoxic and Prosurvival Functions in Tumor Cells

Nikolai N. Khodarev; Andy J. Minn; Elena V. Efimova; Thomas E. Darga; Edwardine Labay; Michael A. Beckett; Helena J. Mauceri; Bernard Roizman; Ralph R. Weichselbaum

Elsewhere, we reported that multiple serial in vivo passage of a squamous cell carcinoma cells (SCC61) concurrent with ionizing radiation (IR) treatment resulted in the selection of radioresistant tumor (nu61) that overexpresses the signal transducer and activator of transcription 1 (Stat1)/IFN-dependent pathway. Here, we report that (a) the Stat1 pathway is induced by IR, (b) constitutive overexpression of Stat1 is linked with failure to transmit a cytotoxic signal by radiation or IFNs, (c) selection of parental cell line SCC61 against IFN-alpha and IFN-gamma leads to the same IR- and IFN-resistant phenotype as was obtained by IR selection, and (d) suppression of Stat1 by short hairpin RNA renders the IR-resistant nu61 cells radiosensitive to IR. We propose a model that transient induction of Stat1 by IFN, IR, or other stress signals activates cytotoxic genes and cytotoxic response. Constitutive overexpression of Stat1 on the other hand leads to the suppression of the cytotoxic response and induces prosurvival genes that, at high levels of Stat1, render the cells resistant to IR or other inducers of cell death.


Cancer Research | 2010

Poly(ADP-Ribose) Polymerase Inhibitor Induces Accelerated Senescence in Irradiated Breast Cancer Cells and Tumors

Elena V. Efimova; Helena J. Mauceri; Daniel W. Golden; Edwardine Labay; Vytautas P. Bindokas; Thomas E. Darga; Chaitali Chakraborty; Juan Camilo Barreto-Andrade; Clayton D. Crawley; Harold G. Sutton; Stephen J. Kron; Ralph R. Weichselbaum

Persistent DNA double-strand breaks (DSB) may determine the antitumor effects of ionizing radiation (IR) by inducing apoptosis, necrosis, mitotic catastrophe, or permanent growth arrest. IR induces rapid modification of megabase chromatin domains surrounding DSBs via poly-ADP-ribosylation, phosphorylation, acetylation, and protein assembly. The dynamics of these IR-induced foci (IRIF) have been implicated in DNA damage signaling and DNA repair. As an IRIF reporter, we tracked the relocalization of green fluorescent protein fused to a chromatin binding domain of the checkpoint adapter protein 53BP1 after IR of breast cancer cells and tumors. To block DSB repair in breast cancer cells and tumors, we targeted poly(ADP-ribose) polymerase (PARP) with ABT-888 (veliparib), one of several PARP inhibitors currently in clinical trials. PARP inhibition markedly enhanced IRIF persistence and increased breast cancer cell senescence both in vitro and in vivo, arguing for targeting IRIF resolution as a novel therapeutic strategy.


Molecular Cancer Therapeutics | 2011

Response of Human Prostate Cancer Cells and Tumors to Combining PARP Inhibition with Ionizing Radiation

Juan Camilo Barreto-Andrade; Elena V. Efimova; Helena J. Mauceri; Michael A. Beckett; Harold G. Sutton; Thomas E. Darga; Everett E. Vokes; Mitchell C. Posner; Stephen J. Kron; Ralph R. Weichselbaum

Radiation therapy remains a promising modality for curative treatment of localized prostate cancer, but dose-limiting toxicities significantly limit its effectiveness. Agents that enhance efficacy at lower radiation doses might have considerable value in increasing tumor control without compromising organ function. Here, we tested the hypothesis that the PARP inhibitor ABT-888 (veliparib) can enhance the response of prostate cancer cells and tumors to ionizing radiation (IR). Following exposure of DU-145 and PC-3 prostate cancer cell lines to the combination of 10 μmol/L ABT-888 and 6 Gy, we observed similar persistence between both cell lines of DNA damage foci and in vitro radiosensitization. We have previously observed that persistent DNA damage foci formed after ABT-888 plus IR efficiently promote accelerated cell senescence, but only PC-3 cells displayed the expected senescent response of G2–M arrest, induction of p21 and β-galactosidase expression, and accumulation as large flat cells. In turn, combining ABT-888 with 6 Gy resulted in delayed tumor regrowth compared with either agent alone only in PC-3 xenograft tumors, whereas DU-145 tumors continued to grow. By 7 days after treatment with ABT-888 plus IR, PC-3 tumors contained abundant senescent cells displaying persistent DNA damage foci, but no evidence of senescence was noted in the DU-145 tumors. That equivalent radiosensitization by ABT-888 plus IR in vitro failed to predict comparable results with tumors in vivo suggests that the efficacy of PARP inhibitors may partially depend on a competent senescence response to accumulated DNA damage. Mol Cancer Ther; 10(7); 1185–93. ©2011 AACR.


International Journal of Radiation Biology | 2009

Radioresistance of Stat1 over-expressing tumour cells is associated with suppressed apoptotic response to cytotoxic agents and increased IL6-IL8 signalling

Elena V. Efimova; Hua Liang; Sean P. Pitroda; Edwardine Labay; Thomas E. Darga; Vera Levina; Anna Lokshin; Bernard Roizman; Ralph R. Weichselbaum; Nikolai N. Khodarev

Purpose: To determine the mechanisms of Signal Transducer and Activator of Transcription 1 (Stat1)-associated radioresistance developed by nu61 tumour selected in vivo by fractionated irradiation of the parental radiosensitive tumour SCC61. Materials and methods: Radioresistence of nu61 and SCC61 in vitro was measured by clonogenic assay. Apoptotic response of nu61 and SCC61 cells to genotoxic stress was examined using caspase-based apoptotic assays. Co-cultivation of carboxyfluorescein diacetate, succinimidyl ester (CFDE-SE)-labeled nu61 with un-labeled SCC61 was performed at 1:1 ratio. Production of interleukin-6, interleukin-8 and soluble receptor of interleukin 6 (IL6, IL8 and sIL6R) was measured using Enzyme-Linked Immunosorbent Assay (ELISA). Results: Radioresistant nu61 was also resistant to interferon-gamma (IFNγ) and the death ligands of tumour necrosis factor alpha receptor (TNFR) family when compared to SCC61. This combined resistance is due to an impaired apoptotic response in nu61. Relative to SCC61, nu61 produced more IL6, IL8 and sIL6R. Using Stat1 knock-downs we demonstrated that IL6 and IL8 production is Stat1-dependent. Treatment with neutralising antibodies to IL6 and IL8, but not to either cytokine alone sensitised nu61 to genotoxic stress induced apoptosis. Conclusion: Nu61, which over-expresses Stat1 pathway, is deficient in apoptotic response to ionising radiation and cytotoxic ligands. This resistance to apoptosis is associated with Stat1-dependent production of IL6 and IL8 and suppression of 8, 9 and 3.


Oncogene | 2004

IG20 (MADD splice variant-5), a proapoptotic protein, interacts with DR4/DR5 and enhances TRAIL-induced apoptosis by increasing recruitment of FADD and caspase-8 to the DISC.

Madhu Ramaswamy; Elena V. Efimova; Osvaldo Martinez; Nirupama U Mulherkar; Surya P. Singh; Bellur S. Prabhakar

Recently, we identified Insulinoma–Glucagonoma clone 20 (IG20) that can render cells more susceptible to tumor necrosis factor-alpha (TNF-α)-induced apoptosis. In addition, it can slow cell proliferation, and enhance drug- and radiation-induced cell death. TNF-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis in some cancer cells and render others susceptible to cotreatment with drugs and irradiation, with little or no effect on most normal cells. In this study, we investigated the potential of IG20 to enhance TRAIL-induced apoptosis and found that it can render cells more susceptible to TRAIL treatment through enhanced activation of caspases. Further, we showed that this effect can be suppressed by caspase inhibitors, p35 and CrmA, and a dominant-negative Fas-associated death domain-containing protein (DN-FADD). Results from colocalization and immunoprecipitation studies showed that IG20 can interact with TRAIL death receptors (DR), DR4 and DR5 and increase recruitment of FADD and caspase-8 into the TRAIL death-inducing signaling complex (DISC). These results indicate that IG20 is a novel protein that can enhance TRAIL-induced apoptosis by facilitating DISC formation.


Molecular Therapy | 2012

Radiation-inducible Immunotherapy for Cancer: Senescent Tumor Cells as a Cancer Vaccine

Yuru Meng; Elena V. Efimova; Khaled W Hamzeh; Thomas E. Darga; Helena J. Mauceri; Yang-Xin Fu; Stephen J. Kron; Ralph R. Weichselbaum

Radiotherapy offers an effective treatment for advanced cancer but local and distant failures remain a significant challenge. Here, we treated melanoma and pancreatic carcinoma in syngeneic mice with ionizing radiation (IR) combined with the poly(ADP-ribose) polymerase inhibitor (PARPi) veliparib to inhibit DNA repair and promote accelerated senescence. Based on prior work implicating cytotoxic T lymphocytes (CTLs) as key mediators of radiation effects, we discovered that senescent tumor cells induced by radiation and veliparib express immunostimulatory cytokines to activate CTLs that mediate an effective antitumor response. When these senescent tumor cells were injected into tumor-bearing mice, an antitumor CTL response was induced which potentiated the effects of radiation, resulting in elimination of established tumors. Applied to human cancers, radiation-inducible immunotherapy may enhance radiotherapy responses to prevent local recurrence and distant metastasis.


Cancer Gene Therapy | 2009

Translational strategies exploiting TNF-|[alpha]| that sensitize tumors to radiation therapy

Helena J. Mauceri; M. A. Beckett; Hua Liang; Harold G. Sutton; Sean P. Pitroda; Eva Galka; Elena V. Efimova; Thomas E. Darga; Nikolai N. Khodarev; C R King; Mitchell C. Posner; Samuel Hellman; Donald Kufe; Ralph R. Weichselbaum

TNFerade is a radioinducible adenoviral vector expressing tumor necrosis factor-α (TNF-α) (Ad.Egr-TNF) currently in a phase III trial for inoperable pancreatic cancer. We studied B16-F1 melanoma tumors in TNF receptor wild-type (C57BL/6) and deficient (TNFR1,2−/− and TNFR1−/−) mice. Ad.Egr-TNF+IR inhibited tumor growth compared with IR in C57BL/6 but not in receptor-deficient mice. Tumors resistant to TNF-α were also sensitive to Ad.Egr-TNF+IR in C57BL/6 mice. Ad.Egr-TNF+IR produced an increase in tumor-associated endothelial cell apoptosis not observed in receptor-deficient animals. Also, B16-F1 tumors in mice with germline deletions of TNFR1,2, TNFR1 or TNF-α, or in mice receiving anti-TNF-α exhibited radiosensitivity. These results show that tumor-associated endothelium is the principal target for Ad.Egr-TNF radiosensitization and implicate TNF-α signaling in tumor radiosensitivity.


Journal of Cellular Biochemistry | 2013

Imaging UVC‐induced DNA damage response in models of minimal cancer

Shinji Miwa; Shuya Yano; Yukihiko Hiroshima; Yasunori Tome; Fuminari Uehara; Sumiyuki Mii; Elena V. Efimova; Hiroaki Kimura; Katsuhiro Hayashi; Hiroyuki Tsuchiya; Robert M. Hoffman

We have previously demonstrated that the ultraviolet (UV) light is effective against a variety of cancer cells in vivo as well as in vitro. In the present report, we imaged the DNA damage repair response of minimal cancer after UVC irradiation. DNA‐damage repair response to UV irradiation was imaged on tumors growing in 3D culture and in superficial tumors grown in vivo. UV‐induced DNA damage repair was imaged with GFP fused to the DNA damage response (DDR)‐related chromatin‐binding protein 53BP1 in MiaPaCa‐2 human pancreatic cancer cells. Three‐dimensional Gelfoam® histocultures and confocal imaging enabled 53BP1‐GFP nuclear foci to be observed within 1 h after UVC irradiation, indicating the onset of DNA damage repair response. A clonogenic assay showed that UVC inhibited MiaPaCa‐2 cell proliferation in a dose‐dependent manner, while UVA and UVB showed little effect on cell proliferation. Induction of UV‐induced 53BP1‐GFP focus formation was limited up to a depth of 40 µm in 3D‐culture of MiaPaCa‐2 cells. The MiaPaCa‐2 cells irradiated by UVC light in a skin‐flap mouse model had a significant decrease of tumor growth compared to untreated controls. Our results also demonstrate that 53BP1‐GFP is an imageable marker of UV‐induced DNA damage repair response of minimal cancer and that UVC is a useful tool for the treatment of residual cancer since UVC can kill superficial cancer cells without damage to deep tissue. J. Cell. Biochem. 114: 2493–2499, 2013.


Journal of Cellular Biochemistry | 2013

Dynamic color‐coded fluorescence imaging of the cell‐cycle phase, mitosis, and apoptosis demonstrates how caffeine modulates cisplatinum efficacy

Shinji Miwa; Shuya Yano; Yasunori Tome; Naotoshi Sugimoto; Yukihiko Hiroshima; Fuminari Uehara; Sumiyuki Mii; Hiroaki Kimura; Katsuhiro Hayashi; Elena V. Efimova; Toshiyoshi Fujiwara; Hiroyuki Tsuchiya; Robert M. Hoffman

Caffeine enhances the effect of certain anticancer drugs, but the mechanism of modulation is poorly understood. In this study, modulation of cisplatinum efficacy induced by caffeine was visualized at the subcellular level by real‐time fluorescent‐protein imaging. Mitotic and apoptotic changes were observed by imaging 143B human osteosarcoma dual‐color cells, in which GFP is expressed in the nucleus and RFP is expressed in the cytoplasm. Modulation of the cell cycle was imaged using time‐lapse imaging of HeLa cells expressing a fluorescent ubiquitination‐based cell cycle indicator (FUCCI) in the nucleus. Clonogenic assays showed that caffeine increased the inhibition by cisplatinum on cell proliferation. Subcellular imaging demonstrated that cisplatinum decreased mitosis and induced apoptosis in 143B cells. The combination of cisplatinum and caffeine enhanced mitosis and subsequently increased apoptosis. Time‐lapse imaging showed that cisplatinum strongly induced cell‐cycle arrest in the S/G2 phase in HeLa‐FUCCI cells. Caffeine overcame the cell‐cycle arrest induced by cisplatinum, thereby increasing its efficacy, since cisplatinum is ineffective against quiescent cells. The data in this report indicate that caffeine modulates the cell cycle in cancer cells, thereby enhancing efficacy of cell‐cycle‐dependent anticancer drugs such as cisplatinum. J. Cell. Biochem. 114: 2454–2460, 2013.


Molecular Cancer Research | 2016

Linking Cancer Metabolism to DNA Repair and Accelerated Senescence

Elena V. Efimova; Satoe Takahashi; Noumaan A. Shamsi; Ding Wu; Edwardine Labay; Olesya A. Ulanovskaya; Ralph R. Weichselbaum; Sergey A. Kozmin; Stephen J. Kron

Conventional wisdom ascribes metabolic reprogramming in cancer to meeting increased demands for intermediates to support rapid proliferation. Prior models have proposed benefits toward cell survival, immortality, and stress resistance, although the recent discovery of oncometabolites has shifted attention to chromatin targets affecting gene expression. To explore further effects of cancer metabolism and epigenetic deregulation, DNA repair kinetics were examined in cells treated with metabolic intermediates, oncometabolites, and/or metabolic inhibitors by tracking resolution of double-strand breaks (DSB) in irradiated MCF7 breast cancer cells. Disrupting cancer metabolism revealed roles for both glycolysis and glutaminolysis in promoting DSB repair and preventing accelerated senescence after irradiation. Targeting pathways common to glycolysis and glutaminolysis uncovered opposing effects of the hexosamine biosynthetic pathway (HBP) and tricarboxylic acid (TCA) cycle. Treating cells with the HBP metabolite N-acetylglucosamine (GlcNAc) or augmenting protein O-GlcNAcylation with small molecules or RNAi targeting O-GlcNAcase each enhanced DSB repair, while targeting O-GlcNAc transferase reversed GlcNAcs effects. Opposing the HBP, TCA metabolites including α-ketoglutarate blocked DSB resolution. Strikingly, DNA repair could be restored by the oncometabolite 2-hydroxyglutarate (2-HG). Targeting downstream effectors of histone methylation and demethylation implicated the PRC1/2 polycomb complexes as the ultimate targets for metabolic regulation, reflecting known roles for Polycomb group proteins in nonhomologous end-joining DSB repair. Our findings that epigenetic effects of cancer metabolic reprogramming may promote DNA repair provide a molecular mechanism by which deregulation of metabolism may not only support cell growth but also maintain cell immortality, drive therapeutic resistance, and promote genomic instability. Implications: By defining a pathway from deregulated metabolism to enhanced DNA damage response in cancer, these data provide a rationale for targeting downstream epigenetic effects of metabolic reprogramming to block cancer cell immortality and overcome resistance to genotoxic stress. Mol Cancer Res; 14(2); 173–84. ©2015 AACR.

Collaboration


Dive into the Elena V. Efimova'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

Yasunori Tome

University of the Ryukyus

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge