Network


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

Hotspot


Dive into the research topics where Preston D. Moore is active.

Publication


Featured researches published by Preston D. Moore.


Journal of Immunology | 2013

STAT6 Deficiency Ameliorates Severity of Oxazolone Colitis by Decreasing Expression of Claudin-2 and Th2-Inducing Cytokines

Michael J. Rosen; Rupesh Chaturvedi; M. Kay Washington; Lindsay A. Kuhnhein; Preston D. Moore; Scott S. Coggeshall; Elizabeth McDonough; Jörn-Hendrik Weitkamp; Amar B. Singh; Lori A. Coburn; Christopher S. Williams; Fang Yan; Luc Van Kaer; R. Stokes Peebles; Keith T. Wilson

Patients suffering from ulcerative colitis (UC) exhibit chronic colonic inflammation caused by a dysregulated mucosal immune response and epithelial barrier disruption. Th2 cytokines, including IL-13, have been implicated in the pathogenesis of UC. IL-13 induces phosphorylation of STAT6, and we previously demonstrated increased epithelial p-STAT6 in children with UC. In this study, we investigated the role of STAT6 in oxazolone colitis, a murine model of UC, by inducing colitis in STAT6-deficient (STAT6−/−) and wild type (WT) mice. We observed increased epithelial cell, T cell, macrophage, and NKT cell STAT6 phosphorylation, as well as increased p-STAT6+ IL-13–producing NKT cells, in colitic WT mice. Colitis was attenuated in STAT6−/− mice, with improvements in weight, colon length, and histopathology. There was decreased induction of the pore-forming tight junction protein claudin-2 in STAT6−/− mice. Similarly, short hairpin RNA STAT6 knockdown reduced claudin-2 induction and transepithelial resistance decrease in IL-13–treated human T84 cells. Tissue expression of IL-13, IFN-γ, IL-17, and IL-10 mRNA was similarly induced in WT and STAT6−/− colitic mice; however, we observed increased mRNA expression for the Th2-inducing cytokines IL-33 and thymic stromal lymphopoietin in WT mice with colitis, which was abrogated in STAT6−/− mice. Mesenteric lymph node cells from STAT6−/− mice with colitis exhibited reduced secretion of IL-4, IL-5, IL-13, and IFN-γ. IL-33 augmented mesenteric lymph node cell secretion of IL-5, IL-13, IL-6, and IFN-γ. These data implicate STAT6 in the pathogenesis of colitis in vivo with important roles in altering epithelial barrier function and regulating Th2-inducing cytokine production.


Cancer Research | 2017

Kinome-wide RNA interference screen reveals a role for PDK1 in acquired resistance to CDK4/6 inhibition in ER-positive breast cancer

Valerie M. Jansen; Neil E. Bhola; Joshua A. Bauer; Luigi Formisano; Lee Km; Katherine E. Hutchinson; Agnieszka K. Witkiewicz; Preston D. Moore; Monica V. Estrada; Violeta Sanchez; Paula I. Gonzalez Ericsson; Melinda E. Sanders; Paula R. Pohlmann; Michael J. Pishvaian; David A. Riddle; Teresa C. Dugger; Wenyi Wei; Erik S. Knudsen; Carlos L. Arteaga

Acquired resistance to cyclin-dependent kinases 4 and 6 (CDK4/6) small-molecule inhibitors in breast cancer arises through mechanisms that are yet uncharacterized. In this study, we used a kinome-wide siRNA screen to identify kinases that, when downregulated, yield sensitivity to the CDK4/6 inhibitor ribociclib. In this manner, we identified 3-phosphoinositide-dependent protein kinase 1 (PDK1) as a key modifier of ribociclib sensitivity in estrogen receptor-positive MCF-7 breast cancer cells. Pharmacologic inhibition of PDK1 with GSK2334470 in combination with ribociclib or palbociclib, another CDK4/6 inhibitor, synergistically inhibited proliferation and increased apoptosis in a panel of ER-positive breast cancer cell lines. Ribociclib-resistant breast cancer cells selected by chronic drug exposure displayed a relative increase in the levels of PDK1 and activation of the AKT pathway. Analysis of these cells revealed that CDK4/6 inhibition failed to induce cell-cycle arrest or senescence. Mechanistic investigations showed that resistant cells coordinately upregulated expression of cyclins A, E, and D1, activated phospho-CDK2, and phospho-S477/T479 AKT. Treatment with GSK2334470 or the CDK2 inhibitor dinaciclib was sufficient to reverse these events and to restore the sensitivity of ribociclib-resistant cells to CDK4/6 inhibitors. Ribociclib, in combination with GSK2334470 or the PI3Kα inhibitor alpelisib, decreased xenograft tumor growth more potently than each drug alone. Taken together, our results highlight a role for the PI3K-PDK1 signaling pathway in mediating acquired resistance to CDK4/6 inhibitors. Cancer Res; 77(9); 2488-99. ©2017 AACR.


