Network


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

Hotspot


Dive into the research topics where Elaine Gavin is active.

Publication


Featured researches published by Elaine Gavin.


Oncogene | 2009

Mechanism of chemoresistance mediated by miR-140 in human osteosarcoma and colon cancer cells

Bo Song; Yuan Wang; Yaguang Xi; Kenji Kudo; Skjalg Bruheim; Galina I. Botchkina; Elaine Gavin; Yu Wan; Andrea Formentini; Marko Kornmann; Øystein Fodstad; Jingfang Ju

In this study, high-throughput microRNA (miRNA) expression analysis revealed that the expression of miR-140 was associated with chemosensitivity in osteosarcoma tumor xenografts. Tumor cells ectopically transfected with miR-140 were more resistant to methotrexate and 5-fluorouracil (5-FU). Overexpression of miR-140 inhibited cell proliferation in both osteosarcoma U-2 OS (wt-p53) and colon cancer HCT 116 (wt-p53) cell lines, but less so in osteosarcoma MG63 (mut-p53) and colon cancer HCT 116 (null-p53) cell lines. miR-140 induced p53 and p21 expression accompanied with G1 and G2 phase arrest only in cell lines containing wild type of p53. Histone deacetylase 4 (HDAC4) was confirmed to be one of the important targets of miR-140. The expression of endogenous miR-140 was significantly elevated in CD133+hiCD44+hi colon cancer stem-like cells that exhibit slow proliferating rate and chemoresistance. Blocking endogenous miR-140 by locked nucleic acid-modified anti-miR partially sensitized resistant colon cancer stem-like cells to 5-FU treatment. Taken together, our findings indicate that miR-140 is involved in the chemoresistance by reduced cell proliferation through G1 and G2 phase arrest mediated in part through the suppression of HDAC4. miR-140 may be a candidate target to develop novel therapeutic strategy to overcome drug resistance.


Clinical Cancer Research | 2008

miR-192 Regulates dihydrofolate reductase and cellular proliferation through the p53-microRNA circuit.

Bo Song; Yuan Wang; Kenji Kudo; Elaine Gavin; Yaguang Xi; Jingfang Ju

Purpose: The purpose of this study is to investigate the molecular mechanism of miR-192 in colon cancer. Experimental Design: Human colon cancer cell lines with different p53 status were used as our model system to study the effect of miR-192 on cell proliferation, cell cycle control, and mechanism of regulation. Results: Our results show that one of the key miR-192 target genes is dihydrofolate reductase (DHFR). miR-192 affects cellular proliferation through the p53-miRNA circuit. Western immunoblot analyses indicated that the expression of DHFR was significantly decreased by miR-192. Further investigation revealed that such suppression was due to translational arrest rather than mRNA degradation. More profound inhibition of cellular proliferation was observed by ectopic expression of miR-192 in colon cancer cell lines containing wild-type p53 than cells containing mutant p53. Thus, the effect of miR-192 on cellular proliferation is mainly p53 dependent. Overexpression of miR-192 triggered both G1 and G2 arrest in HCT-116 (wt-p53) cells but not in HCT-116 (null-p53) cells. The cell cycle checkpoint control genes p53 and p21 were highly overexpressed in cells that overexpressed miR-192. Endogenous miR-192 expression was increased in HCT-116 (wt-p53) and RKO (wt-p53) cells treated with methotrexate, which caused an induction of p53 expression. Chromatin immunoprecipitation-quantitative reverse transcription-PCR analysis revealed that the p53 protein interacted with the miR-192 promoter sequence. Conclusion: These results indicate that miR-192 may be another miRNA candidate that is involved in the p53 tumor suppressor network with significant effect on cell cycle control and cell proliferation.


Oncogene | 2012

Inhibition of Gli1 results in altered c-Jun activation, inhibition of cisplatin-induced upregulation of ERCC1, XPD and XRCC1, and inhibition of platinum–DNA adduct repair

Kenji Kudo; Elaine Gavin; Shamik Das; L Amable; Lalita A. Shevde; Eddie Reed

The transcription of ERCC1 and other nucleotide excision repair (NER) genes is strongly influenced by c-jun. C-jun is transcriptionally regulated by Gli proteins of the Hedgehog pathway. We therefore studied the possible relationships between Gli1, c-jun, and the upregulation of ERCC1, XPD and XRCC1 in cisplatin-resistant human ovarian cancer cells. We studied the paired human ovarian cancer cell lines A2780 and A2780-CP70. We used a shRNA construct that specifically degrades Gli1 message. Genes we assessed for mRNA and/or protein levels included: c-jun, ERCC1, XPD, XRCC1, Gli1, Gli2, SHH, IHH, GAPDH and α-tubulin. Platinum–DNA adduct repair was assessed by atomic absorbance spectrometry with Zeeman background correction. Use of the anti-Gli1 shRNA in cisplatin-resistant cells resulted in a block of the cells ability to upregulate genes in response to cisplatin treatment, including: c-jun, ERCC1, XPD and XRCC1. This block in upregulation of c-jun was concurrent with a change in the phosphorylation pattern of the c-jun protein, shifting that pattern from a Ser63/73 dominant pattern, to a Thr91/93 dominant pattern. A2780-CP70 cells were treated at their cisplatin IC50, and DNA repair was assessed after pretreatment with anti-Gli1 shRNA or scrambled shRNA control. Control cells repaired 78% of platinum–DNA adducts at 12 h, compared with 33% repair in cells pretreated with anti-Gli1 shRNA resulting in a 2.4-fold difference. Pretreatment of A2780-CP70 cells with anti-Gli1 shRNA resulted in supra-additive cell killing with cisplatin; shifting the cisplatin IC50 (half maximal inhibitory concentration) from 30 μM to 5 μM. Pretreatment of these cells with cyclopamine did not shift the cisplatin IC50. We conclude that the transcriptional protein Gli1 is important in the upregulation of these three DNA repair genes in human ovarian cancer cells, and that Gli1 strongly influences platinum–DNA adduct repair, and cellular sensitivity to cisplatin. This Gli1 role has c-jun as an intermediate in the pathway. In all, inhibition of Gli1 by a specific shRNA inhibits the upregulation of c-jun Ser63/73, and also inhibits the upregulation of three genes essential to NER (ERCC1, XPD) and base excision repair (XRCC1).


PLOS ONE | 2008

Reduction of Orc6 Expression Sensitizes Human Colon Cancer Cells to 5-Fluorouracil and Cisplatin

Elaine Gavin; Bo Song; Yuan Wang; Yaguang Xi; Jingfang Ju

Previous studies from our group have shown that the expression levels of Orc6 were highly elevated in colorectal cancer patient specimens and the induction of Orc6 was associated with 5-fluorouracil (5-FU) treatment. The goal of this study was to investigate the molecular and cellular impact of Orc6 in colon cancer. In this study, we use HCT116 (wt-p53) and HCT116 (null-p53) colon cancer cell lines as a model system to investigate the impact of Orc6 on cell proliferation, chemosensitivity and pathways involved with Orc6. We demonstrated that the down regulation of Orc6 sensitizes colon cancer cells to both 5-FU and cisplatin (cis-pt) treatment. Decreased Orc6 expression in HCT-116 (wt-p53) cells by RNA interference triggered cell cycle arrest at G1 phase. Prolonged inhibition of Orc6 expression resulted in multinucleated cells in HCT-116 (wt-p53) cell line. Western immunoblot analysis showed that down regulation of Orc6 induced p21 expression in HCT-116 (wt-p53) cells. The induction of p21 was mediated by increased level of phosphorylated p53 at ser-15. By contrast, there is no elevated expression of p21 in HCT-116 (null-p53) cells. Orc6 down regulation also increased the expression of DNA damaging repair protein GADD45β and reduced the expression level of JNK1. Orc6 may be a potential novel target for future anti cancer therapeutic development in colon cancer.


Oncology Reports | 2014

Gli1 contributes to cellular resistance to cisplatin through altered cellular accumulation of the drug

Lauren Amable; Jason Fain; Elaine Gavin; Eddie Reed

Cellular resistance to platinum anticancer compounds is governed by no less than two molecular processes; DNA repair and cellular accumulation of drug. Gli1 is an upstream regulator of nucleotide excision repair, effecting this process through c-jun. We, therefore, investigated whether Gli1 plays a role in cellular accumulation of cisplatin. Using a Gli1-specific shRNA, we explored the role of Gli1 in the cellular accumulation and efflux of cisplatin, in cisplatin-resistant A2780-CP70 human ovarian cancer cells. When Gli1 is inhibited, cellular uptake of cisplatin was approximately 33% of the level of uptake under control conditions. When Gli1 is inhibited, cellular efflux of cisplatin was completely abrogated, over a 12-h period of observation. We assayed nuclear lysates from these cells, for the ability to bind the DNA sequence that is the Gli-binding site (GBS) in the 5′UTR for each of five known cisplatin transmembrane transporters. Four of these transporters are active in cisplatin uptake; and, one is active in cisplatin efflux. In each case, nuclear lysate from A2780-CP70 cells binds the GBS of the respective cisplatin transport gene. We conclude that Gli1 plays a strong role in total cellular accumulation of cisplatin in these cells; and, that the combined effects on cellular accumulation of drug and on DNA repair may indicate a role for Gli1 in protecting cellular DNA from lethal types of DNA damage.


International Journal of Oncology | 2014

GLI1 upregulates C-JUN through a specific 130-kDa isoform

Lauren Amable; Elaine Gavin; Kenji Kudo; Erhong Meng; Rodney P. Rocconi; Lalita A. Shevde; Eddie Reed

The Hedgehog pathway is molecularly linked to increased resistance to cisplatin and increased repair of platinum-DNA damage, through C-JUN. GLI1, which has five known isoforms, is a positive transcriptional regulator in Hedgehog. Southwestern blot assay, EMSA and ChIP assays indicate that only one of five isoforms of GLI1 may be responsible for the Hedgehog link with C-JUN and thus, increased platinum-DNA adduct repair. Cancer tissues express this 130-kDa isoform at levels 6-fold higher than non-malignant tissues; and this isoform exists in abundance in six of seven ovarian cancer cell lines examined.


Molecular Cancer Research | 2017

EGFR Mutations Compromise Hypoxia-Associated Radiation Resistance through Impaired Replication Fork–Associated DNA Damage Repair

Mohammad Saki; Haruhiko Makino; Prashanthi Javvadi; Nozomi Tomimatsu; Lianghao Ding; Jennifer E. Clark; Elaine Gavin; Kenichi Takeda; Joel Andrews; Debabrata Saha; Michael D. Story; Sandeep Burma; Chaitanya S. Nirodi

EGFR signaling has been implicated in hypoxia-associated resistance to radiation or chemotherapy. Non–small cell lung carcinomas (NSCLC) with activating L858R or ΔE746-E750 EGFR mutations exhibit elevated EGFR activity and downstream signaling. Here, relative to wild-type (WT) EGFR, mutant (MT) EGFR expression significantly increases radiosensitivity in hypoxic cells. Gene expression profiling in human bronchial epithelial cells (HBEC) revealed that MT-EGFR expression elevated transcripts related to cell cycle and replication in aerobic and hypoxic conditions and downregulated RAD50, a critical component of nonhomologous end joining and homologous recombination DNA repair pathways. NSCLCs and HBEC with MT-EGFR revealed elevated basal and hypoxia-induced γ-H2AX–associated DNA lesions that were coincident with replication protein A in the S-phase nuclei. DNA fiber analysis showed that, relative to WT-EGFR, MT-EGFR NSCLCs harbored significantly higher levels of stalled replication forks and decreased fork velocities in aerobic and hypoxic conditions. EGFR blockade by cetuximab significantly increased radiosensitivity in hypoxic cells, recapitulating MT-EGFR expression and closely resembling synthetic lethality of PARP inhibition. Implications: This study demonstrates that within an altered DNA damage response of hypoxic NSCLC cells, mutant EGFR expression, or EGFR blockade by cetuximab exerts a synthetic lethality effect and significantly compromises radiation resistance in hypoxic tumor cells. Mol Cancer Res; 15(11); 1503–16. ©2017 AACR.


Cancer Research | 2017

Abstract 5174: Phosphodiesterase 10A inhibition as a novel approach to suppress β-catenin signaling in ovarian cancer cells

Luciana Madeira da Silva; Elaine Gavin; Kevin Lee; Veronica Ramirez-Alcantara; Kristy L. Berry; Holly Taylor; Alla Musiyenko; Ileana V. Aragon; Adam B. Keeton; Jennifer Scalici; Rodney P. Rocconi; Gary A. Piazza

Canonical Wnt/β-catenin signaling is known to be associated with platinum resistance in ovarian cancer in which inhibitors hold promise for the treatment of refractory disease. Phosphodiesterase 10A (PDE10A) is a dual cyclic AMP and cyclic GMP phosphodiesterase isozyme recently implicated in colon cancer. PDE10A inhibition in colon cancer cells by siRNA or small molecule inhibitors increased cGMP levels and activated PKG to inhibit β-catenin signaling. A novel PDE10 inhibitor, ADT-061, was identified by screening a library of indene derivatives, and showed strong antineoplastic activity in the Apc+/min-FCCC mouse (Lee K et al., unpublished data). Cyclic GMP and phosphodiesterases participate in the ovarian follicular development, although little is known about PDE10A expression in ovaries, especially with regard to a potential role in ovarian tumorigenesis. PDE10A protein was found to be expressed in various established ovarian cancer cell lines at higher levels than immortalized or primary ovarian surface epithelial cell lines. Pf-2545920, a known PDE10A inhibitor, and ADT-061 inhibited the growth of multiple ovarian tumor cell lines with IC50s around 20µM and 0.5µM, respectively. Both compounds induced apoptosis after 24h treatment, as measured by PI/Annexin-V staining and PARP cleavage. Pf-2545920 and ADT-061 induced phosphorylation of VASP at Ser157 and Ser239 in various ovarian cancer cell lines, indicating activation of cyclic AMP and cyclic GMP signaling, respectively. Treatment also decreased levels of β-catenin and downstream targets of TCF-dependent transcription, including c-MYC, survivin and cyclin-D1. Homozygous knockout PDE10A clones of OV-90 ovarian cancer cells obtained using CRISPR/Cas9 showed decreased clonogenic potential, decreased Pf-2545920-mediated VASP phosphorylation and β-catenin, c-MYC and survivin expression. Ongoing efforts are focused on the development of more potent ADT-061 analogs. These observations support further study of a role of PDE10 in ovarian tumorigenesis and the development of ADT-061 or analogs for the treatment of refractory ovarian cancer as well as the prevention of malignant recurrence. Citation Format: Luciana Madeira Da Silva, Elaine Gavin, Kevin J. Lee, Veronica Ramirez-Alcantara, Kristy L. Berry, Holly T. Taylor, Alla Musiyenko, Ileana V. Aragon, Adam B. Keeton, Jennifer Scalici, Rodney P. Rocconi, Gary A. Piazza. Phosphodiesterase 10A inhibition as a novel approach to suppress β-catenin signaling 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 5174. doi:10.1158/1538-7445.AM2017-5174


Clinical Cancer Research | 2016

Abstract A76: Targeting phosphodiesterase 10A for chemoprevention and treatment of ovarian cancer.

Luciana Madeira da Silva; Elaine Gavin; Kevin Lee; Ileana V. Aragon; Veronica Ramirez-Alcantara; J.M. Scalici; Rodney P. Rocconi; Gary A. Piazza

Phosphodiesterase 10A (PDE10A) is a dual cyclic AMP and cyclic GMP phosphodiesterase that is highly expressed in the brain striatum, testis and thyroid and this restricted expression in peripheral tissue has been clinically exploited for the treatment of schizophrenia and Huntington9s disease. Interestingly, our group has recently found that PDE10A is overexpressed in colon, lung and breast tumors when compared to their healthy tissue correlates. Moreover, our studies indicate that PDE10A inhibition in cancer cells by RNA interference or by small molecule inhibitors leads to activation of cGMP/PKG and inhibition of β-catenin signaling pathway that culminates with apoptotic cell death. Here we are investigating the role of PDE10A in ovarian cancer by using genetic approaches (RNA interference and CRISPR/Cas9 genome editing) and novel PDE10A small molecule inhibitors that we generated. We found that PDE10A protein is expressed in various established ovarian cancer cell lines at higher levels than in immortalized ovarian surface epithelial cell lines. Using a known PDE10A inhibitor, Pf-2545920, and novel sulindac derivatives that we generated with high specificity towards the PDE10A isozyme, we observed potent growth inhibition properties (IC50s at low micromolar or sub-micromolar concentrations) for all ovarian cancer cell lines tested. One of our novel PDE10A inhibitors, called MCI-030, which showed IC50~0.5μM for ovarian cancer cells, was orally administered to C57BL/6 mice at a safe dose of 100mg/Kg and revealed a stable concentration of approximately 2μM in the plasma and ovarian tissue for about 8h. Mechanistically, our results showed that PDE10A inhibitors induced apoptosis as measured by PI/Annexin V staining. Pf-2545920 induced phosphorylation of VASP at Ser157 and Ser239 in OV-90, OVCAR3 and SKOV3 cells, indicating activation of cyclic nucleotide signaling pathway. In addition, a decrease in the levels of β-catenin, survivin, c-Myc and cyclin D1 was observed in OV-90 cells treated with Pf-2545920 for 24h. We used CRISPR-Cas9 to delete a 97bp segment in exon 7 of PDE10A in the diploid ovarian cancer cell line OV-90. Homozygous knockout PDE10A clones were obtained and showed decreased clonogenic potential and decreased Pf-2545920-mediated pVASP phosphorylation in vitro. We are currently investigating the expression of PDE10A in clinical specimens of ovarian tumors by qPCR, western-blotting and immunohistochemistry. Our future work includes testing MCI-030 in vivo using xenograft intraperitoneal models of ovarian cancer, as well as in vivo experiments with OV-90 PDE10 knockout cells. Our ultimate goal for the proposed studies of PDE10 as a novel chemoprevention target for ovarian cancer is to develop a potent PDE10 inhibitor with negligible toxicity that can be used as a chemoprevention agent for women at high risk for this malignancy. Citation Format: Luciana Madeira da Silva, Elaine Gavin, Kevin Lee, Ileana Aragon, Veronica Ramirez-Alcantara, Jennifer Scalici, Rodney P. Rocconi, Gary A. Piazza. Targeting phosphodiesterase 10A for chemoprevention and treatment of ovarian cancer. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: Exploiting Vulnerabilities; Oct 17-20, 2015; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(2 Suppl):Abstract nr A76.


Cancer Research | 2013

Abstract LB-223: A specific Isoform of Gli1 binds the promoter region of c-fos in A2780-CP70 human ovarian cancer cells.

Lauren Amable; Kenji Kudo; Elaine Gavin; Jason Fain; Eddie Reed

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The transcription factor Activator Protein 1 (AP1), is an important regulator in cisplatin resistance. AP1 is comprised of the two proteins, c-jun and c-fos. The transcription factor Gli1, a member of the hedgehog signaling pathway, is also an important factor in cisplatin resistance and transcriptionally regulated c-jun. The c-jun interface with Gli1 is mediated by a 130 kDa isoform of Gli1. We have investigated whether Gli1 may regulate c-fos by selectively knocking down Gli1 expression using anti-Gli1 shRNA or by treating cells with a Hedgehog pathway inhibitor, cyclopamine. A2780-CP70 cells were treated with an IC50 dose of anti-Gli1 shRNA and the changes in RNA and protein levels were analyzed over a 72 hour time course. No changes in the expression of c-jun or c-fos RNA transcripts were observed versus control. Western blot analyses showed Gli1 protein expression was reduced at 24 hours and undetectable at 48 and 72 hours. Sonic hedgehog (Shh) protein levels were dramatically reduced by 24 hours, low but measureable at 48 hours, and undetectable at 72 hours. Indian hedgehog (Ihh) expression increased at 24 hours and remained stable for the remainder of the 72 hour time course. Treatment of A2780-CP70 cells with an IC50 dose of cyclopamine resulted in a different intracellular response than treatment with Gli1 shRNA. Cyclopamine specifically targets the cell membrane receptor Smoothened. Experiments were performed analyzing RNA and protein expression over a 72 hour time period after treatment with cyclopamine. C-jun and c-fos RNA transcript levels were higher than control 6 hours after treatment and remained high for the duration of the 72 hour experiment. Western blot analyses after cyclopamine treatment showed Gli1 protein expression gradually decreasing during the 72 hours, but were never fully depleted. Expression of Shh increased over 72 hours, while Ihh levels remained stable. The identical Gli-Binding-Site (GBS) in the c-jun promoter was found in the promoter of c-fos. We investigated whether the Gli1 isoform that binds c-jun, also binds c-fos. Simultaneous Western and Southwestern blots show only one of the known Gli1 isoform binds the promoter of c-fos. Further studies with Chromatin Immunoprecipitation (ChIP) assays confirmed the 130 kDa isoform of Gli1 binds the GBS of the c-fos promoter. We conclude that the 130 kDa Gli1 isoform binds both components of AP1, c-jun and c-fos, and modulates the cellular resistance to cisplatin. Citation Format: Lauren Amable, Kenji Kudo, Elaine Gavin, Jason Fain, Eddie Reed. A specific Isoform of Gli1 binds the promoter region of c-fos in A2780-CP70 human ovarian 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 LB-223. doi:10.1158/1538-7445.AM2013-LB-223

Collaboration


Dive into the Elaine Gavin's collaboration.

Top Co-Authors

Avatar

Eddie Reed

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kenji Kudo

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Lauren Amable

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rodney P. Rocconi

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Jingfang Ju

Stony Brook University

View shared research outputs
Top Co-Authors

Avatar

Yaguang Xi

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Lalita A. Shevde

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gary A. Piazza

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Ileana V. Aragon

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Jason Fain

University of South Alabama

View shared research outputs
Researchain Logo
Decentralizing Knowledge