Molecular & Cellular Proteomics | 2015

Activating PIK3CA Mutations Induce an Epidermal Growth Factor Receptor (EGFR)/Extracellular Signal-regulated Kinase (ERK) Paracrine Signaling Axis in Basal-like Breast Cancer

Christian D. Young; Lisa J. Zimmerman; Daisuke Hoshino; Luigi Formisano; Ariella B. Hanker; Michael L. Gatza; Meghan M. Morrison; Preston D. Moore; Corbin A. Whitwell; Bhuvanesh Dave; Thomas Stricker; Neil E. Bhola; Grace O. Silva; Premal Patel; Dana M. Brantley-Sieders; Maren K. Levin; Marina Horiates; Norma Alonzo Palma; Kai Wang; Philip J. Stephens; Charles M. Perou; Alissa M. Weaver; Joyce O'Shaughnessy; Jenny C. Chang; Ben Ho Park; Daniel C. Liebler; Rebecca S. Cook; Carlos L. Arteaga

Mutations in PIK3CA, the gene encoding the p110α catalytic subunit of phosphoinositide 3-kinase (PI3K) have been shown to transform human mammary epithelial cells (MECs). These mutations are present in all breast cancer subtypes, including basal-like breast cancer (BLBC). Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), we identified 72 protein expression changes in human basal-like MECs with knock-in E545K or H1047R PIK3CA mutations versus isogenic MECs with wild-type PIK3CA. Several of these were secreted proteins, cell surface receptors or ECM interacting molecules and were required for growth of PIK3CA mutant cells as well as adjacent cells with wild-type PIK3CA. The proteins identified by MS were enriched among human BLBC cell lines and pointed to a PI3K-dependent amphiregulin/EGFR/ERK signaling axis that is activated in BLBC. Proteins induced by PIK3CA mutations correlated with EGFR signaling and reduced relapse-free survival in BLBC. Treatment with EGFR inhibitors reduced growth of PIK3CA mutant BLBC cell lines and murine mammary tumors driven by a PIK3CA mutant transgene, all together suggesting that PIK3CA mutations promote tumor growth in part by inducing protein changes that activate EGFR.


Inflammatory Bowel Diseases | 2015

IL-33 signaling protects from murine oxazolone colitis by supporting intestinal epithelial function

Amanda Waddell; Jefferson Vallance; Preston D. Moore; Amy T. Hummel; David Wu; Shiva Kumar Shanmukhappa; Lin Fei; M. Kay Washington; Phillip Minar; Lori A. Coburn; Susumu Nakae; Keith T. Wilson; Lee A. Denson; Simon P. Hogan; Michael J. Rosen

Background:IL-33, a member of the IL-1 cytokine family that signals through ST2, is upregulated in ulcerative colitis (UC); however, the role of IL-33 in colitis remains unclear. IL-33 augments type 2 immune responses, which have been implicated in UC pathogenesis. We sought to determine the role of IL-33 signaling in oxazolone (OXA) colitis, a type 2 cytokine-mediated murine model of UC. Methods:Colon mucosal IL-33 expression was compared between pediatric and adult UC and non-IBD patients using immunohistochemistry and real-time PCR. OXA colitis was induced in WT, IL-33−/−, and ST2−/− mice, and histopathology, cytokine levels, and goblet cells were assessed. Transepithelial resistance was measured across IL-33-treated T84 cell monolayers. Results:Colon mucosal IL-33 was increased in pediatric patients with active UC and in OXA colitis. IL-33−/− and ST2−/− OXA mice exhibited increased disease severity compared with WT OXA mice. OXA induced a mixed mucosal cytokine response, but few differences were observed between OXA WT and IL-33−/− or ST2−/− mice. Goblet cells were significantly decreased in IL-33−/− and ST2−/− OXA compared with WT OXA mice. IL-33 augmented transepithelial resistance in T84 cells, and this effect was blocked by the ERK1/2 inhibitor PD98,059. Conclusions:OXA colitis is exacerbated in IL-33−/− and ST2−/− mice. Increased mucosal IL-33 in human UC and murine colitis may be a homeostatic response to limit inflammation, potentially through effects on epithelial barrier function. Further investigation of IL-33 protective mechanisms would inform the development of novel therapeutic approaches.


Clinical Cancer Research | 2017

HER2-overexpressing breast cancers amplify FGFR signaling upon acquisition of resistance to dual therapeutic blockade of HER2

Ariella B. Hanker; Joan T. Garrett; Monica V. Estrada; Preston D. Moore; Paula I. Gonzalez Ericsson; James P. Koch; Emma Langley; Sharat Singh; Phillip Kim; Garrett Michael Frampton; Eric M. Sanford; Philip Owens; Jennifer Becker; M. Reid Groseclose; Stephen Castellino; Heikki Joensuu; Jens Huober; Jan C. Brase; Samira Majjaj; Sylvain Brohée; David Venet; David Norman Brown; José Baselga; Martine Piccart; Christos Sotiriou; Carlos L. Arteaga

Purpose: Dual blockade of HER2 with trastuzumab and lapatinib or pertuzumab has been shown to be superior to single-agent trastuzumab. However, a significant fraction of HER2-overexpressing (HER2+) breast cancers escape from these drug combinations. In this study, we sought to discover the mechanisms of acquired resistance to the combination of lapatinib + trastuzumab. Experimental Design: HER2+ BT474 xenografts were treated with lapatinib + trastuzumab long-term until resistance developed. Potential mechanisms of acquired resistance were evaluated in lapatinib + trastuzumab-resistant (LTR) tumors by targeted capture next-generation sequencing. In vitro experiments were performed to corroborate these findings, and a novel drug combination was tested against LTR xenografts. Gene expression and copy-number analyses were performed to corroborate our findings in clinical samples. Results: LTR tumors exhibited an increase in FGF3/4/19 copy number, together with an increase in FGFR phosphorylation, marked stromal changes in the tumor microenvironment, and reduced tumor uptake of lapatinib. Stimulation of BT474 cells with FGF4 promoted resistance to lapatinib + trastuzumab in vitro. Treatment with FGFR tyrosine kinase inhibitors reversed these changes and overcame resistance to lapatinib + trastuzumab. High expression of FGFR1 correlated with a statistically shorter progression-free survival in patients with HER2+ early breast cancer treated with adjuvant trastuzumab. Finally, FGFR1 and/or FGF3 gene amplification correlated with a lower pathologic complete response in patients with HER2+ early breast cancer treated with neoadjuvant anti-HER2 therapy. Conclusions: Amplification of FGFR signaling promotes resistance to HER2 inhibition, which can be diminished by the combination of HER2 and FGFR inhibitors. Clin Cancer Res; 23(15); 4323–34. ©2017 AACR.


Cancer Research | 2017

Extracellular Matrix/Integrin Signaling Promotes Resistance to Combined Inhibition of HER2 and PI3K in HER2+ Breast Cancer

Ariella B. Hanker; Monica V. Estrada; Giampaolo Bianchini; Preston D. Moore; Junfei Zhao; Feixiong Cheng; James P. Koch; Luca Gianni; Darren R. Tyson; Violeta Sanchez; Brent N. Rexer; Melinda E. Sanders; Zhongming Zhao; Thomas Stricker; Carlos L. Arteaga

PIK3CA mutations are associated with resistance to HER2-targeted therapies. We previously showed that HER2+/PIK3CAH1047R transgenic mammary tumors are resistant to the HER2 antibodies trastuzumab and pertuzumab but respond to PI3K inhibitor buparlisib (TPB). In this study, we identified mechanisms of resistance to combined inhibition of HER2 and PI3K. TPB-resistant tumors were generated by treating HER2+/PIK3CAH1047R tumor-bearing mice long term with the drug combination. RNA sequencing of TPB-resistant tumors revealed that extracellular matrix and cell adhesion genes, including collagen II (Col2a1), were markedly upregulated, accompanied by activation of integrin β1/Src. Cells derived from drug-resistant tumors were sensitive to TBP when grown in vitro, but exhibited resistance when plated on collagen or when reintroduced into mice. Drug resistance was partially reversed by the collagen synthesis inhibitor ethyl-3,4-dihydroxybenzoate. Inhibition of integrin β1/Src blocked collagen-induced resistance to TPB and inhibited growth of drug-resistant tumors. High collagen II expression was associated with significantly lower clinical response to neoadjuvant anti-HER2 therapy in HER2+ breast cancer patients. Overall, these data suggest that upregulation of collagen/integrin/Src signaling contributes to resistance to combinatorial HER2 and PI3K inhibition. Cancer Res; 77(12); 3280-92. ©2017 AACR.


Cancer Research | 2016

Abstract PD2-06: Inhibition of 3-phosphoinositide dependent protein kinase 1 (PDK1) synergizes with CDK4/6 inhibitors against ER-positive breast cancer

Valerie M. Jansen; Neil E. Bhola; Ja Bauer; Luigi Formisano; Preston D. Moore; James P. Koch; Cl Arteaga

Background: Dysregulation in cell cycle checkpoints is common in cancer. Small molecule inhibitors that target the CDK4/6/cyclinD1 pathway of the cell cycle are in clinical development. Recently the combination of the CDK4/6 inhibitor palbociclib and the aromatase inhibitor letrozole was approved for the treatment of post-menopausal women with ER+/HER2- advanced breast cancer. However, not all patients benefit from CDK4/6 inhibitors and a significant fraction of them eventually progress on these agents, underscoring the need to develop potent therapeutic strategies to circumvent drug resistance. Methods: We performed a high-throughput RNA interference (RNAi) kinome screen targeting 720 kinases to identify targetable molecules whose inhibition, in combination with the CDK4/6 inhibitor LEE011 (ribociclib), induced synthetic lethality in MCF7 ER+ breast cancer cells. PDK1 RNAi oligonucleotides and the PDK1 inhibitor GSK2334470 in combination with each of the CDK4/6 inhibitors, palbociclib and LEE011, were tested against ER+ breast cancer cells. In vivo anti-tumor efficacy of LEE011 and GSK2334470 was assessed in ovariectomized athymic nude mice bearing MCF7 xenografts. Results: A siRNA kinome screen identified PDK1 as the top RNA whose downregulation sensitized MCF7 cells to CDK4/6 inhibitors. This was confirmed with independent siRNAs in ER+ MCF7, T47D, HCC1428 and HCC1500 breast cancer cells. Pharmacological inhibition of PDK1 with the ATP-competitive, small molecule inhibitor GSK2334470 in combination with each of the CDK4/6 inhibitors, LEE011 and palbociclib, synergistically inhibited proliferation and increased apoptosis of MCF7 and T47D cells (combination index 0.19-0.89). LEE011-resistant MCF7 and T47D cells were generated by chronic treatment with doses of LEE011 up to 1 µM. Drug-resistant cells displayed increased levels of PDK1, phosphorylated Rb, and phosphorylated S6 ribosomal protein (pS6), an effector of the PDK1 substrate p70S6K, compared to parental drug-sensitive cells. Inhibition of PDK1 with siRNA or GSK2334470 re-sensitized the LEE011-resistant cells to the CDK4/6 inhibitors. Genetic (RNAi) and pharmacological inhibition of PDK1 (with GSK2334470) abrogated pS6 levels whereas inhibition of AKT with the small molecule inhibitor MK2206 did not affect pS6 levels, suggesting PDK1 can induce resistance to CDK4/6 inhibitors via p70S6K/pS6 signaling in an AKT-independent manner. The effects observed in cell lines in culture were recapitulated in vivo using MCF7 xenografts established in ovariectomized nude mice in the absence of estrogen supplementation. Treatment with GSK2334470 and LEE011 induced tumor regressions (8/8 tumors by RECIST criteria) more potently than either drug alone. Conclusions: These data support a critical role of PDK1 in mediating acquired resistance to CDK4/6 inhibitors in ER+ breast cancer cells. Co-targeting of the PDK1 and CDK4/6 pathways may overcome resistance to CDK4/6 inhibitors and is worthy of further translational and clinical investigation in patients with ER+ breast cancer. Citation Format: Jansen VM, Bhola NE, Bauer JA, Formisano L, Moore P, Koch J, Arteaga CL. Inhibition of 3-phosphoinositide dependent protein kinase 1 (PDK1) synergizes with CDK4/6 inhibitors against ER-positive breast cancer. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr PD2-06.


Cancer Research | 2016

Abstract PD6-01: Cooperation of clinically-relevant genomic alterations transforms mammary progenitor cells to metaplastic breast cancer

Mellissa Hicks; Philip Owens; Preston D. Moore; Jennifer M. Giltnane; Monica V. Estrada; Melinda E. Sanders; Rebecca S. Cook; Cl Arteaga; Justin M. Balko

Background: We have previously identified the MAPK phosphatase dual specificity phosphatase 4 (Dusp4) as frequently down-regulated in triple-negative and basal-like breast cancer (TNBC and BLBC, respectively). DUSP4 is deregulated in BLBCs by heterozygous and homozygous loss, as well as by promoter CpG methylation. In TNBC and BLBC, DUSP4 deregulation frequently co-occurs with MYC amplification and mutations in TP53. In addition, loss of DUSP4 promotes activation of ETS-1 and cJUN, transcription factors downstream of Ras/MAPK and JNK, while imparting resistance to chemotherapy and promoting tumor-initiating cells, suggesting DUSP4 is a tumor suppressor. Thus, we hypothesized that loss of Dusp4 in the mammary epithelium, together with MYC amplification and mutations in Trp53, would promote tumorigenesis. Methods: We generated transgenic mice with a conditional floxed Dusp4 allele (Dusp4FLOX) and isolated mammary epithelial progenitor cells. The resulting cells were transduced with adenovirus to deliver either Cre recombinase to excise the Dusp4 locus (Dusp4NULL) or GFP control. CRISPR-mediated mutagenesis (or non-targeting CRISPR control) was used to disrupt exon 7 or 8 of Trp53 causing relevant p53 mutations and MYC was overexpressed (or LACZ control). Isogenic cell lines were assessed for tumorigenicity, ploidy, sensitivity to chemotherapy, proliferation, and cell signaling. Results: The isolated parental (Dusp4FLOX) mammary cell line maintained proliferation through >50 passages without senescing and was highly cytokeratin (Ck) 5/14+ and moderately Ck 8/18+, suggestive of a bipotent progenitor-like lineage. As expected, p53 disruption, regardless of Dusp4 status, resulted in tetraploidy and loss of p21 expression. With concurrent Dusp4, p53 and MYC alterations, cells became increasingly resistant to chemotherapy and targeted agents, as measured with proliferation assays. In order to determine whether cells formed tumors in vivo, we implanted isogenic cell lines subcutaneously into nude mice. We found that only Dusp4NULL cells, together with p53 disruption and MYC overexpression (DPM) generated aggressive tumors (forming and ulcerating within 1-2 weeks), while the Dusp4FLOX counterparts (p53 disruption and MYC overexpression; PM) did not. Syngeneic transplantation revealed that DPM cells could also form tumors in an immune competent setting. Cells with Dusp4 loss and p53 mutation were also able to develop tumors without MYC amplification, but at a much slower rate. Tumors derived from the DPM cells were primarily metaplastic or basal-like in nature, including intra-tumor heterogeneity (pockets of Ck5 and pockets of Ck8, with regions of Ck8+ keratin pearls). Conclusions: We have developed a genetically relevant mouse model to study BLBC in immune competent mice for unique opportunities to understand the pathogenesis and therapy response in BLBC, including immunotherapy. These results support the hypothesis that DUSP4 loss promotes tumorigenesis in BLBC. An understanding of the mechanism whereby Dusp4 loss contributes to this process in vivo should provide a rational for novel therapeutic strategies to treat BLBCs with DUSP4 loss. Citation Format: Hicks M, Owens P, Moore P, Giltnane J, Estrada M, Sanders M, Cook R, Arteaga C, Balko J. Cooperation of clinically-relevant genomic alterations transforms mammary progenitor cells to metaplastic breast cancer. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr PD6-01.


Cancer Research | 2016

Abstract 302: ECM/Integrin signaling promotes resistance to the combination of HER2 and PI3K inhibitors in HER2+, PIK3CA-mutant breast cancer

Ariella B. Hanker; Monica V. Estrada; Junfei Zhao; Feixiong Cheng; Preston D. Moore; Darren R. Tyson; Violeta Sanchez; Brent N. Rexer; Melinda E. Sanders; Zhongming Zhao; Thomas Stricker; Carlos L. Arteaga


Gastroenterology | 2015

20 IL-33 Signaling Augments Epithelial Barrier Function In Vitro and Protects Mice From Oxazolone Colitis

Amanda Waddell; Jefferson Vallance; Preston D. Moore; Amy T. Hummel; Shiva Kumar Shanmukhappa; Kay Washington; Susumu Nakae; Andrew N. J. McKenzie; Keith T. Wilson; Lee A. Denson; Simon P. Hogan; Michael J. Rosen

Collaboration


Dive into the Preston D. Moore's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keith T. Wilson

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Melinda E. Sanders

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kay Washington

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge