145 results on '"Fields AP"'
Search Results
2. ECT2 (epithelial cell transforming sequence 2 oncogene)
- Author
-
Justilien, V, primary and Fields, AP, additional
- Published
- 2013
- Full Text
- View/download PDF
3. PRKCI (protein kinase C, iota)
- Author
-
Justilien, V, primary and Fields, AP, additional
- Published
- 2012
- Full Text
- View/download PDF
4. Endoxifen downregulates AKT phosphorylation through protein kinase C beta 1 inhibition in ERα+ breast cancer.
- Author
-
Jayaraman S, Wu X, Kalari KR, Tang X, Kuffel MJ, Bruinsma ES, Jalali S, Peterson KL, Correia C, Kudgus RA, Kaufmann SH, Renuse S, Ingle JN, Reid JM, Ames MM, Fields AP, Schellenberg MJ, Hawse JR, Pandey A, and Goetz MP
- Abstract
Endoxifen, a secondary tamoxifen metabolite, is a potent antiestrogen exhibiting estrogen receptor alpha (ERα) binding at nanomolar concentrations. Phase I/II clinical trials identified clinical activity of Z-endoxifen (ENDX), in endocrine-refractory metastatic breast cancer as well as ERα+ solid tumors, raising the possibility that ENDX may have a second, ERα-independent, mechanism of action. An unbiased mass spectrometry approach revealed that ENDX concentrations achieved clinically with direct ENDX administration (5 µM), but not low concentrations observed during tamoxifen treatment (<0.1 µM), profoundly altered the phosphoproteome of the aromatase expressing MCF7AC1 cells with limited impact on the total proteome. Computational analysis revealed protein kinase C beta (PKCβ) and protein kinase B alpha or AKT1 as potential kinases responsible for mediating ENDX effects on protein phosphorylation. ENDX more potently inhibited PKCβ1 kinase activity compared to other PKC isoforms, and ENDX binding to PKCβ1 was confirmed using Surface Plasma Resonance. Under conditions that activated PKC/AKT signaling, ENDX induced PKCβ1 degradation, attenuated PKCβ1-activated AKT
Ser473 phosphorylation, diminished AKT substrate phosphorylation, and induced apoptosis. ENDX's effects on AKT were phenocopied by siRNA-mediated PKCβ1 knockdown or treatment with the pan-AKT inhibitor, MK-2206, while overexpression of constitutively active AKT diminished ENDX-induced apoptosis. These findings, which identify PKCβ1 as an ENDX target, indicate that PKCβ1/ENDX interactions suppress AKT signaling and induce apoptosis in breast cancer., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
5. Evaluation of cell-free DNA approaches for multi-cancer early detection.
- Author
-
Jamshidi A, Liu MC, Klein EA, Venn O, Hubbell E, Beausang JF, Gross S, Melton C, Fields AP, Liu Q, Zhang N, Fung ET, Kurtzman KN, Amini H, Betts C, Civello D, Freese P, Calef R, Davydov K, Fayzullina S, Hou C, Jiang R, Jung B, Tang S, Demas V, Newman J, Sakarya O, Scott E, Shenoy A, Shojaee S, Steffen KK, Nicula V, Chien TC, Bagaria S, Hunkapiller N, Desai M, Dong Z, Richards DA, Yeatman TJ, Cohn AL, Thiel DD, Berry DA, Tummala MK, McIntyre K, Sekeres MA, Bryce A, Aravanis AM, Seiden MV, and Swanton C
- Subjects
- Humans, Early Detection of Cancer, Biomarkers, Tumor genetics, DNA Methylation, Cell-Free Nucleic Acids genetics, Neoplasms diagnosis, Neoplasms genetics
- Abstract
In the Circulating Cell-free Genome Atlas (NCT02889978) substudy 1, we evaluate several approaches for a circulating cell-free DNA (cfDNA)-based multi-cancer early detection (MCED) test by defining clinical limit of detection (LOD) based on circulating tumor allele fraction (cTAF), enabling performance comparisons. Among 10 machine-learning classifiers trained on the same samples and independently validated, when evaluated at 98% specificity, those using whole-genome (WG) methylation, single nucleotide variants with paired white blood cell background removal, and combined scores from classifiers evaluated in this study show the highest cancer signal detection sensitivities. Compared with clinical stage and tumor type, cTAF is a more significant predictor of classifier performance and may more closely reflect tumor biology. Clinical LODs mirror relative sensitivities for all approaches. The WG methylation feature best predicts cancer signal origin. WG methylation is the most promising technology for MCED and informs development of a targeted methylation MCED test., Competing Interests: Declaration of interests A.J., O.V., E.H., J.F.B., S.G., Q.L., N.Z., E.T.F., K.N.K., H.A., C.B., D.C., K.D., S.F., C.H., R.J., B.J., S.T., C.M., V.D., J.N., O.S., E.S., A.S., S.S., K.K.S., V.N., A.P.F., T.C.C., S.B., N.H., M.D., Z.D., and M.P.H. are employees of GRAIL, LLC, with equity in Illumina, Inc. C.M. also holds stock in Novartis, Clovis, Cara, Gilead, and Bluebird. M.C.L. is an uncompensated consultant for GRAIL, LLC. The Mayo Clinic was compensated for M.C.L.’s and D.D.T.’s advisory board activities for GRAIL, LLC. E.A.K. is a consultant for GRAIL, LLC. D.A.R. is a consultant for Ipsen. M.A.S. is a consultant for Celgene, Millennium, and Syros Pharmaceuticals. A.M.A. was previously employed by GRAIL, LLC; has equity in Illumina, Inc.; is currently employed by Illumina, Inc.; and is an advisor to and an equity holder in Foresite Labs and Myst Therapeutics. M.V.S. is an employee of and holds stock in McKesson Corporation, and is a clinical adviser for GRAIL, LLC. D.A.B. is a co-owner of Berry Consultants, LLC. A.H.B. is a consultant for Pfizer, Merck, Bayer, and Astellas Pharmaceuticals. C.S. holds stock in Illumina, Inc., Epic Biosciences, and Apogen Biotech; receives grants from Pfizer and AstraZeneca; receives honoraria or consultant fees from Roche Ventana, Celgene, Pfizer, Novartis, Genentech, and BMS; and is a co-founder of Achilles Therapeutics. S.G., O.V., A.P.F., A.J., K.D., V.N., J.F.B., C.M., E.H., Q.L., N.Z., P.F., and O.S. are inventors on pending patent applications related to this work, for which GRAIL, LLC, has ownership rights. GRAIL, LLC, a subsidiary of Illumina, Inc., is currently held separate from Illumina, Inc., under the terms of the Interim Measures Order of the European Commission dated October 29, 2021. C.S. recieved grant support from AstraZeneca, Boehringer-Ingelheim, Bristol Myers Squibb, Pfizer, Roche-Ventana, Invitae (previously Archer Dx Inc), and Ono Pharmaceutical; is an AstraZeneca Advisory Board member and Chief Investigator for the AZ MeRmaiD 1 and 2 clinical trials and is also Co-Chief Investigator of the NHS Galleri trial funded by GRAIL and a paid member of GRAIL’s Scientific Advisory Board (SAB); received consultant fees from Achilles Therapeutics (also SAB member), Bicycle Therapeutics (also a SAB member), Genentech, Medicxi, Roche Innovation Centre – Shanghai, Metabomed, and the Sarah Cannon Research Institute; received honoraria from Amgen, AstraZeneca, Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol Myers Squibb, Illumina, and Roche-Ventana; had stock options in Apogen Biotechnologies and GRAIL until June 2021, and currently has stock options in Epic Bioscience, Bicycle Therapeutics, and Achilles Therapeutics; and is a co-founder of Achilles Therapeutics; holds patents relating to assay technology to detect tumour recurrence (PCT/GB2017/053289), targeting neoantigens (PCT/EP2016/059401), identifying patent response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), and identifying patients who respond to cancer treatment (PCT/GB2018/051912); holds US patent relating to detecting tumour mutations (PCT/US2017/28013), methods for lung cancer detection (US20190106751A1); and holds both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892). C.S. is a Royal Society Napier Research Professor (RSRP\R\210001) and has received funding from the Francis Crick Institute that receives its core funding from Cancer Research UK (CC2041), the UK Medical Research Council (CC2041), and the Wellcome Trust (CC2041); Cancer Research UK (TRACERx [C11496/A17786], PEACE [C416/A21999], and CRUK Cancer Immunotherapy Catalyst Network); Cancer Research UK Lung Cancer Centre of Excellence (C11496/A30025); the Rosetrees Trust, Butterfield and Stoneygate Trusts; NovoNordisk Foundation (ID16584); Royal Society Professorship Enhancement Award (RP/EA/180007); National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre; the Cancer Research UK-University College London Centre; Experimental Cancer Medicine Centre; the Breast Cancer Research Foundation (US) (BCRF-22-157); Cancer Research UK Early Detection and Diagnosis Primer Award (Grant EDDPMA-Nov21/100034); The Mark Foundation for Cancer Research Aspire Award (Grant 21-029-ASP); Stand Up To Cancer-LUNGevity American Lung Association Lung Cancer Interception Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT23-17); and an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 835297)., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
6. Protein kinase Cι mediates immunosuppression in lung adenocarcinoma.
- Author
-
Yin N, Liu Y, Weems C, Shreeder B, Lou Y, Knutson KL, Murray NR, and Fields AP
- Subjects
- Mice, Animals, CD8-Positive T-Lymphocytes, Immunosuppression Therapy, B7-H1 Antigen, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms pathology, Adenocarcinoma of Lung genetics
- Abstract
Lung adenocarcinoma (LUAD) is the most prevalent form of non-small cell lung cancer (NSCLC) and a leading cause of cancer death. Immune checkpoint inhibitors (ICIs) of programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) signaling induce tumor regressions in a subset of LUAD, but many LUAD tumors exhibit resistance to ICI therapy. Here, we identified Prkci as a major determinant of response to ICI in a syngeneic mouse model of oncogenic mutant Kras / Trp53 loss (KP)-driven LUAD. Protein kinase Cι (PKCι)-dependent KP tumors exhibited resistance to anti-PD-1 antibody therapy (α-PD-1), whereas KP tumors in which Prkci was genetically deleted (KPI tumors) were highly responsive. Prkci- dependent resistance to α-PD-1 was characterized by enhanced infiltration of myeloid-derived suppressor cells (MDSCs) and decreased infiltration of CD8
+ T cells in response to α-PD-1. Mechanistically, Prkci regulated YAP1-dependent expression of Cxcl5 , which served to attract MDSCs to KP tumors. The PKCι inhibitor auranofin inhibited KP tumor growth and sensitized these tumors to α-PD-1, whereas expression of either Prkci or its downstream effector Cxcl5 in KPI tumors induced intratumoral infiltration of MDSCs and resistance to α-PD-1. PRKCI expression in tumors of patients with LUAD correlated with genomic signatures indicative of high YAP1-mediated transcription, elevated MDSC infiltration and low CD8+ T cell infiltration, and with elevated CXCL5 / 6 expression. Last, PKCι-YAP1 signaling was a biomarker associated with poor response to ICI in patients with LUAD. Our data indicate that immunosuppressive PKCι-YAP1-CXCL5 signaling is a key determinant of response to ICI, and pharmacologic inhibition of PKCι may improve therapeutic response to ICI in patients with LUAD.- Published
- 2022
- Full Text
- View/download PDF
7. Prkci Regulates Autophagy and Pancreatic Tumorigenesis in Mice.
- Author
-
Inman KS, Liu Y, Scotti Buzhardt ML, Leitges M, Krishna M, Crawford HC, Fields AP, and Murray NR
- Abstract
Protein kinase C iota (PKCι) functions as a bonafide human oncogene in lung and ovarian cancer and is required for Kras
G12D -mediated lung cancer initiation and progression. PKCι expression is required for pancreatic cancer cell growth and maintenance of the transformed phenotype; however, nothing is known about the role of PKCι in pancreas development or pancreatic tumorigenesis. In this study, we investigated the effect of pancreas-specific ablation of PKCι expression on pancreatic cellular homeostasis, susceptibility to pancreatitis, and KrasG12D -mediated pancreatic cancer development. Knockout of pancreatic Prkci significantly increased pancreatic immune cell infiltration, acinar cell DNA damage, and apoptosis, but reduced sensitivity to caerulein-induced pancreatitis. Prkci -ablated pancreatic acinar cells exhibited P62 aggregation and a loss of autophagic vesicles. Loss of pancreatic Prkci promoted KrasG12D -mediated pancreatic intraepithelial neoplasia formation but blocked progression to adenocarcinoma, consistent with disruption of autophagy. Our results reveal a novel promotive role for PKCι in pancreatic epithelial cell autophagy and pancreatic cancer progression.- Published
- 2022
- Full Text
- View/download PDF
8. Aberrant Expression and Subcellular Localization of ECT2 Drives Colorectal Cancer Progression and Growth.
- Author
-
Cook DR, Kang M, Martin TD, Galanko JA, Loeza GH, Trembath DG, Justilien V, Pickering KA, Vincent DF, Jarosch A, Jurmeister P, Waters AM, Hibshman PS, Campbell AD, Ford CA, Keku TO, Yeh JJ, Lee MS, Cox AD, Fields AP, Sandler RS, Sansom OJ, Sers C, Schaefer A, and Der CJ
- Subjects
- Aged, Animals, Disease Models, Animal, Disease Progression, Female, Humans, Male, Mice, Colorectal Neoplasms genetics, Genomics methods, Proto-Oncogene Proteins metabolism
- Abstract
ECT2 is an activator of RHO GTPases that is essential for cytokinesis. In addition, ECT2 was identified as an oncoprotein when expressed ectopically in NIH/3T3 fibroblasts. However, oncogenic activation of ECT2 resulted from N-terminal truncation, and such truncated ECT2 proteins have not been found in patients with cancer. In this study, we observed elevated expression of full-length ECT2 protein in preneoplastic colon adenomas, driven by increased ECT2 mRNA abundance and associated with APC tumor-suppressor loss. Elevated ECT2 levels were detected in the cytoplasm and nucleus of colorectal cancer tissue, suggesting cytoplasmic mislocalization as one mechanism of early oncogenic ECT2 activation. Importantly, elevated nuclear ECT2 correlated with poorly differentiated tumors, and a low cytoplasmic:nuclear ratio of ECT2 protein correlated with poor patient survival, suggesting that nuclear and cytoplasmic ECT2 play distinct roles in colorectal cancer. Depletion of ECT2 reduced anchorage-independent cancer cell growth and invasion independent of its function in cytokinesis, and loss of Ect2 extended survival in a Kras
G12D Apc -null colon cancer mouse model. Expression of ECT2 variants with impaired nuclear localization or guanine nucleotide exchange catalytic activity failed to restore cancer cell growth or invasion, indicating that active, nuclear ECT2 is required to support tumor progression. Nuclear ECT2 promoted ribosomal DNA transcription and ribosome biogenesis in colorectal cancer. These results support a driver role for both cytoplasmic and nuclear ECT2 overexpression in colorectal cancer and emphasize the critical role of precise subcellular localization in dictating ECT2 function in neoplastic cells. SIGNIFICANCE: ECT2 overexpression and mislocalization support its role as a driver in colon cancer that is independent from its function in normal cell cytokinesis., (©2021 American Association for Cancer Research.)- Published
- 2022
- Full Text
- View/download PDF
9. Protein kinase C ι and SRC signaling define reciprocally related subgroups of glioblastoma with distinct therapeutic vulnerabilities.
- Author
-
Kenchappa RS, Liu Y, Argenziano MG, Banu MA, Mladek AC, West R, Luu A, Quiñones-Hinojosa A, Hambardzumyan D, Justilien V, Leitges M, Sarkaria JN, Sims PA, Canoll P, Murray NR, Fields AP, and Rosenfeld SS
- Subjects
- Animals, Carcinogenesis genetics, Cell Line, Tumor, Disease Models, Animal, Gene Expression genetics, Gene Expression Regulation, Neoplastic genetics, Glioblastoma classification, Humans, Isoenzymes genetics, Mice, Oncogenes genetics, Protein Kinase C genetics, Protein Kinase C physiology, Signal Transduction physiology, Glioblastoma genetics, Glioblastoma metabolism, Isoenzymes metabolism, Protein Kinase C metabolism
- Abstract
We report that atypical protein kinase Cι (PKCι) is an oncogenic driver of glioblastoma (GBM). Deletion or inhibition of PKCι significantly impairs tumor growth and prolongs survival in murine GBM models. GBM cells expressing elevated PKCι signaling are sensitive to PKCι inhibitors, whereas those expressing low PKCι signaling exhibit active SRC signaling and sensitivity to SRC inhibitors. Resistance to the PKCι inhibitor auranofin is associated with activated SRC signaling and response to a SRC inhibitor, whereas resistance to a SRC inhibitor is associated with activated PKCι signaling and sensitivity to auranofin. Interestingly, PKCι- and SRC-dependent cells often co-exist in individual GBM tumors, and treatment of GBM-bearing mice with combined auranofin and SRC inhibitor prolongs survival beyond either drug alone. Thus, we identify PKCι and SRC signaling as distinct therapeutic vulnerabilities that are directly translatable into an improved treatment for GBM., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. Editorial.
- Author
-
Fields AP
- Subjects
- Antineoplastic Agents therapeutic use, Cell Movement, Cell Proliferation, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Humans, Isoenzymes genetics, Isoenzymes metabolism, Neoplasms classification, Neoplasms drug therapy, Neoplasms enzymology, Protein Kinase C metabolism, Cell Transformation, Neoplastic genetics, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Protein Kinase C genetics, Signal Transduction genetics
- Published
- 2021
- Full Text
- View/download PDF
11. Recurrent copy number gains drive PKCι expression and PKCι-dependent oncogenic signaling in human cancers.
- Author
-
Liu Y, Justilien V, Fields AP, and Murray NR
- Subjects
- Chromosomes, Human, Pair 3, Humans, Isoenzymes genetics, Neoplasms enzymology, Neoplasms pathology, Protein Kinase C genetics, Survival Analysis, DNA Copy Number Variations, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Isoenzymes metabolism, Neoplasms genetics, Oncogenes, Protein Kinase C metabolism, Signal Transduction genetics
- Abstract
PRKCI is frequently overexpressed in multiple human cancers, and PKCι expression is often prognostic for poor patient survival, indicating that elevated PKCι broadly plays an oncogenic role in the cancer phenotype. PKCι drives multiple oncogenic signaling pathways involved in transformed growth, and transgenic mouse models have revealed that PKCι is a critical oncogenic driver in both lung and ovarian cancers. We now report that recurrent 3q26 copy number gain (CNG) is the predominant genetic driver of PRKCI mRNA expression in all major human cancer types exhibiting such CNGs. In addition to PRKCI, CNG at 3q26 leads to coordinate CNGs of ECT2 and SOX2, two additional 3q26 genes that collaborate with PRKCI to drive oncogenic signaling and tumor initiation in lung squamous cell carcinoma. Interestingly however, whereas 3q26 CNG is a strong driver of PRKCI mRNA expression across all tumor types examined, it has differential effects on ECT2 and SOX2 mRNA expression. In some tumors types, particularly those with squamous histology, all three 3q26 oncogenes are coordinately overexpressed as a consequence of 3q26 CNG, whereas in other cancers only PRKCI and ECT2 mRNA are coordinately overexpressed. This distinct pattern of expression of 3q26 genes corresponds to differences in genomic signatures reflective of activation of specific PKCι oncogenic signaling pathways. In addition to highly prevalent CNG, some tumor types exhibit monoallelic loss of PRKCI. Interestingly, many tumors harboring monoallelic loss of PRKCI express significantly lower PRKCI mRNA and exhibit evidence of WNT/β-catenin signaling pathway activation, which we previously characterized as a major oncogenic pathway in a newly described, PKCι-independent molecular subtype of lung adenocarcinoma. Finally, we show that CNG-driven activation of PKCι oncogenic signaling predicts poor patient survival in many major cancer types. We conclude that CNG and monoallelic loss are the major determinants of tumor PRKCI mRNA expression across virtually all tumor types, but that tumor-type specific mechanisms determine whether these copy number alterations also drive expression of the collaborating 3q26 oncogenes ECT2 and SOX2, and the oncogenic PKCι signaling pathways activated through the collaborative action of these genes. Our analysis may be useful in identifying tumor-specific predictive biomarkers and effective PKCι-targeted therapeutic strategies in the multitude of human cancers harboring genetic activation of PRKCI., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
12. FoxM1 insufficiency hyperactivates Ect2-RhoA-mDia1 signaling to drive cancer.
- Author
-
Limzerwala JF, Jeganathan KB, Kloeber JA, Davies BA, Zhang C, Sturmlechner I, Zhong J, Fierro Velasco R, Fields AP, Yuan Y, Baker DJ, Zhou D, Li H, Katzmann DJ, and van Deursen JM
- Subjects
- Animals, GTP Phosphohydrolases, Mice, Signal Transduction, Actins metabolism, Forkhead Box Protein M1 metabolism, Neoplasms genetics
- Abstract
FoxM1 activates genes that regulate S-G2-M cell-cycle progression and, when overexpressed, is associated with poor clinical outcome in multiple cancers. Here we identify FoxM1 as a tumor suppressor in mice that, through its N-terminal domain, binds to and inhibits Ect2 to limit the activity of RhoA GTPase and its effector mDia1, a catalyst of cortical actin nucleation. FoxM1 insufficiency impedes centrosome movement through excessive cortical actin polymerization, thereby causing the formation of non-perpendicular mitotic spindles that missegregate chromosomes and drive tumorigenesis in mice. Importantly, low FOXM1 expression correlates with RhoA GTPase hyperactivity in multiple human cancer types, indicating that suppression of the newly discovered Ect2-RhoAmDia1 oncogenic axis by FoxM1 is clinically relevant. Furthermore, by dissecting the domain requirements through which FoxM1 inhibits Ect2 GEF activity, we provide mechanistic insight for the development of pharmacological approaches that target protumorigenic RhoA activity., Competing Interests: Competing interests: None.
- Published
- 2020
- Full Text
- View/download PDF
13. Translation Initiation Site Profiling Reveals Widespread Synthesis of Non-AUG-Initiated Protein Isoforms in Yeast.
- Author
-
Eisenberg AR, Higdon AL, Hollerer I, Fields AP, Jungreis I, Diamond PD, Kellis M, Jovanovic M, and Brar GA
- Subjects
- Codon metabolism, Peptide Chain Initiation, Translational genetics, Protein Biosynthesis genetics, Protein Isoforms metabolism, Saccharomyces cerevisiae genetics
- Abstract
Genomic analyses in budding yeast have helped define the foundational principles of eukaryotic gene expression. However, in the absence of empirical methods for defining coding regions, these analyses have historically excluded specific classes of possible coding regions, such as those initiating at non-AUG start codons. Here, we applied an experimental approach to globally annotate translation initiation sites in yeast and identified 149 genes with alternative N-terminally extended protein isoforms initiating from near-cognate codons upstream of annotated AUG start codons. These isoforms are produced in concert with canonical isoforms and translated with high specificity, resulting from initiation at only a small subset of possible start codons. The non-AUG initiation driving their production is enriched during meiosis and induced by low eIF5A, which is seen in this context. These findings reveal widespread production of non-canonical protein isoforms and unexpected complexity to the rules by which even a simple eukaryotic genome is decoded., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
14. Protein kinase Cι promotes UBF1-ECT2 binding on ribosomal DNA to drive rRNA synthesis and transformed growth of non-small-cell lung cancer cells.
- Author
-
Justilien V, Lewis KC, Meneses KM, Jamieson L, Murray NR, and Fields AP
- Subjects
- Amino Acid Motifs, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic pathology, Humans, Lung Neoplasms pathology, Models, Biological, Phosphopeptides metabolism, Phosphorylation, Protein Binding, Protein Domains, Proto-Oncogene Proteins chemistry, Carcinoma, Non-Small-Cell Lung metabolism, Cell Transformation, Neoplastic metabolism, DNA, Ribosomal metabolism, Isoenzymes metabolism, Lung Neoplasms metabolism, Pol1 Transcription Initiation Complex Proteins metabolism, Protein Kinase C metabolism, Proto-Oncogene Proteins metabolism, RNA, Ribosomal metabolism
- Abstract
Epithelial cell-transforming sequence 2 (ECT2) is a guanine nucleotide exchange factor for Rho GTPases that is overexpressed in many cancers and involved in signal transduction pathways that promote cancer cell proliferation, invasion, and tumorigenesis. Recently, we demonstrated that a significant pool of ECT2 localizes to the nucleolus of non-small-cell lung cancer (NSCLC) cells, where it binds the transcription factor upstream binding factor 1 (UBF1) on the promoter regions of ribosomal DNA (rDNA) and activates rDNA transcription, transformed cell growth, and tumor formation. Here, we investigated the mechanism by which ECT2 engages UBF1 on rDNA promoters. Results from ECT2 mutagenesis indicated that the tandem BRCT domain of ECT2 mediates binding to UBF1. Biochemical and MS-based analyses revealed that protein kinase Cι (PKCι) directly phosphorylates UBF1 at Ser-412, thereby generating a phosphopeptide-binding epitope that binds the ECT2 BRCT domain. Lentiviral shRNA knockdown and reconstitution experiments revealed that both a functional ECT2 BRCT domain and the UBF1 Ser-412 phosphorylation site are required for UBF1-mediated ECT2 recruitment to rDNA, elevated rRNA synthesis, and transformed growth. Our findings provide critical molecular insight into ECT2-mediated regulation of rDNA transcription in cancer cells and offer a rationale for therapeutic targeting of UBF1- and ECT2-stimulated rDNA transcription for the management of NSCLC., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Justilien et al.)
- Published
- 2020
- Full Text
- View/download PDF
15. Pervasive functional translation of noncanonical human open reading frames.
- Author
-
Chen J, Brunner AD, Cogan JZ, Nuñez JK, Fields AP, Adamson B, Itzhak DN, Li JY, Mann M, Leonetti MD, and Weissman JS
- Subjects
- CRISPR-Cas Systems, Humans, Operon, Ribosomes metabolism, Transcriptome, Open Reading Frames, Peptides genetics, Protein Biosynthesis genetics, RNA, Messenger genetics, RNA, Messenger metabolism
- Abstract
Ribosome profiling has revealed pervasive but largely uncharacterized translation outside of canonical coding sequences (CDSs). In this work, we exploit a systematic CRISPR-based screening strategy to identify hundreds of noncanonical CDSs that are essential for cellular growth and whose disruption elicits specific, robust transcriptomic and phenotypic changes in human cells. Functional characterization of the encoded microproteins reveals distinct cellular localizations, specific protein binding partners, and hundreds of microproteins that are presented by the human leukocyte antigen system. We find multiple microproteins encoded in upstream open reading frames, which form stable complexes with the main, canonical protein encoded on the same messenger RNA, thereby revealing the use of functional bicistronic operons in mammals. Together, our results point to a family of functional human microproteins that play critical and diverse cellular roles., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
16. Chromosome 3q26 Gain Is an Early Event Driving Coordinated Overexpression of the PRKCI, SOX2, and ECT2 Oncogenes in Lung Squamous Cell Carcinoma.
- Author
-
Liu Y, Yin N, Wang X, Khoor A, Sambandam V, Ghosh AB, Fields ZA, Murray NR, Justilien V, and Fields AP
- Subjects
- Carcinogenesis genetics, Carcinogenesis pathology, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cell Proliferation genetics, Cell Transformation, Neoplastic, Gene Dosage, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Lung Neoplasms pathology, Male, Signal Transduction, Transcription, Genetic, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Squamous Cell genetics, Chromosomes, Human, Pair 3 genetics, Isoenzymes genetics, Lung Neoplasms genetics, Oncogenes, Protein Kinase C genetics, Proto-Oncogene Proteins genetics, SOXB1 Transcription Factors genetics
- Abstract
Lung squamous cell carcinoma (LSCC) is a prevalent form of lung cancer exhibiting distinctive histological and genetic characteristics. Chromosome 3q26 copy number gain (CNG) is a genetic hallmark of LSCC present in >90% of tumors. We report that 3q26 CNGs occur early in LSCC tumorigenesis, persist during tumor progression, and drive coordinate overexpression of PRKCI, SOX2, and ECT2. Overexpression of PRKCI, SOX2, and ECT2 in the context of Trp53 loss is sufficient to transform mouse lung basal stem cells into tumors with histological and genomic features of LSCC. Functionally, PRKCI and SOX2 collaborate to activate an extensive transcriptional program that enforces a lineage-restricted LSCC phenotype, whereas PRKCI and ECT2 collaborate to promote oncogenic growth. Gene signatures indicative of PKCι-SOX2 and PKCι-ECT2 signaling activity are enriched in the classical subtype of human LSCC and predict distinct therapeutic vulnerabilities. Thus, the PRKCI, SOX2, and ECT2 oncogenes represent a multigenic driver of LSCC., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. Oncogenic protein kinase Cι signaling mechanisms in lung cancer: Implications for improved therapeutic strategies.
- Author
-
Yin N, Liu Y, Murray NR, and Fields AP
- Subjects
- Humans, Adenocarcinoma of Lung enzymology, Adenocarcinoma of Lung genetics, Carcinoma, Non-Small-Cell Lung enzymology, Carcinoma, Non-Small-Cell Lung genetics, Isoenzymes genetics, Isoenzymes metabolism, Lung Neoplasms enzymology, Lung Neoplasms genetics, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Protein Kinase C genetics, Protein Kinase C metabolism, Signal Transduction
- Abstract
Protein Kinase Cι (PKCι) is a major oncogene involved in the initiation, maintenance and progression of numerous forms of human cancer. In the lung, PKCι is necessary for the maintenance of the transformed phenotype of the two major forms of non-small cell lung cancer (NSCLC), lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC). In addition, PKCι is necessary for both LADC and LSCC tumorigenesis by establishing and maintaining a highly aggressive stem-like, tumor-initiating cell phenotype. Interestingly however, while PKCι signaling in these two major lung cancer subtypes shares some common elements, it also drives distinct, sub-type specific pathways. Furthermore, recent analysis has revealed both PKCι-dependent and PKCι-independent pathways to LADC development. Herein, we discussion our current knowledge of oncogenic PKCι signaling in LADC and LSCC, and discuss these findings in the context of how they may inform strategies for improved therapeutic intervention in these deadly diseases., Competing Interests: Declaration of competing interest The authors have no conflicts of interest to declare., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
18. Protein Kinase Cι and Wnt/β-Catenin Signaling: Alternative Pathways to Kras/Trp53-Driven Lung Adenocarcinoma.
- Author
-
Yin N, Liu Y, Khoor A, Wang X, Thompson EA, Leitges M, Justilien V, Weems C, Murray NR, and Fields AP
- Published
- 2019
- Full Text
- View/download PDF
19. Oncogenic Ect2 signaling regulates rRNA synthesis in NSCLC.
- Author
-
Justilien V, Lewis KC, Murray NR, and Fields AP
- Subjects
- Animals, Carcinogenesis pathology, Carcinoma, Non-Small-Cell Lung pathology, Humans, Lung Neoplasms pathology, Neoplasm Invasiveness, Ribosomes metabolism, Ribosomes pathology, Carcinogenesis metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Lung Neoplasms metabolism, Proto-Oncogene Proteins metabolism, RNA, Neoplasm biosynthesis, RNA, Ribosomal biosynthesis, Signal Transduction
- Abstract
The Rho GTPase family members Rac1, Cdc42 and RhoA play key contributory roles in the transformed phenotype of human cancers. Epithelial Cell Transforming Sequence 2 (Ect2), a guanine nucleotide exchange factor (GEF) for these Rho GTPases, has also been implicated in a variety of human cancers. We have shown that Ect2 is frequently overexpressed in both major forms of non-small cell lung cancer (NSCLC), lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC), which together make up approximately 70% of all lung cancer diagnoses. Furthermore, we have found that Ect2 is required for multiple aspects of the transformed phenotype of NSCLC cells including transformed growth and invasion in vitro and tumorigenesis in vivo . More recently, we showed that a major mechanism by which Ect2 drives KRAS -mediated LADC transformation is by regulating rRNA (rRNA) synthesis. However, it remains unclear whether Ect2 plays a similar role in ribosome biogenesis in LSCC. Here we demonstrate that Ect2 expression correlates positively with expression of ribosome biogenesis genes and with pre-ribosomal 45S RNA abundance in primary LSCC tumors. Furthermore, we demonstrate that Ect2 functionally regulates rRNA synthesis in LSCC cells. Based on these data, we propose that inhibition of Ect2-mediated nucleolar signaling holds promise as a potential therapeutic strategy for improved treatment of both LADC and LSCC.
- Published
- 2019
- Full Text
- View/download PDF
20. Protein kinase C ι : A versatile oncogene in the lung.
- Author
-
Fields AP, Ali SA, Justilien V, and Murray NR
- Abstract
We have recently demonstrated that protein kinase C ι (PKC ι ) promotes a stem-like, tumor-initiating cell phenotype in KRAS -driven lung adenocarcinoma by activating a novel ELF3-NOTCH3 signaling axis.
1 Combined PKC ι and NOTCH inhibition was identified as a novel strategy for the treatment of KRAS -driven lung adenocarcinoma.- Published
- 2018
- Full Text
- View/download PDF
21. A proof-of-concept trial of protein kinase C iota inhibition with auranofin for the paclitaxel-induced acute pain syndrome.
- Author
-
Jatoi A, Grudem ME, Dockter TJ, Block MS, Villasboas JC, Tan A, Deering E, Kasi PM, Mansfield AS, Botero JP, Okuno SH, Smith DR, and Fields AP
- Subjects
- Acute Pain enzymology, Administration, Oral, Antineoplastic Agents, Phytogenic adverse effects, Antineoplastic Agents, Phytogenic therapeutic use, Double-Blind Method, Female, Humans, Male, Middle Aged, Paclitaxel therapeutic use, Peripheral Nervous System Diseases chemically induced, Peripheral Nervous System Diseases drug therapy, Syndrome, Acute Pain chemically induced, Acute Pain drug therapy, Auranofin administration & dosage, Isoenzymes antagonists & inhibitors, Paclitaxel adverse effects, Protein Kinase C antagonists & inhibitors, Protein Kinase Inhibitors administration & dosage
- Abstract
Purpose: Paclitaxel causes the paclitaxel-induced acute pain (PIAP) syndrome. Based on preclinical data, we hypothesized that the protein kinase C (PKC) iota inhibitor, auranofin (a gold salt used for other pain conditions), palliates this pain., Methods: In a randomized, double-blinded manner, patients who had suffered this syndrome were assigned a one-time dose of auranofin 6 mg orally on day #2 of the chemotherapy cycle (post-paclitaxel) versus placebo. Patients completed the Brief Pain Inventory and a pain diary on days 2 through 8 and at the end of the cycle. The primary endpoint was pain scores, as calculated by area under the curve, in response to "Please rate your pain by circling the one number that best describes your pain at its worse in the last 24 hours.", Results: Thirty patients were enrolled. For the primary endpoint, mean area under the curve of 55 units (standard deviation 19) and 61 units (standard deviation 22) were observed in auranofin-treated and placebo-exposed patients, respectively (p = 0.44). On day 8 and at the end of the cycle, pain scores in auranofin-treated patients were more favorable, although differences were not statistically significant., Conclusions: In the dose schedule studied, auranofin did not palliate the PIAP syndrome, but delayed beneficial trends suggest further study for this indication.
- Published
- 2017
- Full Text
- View/download PDF
22. Ect2-Dependent rRNA Synthesis Is Required for KRAS-TRP53-Driven Lung Adenocarcinoma.
- Author
-
Justilien V, Ali SA, Jamieson L, Yin N, Cox AD, Der CJ, Murray NR, and Fields AP
- Subjects
- Adenocarcinoma of Lung, Animals, Auranofin pharmacology, Cell Line, Tumor, Cell Nucleolus metabolism, Cytokinesis, Humans, Isoenzymes physiology, Mice, Nuclear Proteins physiology, Nucleophosmin, Protein Kinase C physiology, Signal Transduction physiology, rac1 GTP-Binding Protein physiology, Adenocarcinoma etiology, Lung Neoplasms etiology, Proto-Oncogene Proteins physiology, Proto-Oncogene Proteins p21(ras) physiology, RNA, Ribosomal biosynthesis, Tumor Suppressor Protein p53 physiology
- Abstract
The guanine nucleotide exchange factor (GEF) epithelial cell transforming sequence 2 (Ect2) has been implicated in cancer. However, it is not clear how Ect2 causes transformation and whether Ect2 is necessary for tumorigenesis in vivo. Here, we demonstrate that nuclear Ect2 GEF activity is required for Kras-Trp53 lung tumorigenesis in vivo and that Ect2-mediated transformation requires Ect2-dependent rDNA transcription. Ect2 activates rRNA synthesis by binding the nucleolar transcription factor upstream binding factor 1 (UBF1) on rDNA promoters and recruiting Rac1 and its downstream effector nucleophosmin (NPM) to rDNA. Protein kinase Cι (PKCι)-mediated Ect2 phosphorylation stimulates Ect2-dependent rDNA transcription. Thus, Ect2 regulates rRNA synthesis through a PKCι-Ect2-Rac1-NPM signaling axis that is required for lung tumorigenesis., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
23. PKCι regulates nuclear YAP1 localization and ovarian cancer tumorigenesis.
- Author
-
Wang Y, Justilien V, Brennan KI, Jamieson L, Murray NR, and Fields AP
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Angiomotins, Animals, Carcinogenesis pathology, Cell Cycle Proteins, Cell Line, Tumor, Female, Heterografts, Humans, Intercellular Signaling Peptides and Proteins metabolism, Isoenzymes genetics, Membrane Proteins metabolism, Mice, Mice, Nude, Microfilament Proteins, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Phosphoproteins genetics, Protein Kinase C genetics, Signal Transduction, Transfection, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Carcinogenesis metabolism, Isoenzymes metabolism, Ovarian Neoplasms metabolism, Phosphoproteins metabolism, Protein Kinase C metabolism
- Abstract
Atypical protein kinase Cι (PKCι) is an oncogene in lung and ovarian cancer. The PKCι gene PRKCI is targeted for frequent tumor-specific copy number gain (CNG) in both lung squamous cell carcinoma (LSCC) and ovarian serous carcinoma (OSC). We recently demonstrated that in LSCC cells PRKCI CNG functions to drive transformed growth and tumorigenicity by activating PKCι-dependent cell autonomous Hedgehog (Hh) signaling. Here, we assessed whether OSC cells harboring PRKCI CNG exhibit similar PKCι-dependent Hh signaling. Surprisingly, we find that whereas PKCι is required for the transformed growth of OSC cells harboring PRKCI CNG, these cells do not exhibit PKCι-dependent Hh signaling or Hh-dependent proliferation. Rather, transformed growth of OSC cells is regulated by PKCι-dependent nuclear localization of the oncogenic transcription factor, YAP1. Lentiviral shRNA-mediated knockdown (KD) of PKCι leads to decreased nuclear YAP1 and increased YAP1 binding to angiomotin (AMOT), which sequesters YAP1 in the cytoplasm. Biochemical analysis reveals that PKCι directly phosphorylates AMOT at a unique site, Thr750, whose phosphorylation inhibits YAP1 binding. Pharmacologic inhibition of PKCι decreases YAP1 nuclear localization and blocks OSC tumor growth in vitro and in vivo. Immunohistochemical analysis reveals a strong positive correlation between tumor PKCι expression and nuclear YAP1 in primary OSC tumor samples, indicating the clinical relevance of PKCι-YAP1 signaling. Our results uncover a novel PKCι-AMOT-YAP1 signaling axis that promotes OSC tumor growth, and provide a rationale for therapeutic targeting of this pathway for treatment of OSC., Competing Interests: The authors declare that they have no conflicts of interest to disclose.
- Published
- 2017
- Full Text
- View/download PDF
24. Targeting oncogenic protein kinase Cι for treatment of mutant KRAS LADC.
- Author
-
Fields AP, Ali SA, Justilien V, and Murray NR
- Subjects
- A549 Cells, Animals, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Line, Tumor, Gold Sodium Thiomalate pharmacology, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Mice, Mutation, Neoplastic Stem Cells drug effects, Sirolimus pharmacology, Treatment Outcome, Xenograft Model Antitumor Assays, Gold Sodium Thiomalate administration & dosage, Isoenzymes metabolism, Lung Neoplasms drug therapy, Protein Kinase C metabolism, Proto-Oncogene Proteins p21(ras) genetics, Sirolimus administration & dosage
- Abstract
Lung cancer is the leading cause of cancer death in the US with ∼124,000 new cases annually, and a 5 y survival rate of ∼16%. Mutant KRAS-driven lung adenocarcinoma (KRAS LADC) is a particularly prevalent and deadly form of lung cancer. Protein kinase Cι (PKCι) is an oncogenic effector of KRAS that activates multiple signaling pathways that stimulate transformed growth and invasion, and maintain a KRAS LADC tumor-initiating cell (TIC) phenotype. PKCι inhibitors used alone and in strategic combination show promise as new therapeutic approaches to treatment of KRAS LADC. These novel drug combinations may improve clinical management of KRAS LADC.
- Published
- 2017
- Full Text
- View/download PDF
25. SOX2 Determines Lineage Restriction: Modeling Lung Squamous Cell Carcinoma in the Mouse.
- Author
-
Murray NR, Justilien V, and Fields AP
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung, Lung Neoplasms genetics, Mice, Oncogenes, Carcinoma, Squamous Cell genetics, SOXB1 Transcription Factors genetics
- Abstract
In this issue of Cancer Cell, Ferone et al. demonstrate that SOX2 not only drives lung tumor formation but also restricts tumor lineage to squamous cell carcinoma (LSCC), regardless of cell of origin. This novel LSCC model should facilitate identification of key oncogenic drivers and treatment strategies for this lung cancer subtype., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation.
- Author
-
Horlbeck MA, Gilbert LA, Villalta JE, Adamson B, Pak RA, Chen Y, Fields AP, Park CY, Corn JE, Kampmann M, and Weissman JS
- Subjects
- Animals, CRISPR-Associated Protein 9, Chromosome Mapping, Humans, Mice, Bacterial Proteins metabolism, Clustered Regularly Interspaced Short Palindromic Repeats, Endonucleases metabolism, Gene Targeting methods, Nucleosomes metabolism, RNA, Guide, CRISPR-Cas Systems metabolism
- Abstract
We recently found that nucleosomes directly block access of CRISPR/Cas9 to DNA (Horlbeck et al., 2016). Here, we build on this observation with a comprehensive algorithm that incorporates chromatin, position, and sequence features to accurately predict highly effective single guide RNAs (sgRNAs) for targeting nuclease-dead Cas9-mediated transcriptional repression (CRISPRi) and activation (CRISPRa). We use this algorithm to design next-generation genome-scale CRISPRi and CRISPRa libraries targeting human and mouse genomes. A CRISPRi screen for essential genes in K562 cells demonstrates that the large majority of sgRNAs are highly active. We also find CRISPRi does not exhibit any detectable non-specific toxicity recently observed with CRISPR nuclease approaches. Precision-recall analysis shows that we detect over 90% of essential genes with minimal false positives using a compact 5 sgRNA/gene library. Our results establish CRISPRi and CRISPRa as premier tools for loss- or gain-of-function studies and provide a general strategy for identifying Cas9 target sites., Competing Interests: MAH: patent application related to CRISPRi and CRISPRa screening (PCT/US15/40449). JSW is a founder of, and MAH and LAG are consultants for, KSQ Therapeutics, a CRISPR functional genomics company. LAG: filed a patent application related to CRISPRi and CRISPRa screening (PCT/US15/40449). JSW is a founder of, and MAH and LAG are consultants for, KSQ Therapeutics, a CRISPR functional genomics company. MK: patent application related to CRISPRi and CRISPRa screening (PCT/US15/40449). JSW: filed a patent application related to CRISPRi and CRISPRa screening (PCT/US15/40449). JSW is a founder of, and MAH and LAG are consultants for, KSQ Therapeutics, a CRISPR functional genomics company. The other authors declare that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
27. Oncogenic PKCι decides tumor-initiating cell fate.
- Author
-
Fields AP, Ali SA, and Murray NR
- Subjects
- Carcinogenesis, Oncogenes, Protein Kinase C, Neoplastic Stem Cells
- Published
- 2016
- Full Text
- View/download PDF
28. Stabilin-1 is expressed in human breast cancer and supports tumor growth in mammary adenocarcinoma mouse model.
- Author
-
Riabov V, Yin S, Song B, Avdic A, Schledzewski K, Ovsiy I, Gratchev A, Llopis Verdiell M, Sticht C, Schmuttermaier C, Schönhaber H, Weiss C, Fields AP, Simon-Keller K, Pfister F, Berlit S, Marx A, Arnold B, Goerdt S, and Kzhyshkowska J
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma pathology, Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Adhesion Molecules, Neuronal genetics, Cell Growth Processes physiology, Disease Models, Animal, Female, HEK293 Cells, Humans, Macrophages metabolism, Macrophages pathology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred BALB C, Mice, Knockout, Receptors, Lymphocyte Homing genetics, Transfection, Adenocarcinoma metabolism, Breast Neoplasms metabolism, Cell Adhesion Molecules, Neuronal biosynthesis, Mammary Neoplasms, Experimental metabolism, Receptors, Lymphocyte Homing biosynthesis
- Abstract
Stabilin-1 is a multifunctional scavenger receptor expressed on alternatively-activated macrophages. Stabilin-1 mediates phagocytosis of "unwanted-self" components, intracellular sorting, and endocytic clearance of extracellular ligands including SPARC that modulates breast cancer growth. The expression of stabilin-1 was found on tumor-associated macrophages (TAM) in mouse and human cancers including melanoma, lymphoma, glioblastoma, and pancreatic insulinoma. Despite its tumor-promoting role in mouse models of melanoma and lymphoma the expression and functional role of stabilin-1 in breast cancer was unknown. Here, we demonstrate that stabilin-1 is expressed on TAM in human breast cancer, and its expression is most pronounced on stage I disease. Using stabilin-1 knockout (ko) mice we show that stabilin-1 facilitates growth of mouse TS/A mammary adenocarcinoma. Endocytosis assay on stabilin-1 ko TAM demonstrated impaired clearance of stabilin-1 ligands including SPARC that was capable of inducing cell death in TS/A cells. Affymetrix microarray analysis on purified TAM and reporter assays in stabilin-1 expressing cell lines demonstrated no influence of stabilin-1 expression on intracellular signalling. Our results suggest stabilin-1 mediated silent clearance of extracellular tumor growth-inhibiting factors (e.g. SPARC) as a mechanism of stabilin-1 induced tumor growth. Silent clearance function of stabilin-1 makes it an attractive candidate for delivery of immunomodulatory anti-cancer therapeutic drugs to TAM., Competing Interests: The authors declare that there are no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
29. Protein Kinase Cι Drives a NOTCH3-dependent Stem-like Phenotype in Mutant KRAS Lung Adenocarcinoma.
- Author
-
Ali SA, Justilien V, Jamieson L, Murray NR, and Fields AP
- Subjects
- Adenocarcinoma of Lung, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, DNA-Binding Proteins metabolism, Humans, Mutation genetics, Neoplastic Stem Cells metabolism, Phenotype, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins c-ets metabolism, Receptor, Notch3, Signal Transduction genetics, Transcription Factors metabolism, Adenocarcinoma genetics, Adenocarcinoma metabolism, Cell Transformation, Neoplastic metabolism, Lung Neoplasms genetics, Lung Neoplasms metabolism, Protein Kinase C metabolism, Proto-Oncogene Proteins p21(ras) genetics, Receptors, Notch metabolism, Stem Cells metabolism
- Abstract
We report that the protein kinase Cι (PKCι) oncogene controls expression of NOTCH3, a key driver of stemness, in KRAS-mediated lung adenocarcinoma (LADC). PKCι activates NOTCH3 expression by phosphorylating the ELF3 transcription factor and driving ELF3 occupancy on the NOTCH3 promoter. PKCι-ELF3-NOTCH3 signaling controls the tumor-initiating cell phenotype by regulating asymmetric cell division, a process necessary for tumor initiation and maintenance. Primary LADC tumors exhibit PKCι-ELF3-NOTCH3 signaling, and combined pharmacologic blockade of PKCι and NOTCH synergistically inhibits tumorigenic behavior in vitro and LADC growth in vivo demonstrating the therapeutic potential of PKCι-ELF3-NOTCH3 signal inhibition to more effectively treat KRAS LADC., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
30. The chromosome 3q26 OncCassette: A multigenic driver of human cancer.
- Author
-
Fields AP, Justilien V, and Murray NR
- Subjects
- Animals, Chromosomes, Human, Pair 3 metabolism, DNA Copy Number Variations, Gene Amplification, Humans, Mice, Neoplasms metabolism, Oncogenes, Chromosomes, Human, Pair 3 genetics, Neoplasms genetics
- Abstract
Recurrent copy number variations (CNVs) are genetic alterations commonly observed in human tumors. One of the most frequent CNVs in human tumors involves copy number gains (CNGs) at chromosome 3q26, which is estimated to occur in >20% of human tumors. The high prevalence and frequent occurrence of 3q26 CNG suggest that it drives the biology of tumors harboring this genetic alteration. The chromosomal region subject to CNG (the 3q26 amplicon) spans from chromosome 3q26 to q29, a region containing ∼200 protein-encoding genes. The large number of genes within the amplicon makes it difficult to identify relevant oncogenic target(s). Whereas a number of genes in this region have been linked to the transformed phenotype, recent studies indicate a high level of cooperativity among a subset of frequently amplified 3q26 genes. Here we use a novel bioinformatics approach to identify potential driver genes within the recurrent 3q26 amplicon in lung squamous cell carcinoma (LSCC). Our analysis reveals a set of 35 3q26 amplicon genes that are coordinately amplified and overexpressed in human LSCC tumors, and that also map to a major LSCC susceptibility locus identified on mouse chromosome 3 that is syntenic with human chromosome 3q26. Pathway analysis reveals that 21 of these genes exist within a single predicted network module. Four 3q26 genes, SOX2, ECT2, PRKCI and PI3KCA occupy the hub of this network module and serve as nodal genes around which the network is organized. Integration of available genetic, genomic, biochemical and functional data demonstrates that SOX2, ECT2, PRKCI and PIK3CA are cooperating oncogenes that function within an integrated cell signaling network that drives a highly aggressive, stem-like phenotype in LSCC tumors harboring 3q26 amplification. Based on the high level of genomic, genetic, biochemical and functional integration amongst these 4 3q26 nodal genes, we propose that they are the key oncogenic targets of the 3q26 amplicon and together define a "3q26 OncCassette" that mediates 3q26 CNG-driven tumorigenesis. Genomic analysis indicates that the 3q26 OncCassette also operates in other major tumor types that exhibit frequent 3q26 CNGs, including head and neck squamous cell carcinoma (HNSCC), ovarian serous cancer and cervical cancer. Finally, we discuss how the 3q26 OncCassette represents a tractable target for development of novel therapeutic intervention strategies that hold promise for improving treatment of 3q26-driven cancers., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
31. A Regression-Based Analysis of Ribosome-Profiling Data Reveals a Conserved Complexity to Mammalian Translation.
- Author
-
Fields AP, Rodriguez EH, Jovanovic M, Stern-Ginossar N, Haas BJ, Mertins P, Raychowdhury R, Hacohen N, Carr SA, Ingolia NT, Regev A, and Weissman JS
- Subjects
- Amino Acid Sequence, Animals, Cells, Cultured, Conserved Sequence, Dendritic Cells drug effects, Humans, Lipopolysaccharides pharmacology, Mice, Open Reading Frames, Regression Analysis, Proteome metabolism, Proteomics methods, Ribosomes metabolism
- Abstract
A fundamental goal of genomics is to identify the complete set of expressed proteins. Automated annotation strategies rely on assumptions about protein-coding sequences (CDSs), e.g., they are conserved, do not overlap, and exceed a minimum length. However, an increasing number of newly discovered proteins violate these rules. Here we present an experimental and analytical framework, based on ribosome profiling and linear regression, for systematic identification and quantification of translation. Application of this approach to lipopolysaccharide-stimulated mouse dendritic cells and HCMV-infected human fibroblasts identifies thousands of novel CDSs, including micropeptides and variants of known proteins, that bear the hallmarks of canonical translation and exhibit translation levels and dynamics comparable to that of annotated CDSs. Remarkably, many translation events are identified in both mouse and human cells even when the peptide sequence is not conserved. Our work thus reveals an unexpected complexity to mammalian translation suited to provide both conserved regulatory or protein-based functions., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. A small molecule inhibitor of atypical protein kinase C signaling inhibits pancreatic cancer cell transformed growth and invasion.
- Author
-
Butler AM, Scotti Buzhardt ML, Erdogan E, Li S, Inman KS, Fields AP, and Murray NR
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic pathology, Humans, Isoenzymes genetics, Neoplasm Invasiveness pathology, Protein Binding, Protein Kinase C genetics, Protein Kinase C metabolism, RNA Interference, RNA, Small Interfering, Signal Transduction drug effects, Adaptor Proteins, Signal Transducing metabolism, Gold Sodium Thiomalate pharmacology, Isoenzymes antagonists & inhibitors, Pancreatic Neoplasms pathology, Protein Kinase C antagonists & inhibitors
- Abstract
Pancreatic cancer is highly resistant to current chemotherapies. Identification of the critical signaling pathways that mediate pancreatic cancer transformed growth is necessary for the development of more effective therapeutic treatments. Recently, we demonstrated that protein kinase C iota (PKCι) and zeta (PKCζ) promote pancreatic cancer transformed growth and invasion, by activating Rac1→ERK and STAT3 signaling pathways, respectively. However, a key question is whether PKCι and PKCζ play redundant (or non-redundant) roles in pancreatic cancer cell transformed growth. Here we describe the novel observations that 1) PKCι and PKCζ are non-redundant in the context of the transformed growth of pancreatic cancer cells; 2) a gold-containing small molecule known to disrupt the PKCι/Par6 interaction, aurothiomalate, also disrupts PKCζ/Par6 interaction; 3) aurothiomalate inhibits downstream signaling of both PKCι and PKCζ, and blocks transformed growth of pancreatic cancer cells in vitro; and 4) aurothiomalate inhibits pancreatic cancer tumor growth and metastasis in vivo. Taken together, these data provide convincing evidence that an inhibitor of atypical PKC signaling inhibits two key oncogenic signaling pathways, driven non-redundantly by PKCι and PKCζ, to significantly reduce tumor growth and metastasis. Our results demonstrate that inhibition of atypical PKC signaling is a promising therapeutic strategy to treat pancreatic cancer.
- Published
- 2015
- Full Text
- View/download PDF
33. Immunogenetics. Dynamic profiling of the protein life cycle in response to pathogens.
- Author
-
Jovanovic M, Rooney MS, Mertins P, Przybylski D, Chevrier N, Satija R, Rodriguez EH, Fields AP, Schwartz S, Raychowdhury R, Mumbach MR, Eisenhaure T, Rabani M, Gennert D, Lu D, Delorey T, Weissman JS, Carr SA, Hacohen N, and Regev A
- Subjects
- Amino Acids chemistry, Amino Acids metabolism, Animals, Cell Culture Techniques, Isotope Labeling methods, Lipopolysaccharides immunology, Mice, Mitochondrial Proteins metabolism, RNA, Messenger biosynthesis, RNA, Messenger genetics, Sequence Analysis, RNA, Bone Marrow Cells immunology, Dendritic Cells immunology, Host-Pathogen Interactions immunology, Molecular Dynamics Simulation, Protein Biosynthesis, Proteolysis
- Abstract
Protein expression is regulated by the production and degradation of messenger RNAs (mRNAs) and proteins, but their specific relationships remain unknown. We combine measurements of protein production and degradation and mRNA dynamics so as to build a quantitative genomic model of the differential regulation of gene expression in lipopolysaccharide-stimulated mouse dendritic cells. Changes in mRNA abundance play a dominant role in determining most dynamic fold changes in protein levels. Conversely, the preexisting proteome of proteins performing basic cellular functions is remodeled primarily through changes in protein production or degradation, accounting for more than half of the absolute change in protein molecules in the cell. Thus, the proteome is regulated by transcriptional induction for newly activated cellular functions and by protein life-cycle changes for remodeling of preexisting functions., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
34. Protein kinase D1 drives pancreatic acinar cell reprogramming and progression to intraepithelial neoplasia.
- Author
-
Liou GY, Döppler H, Braun UB, Panayiotou R, Scotti Buzhardt M, Radisky DC, Crawford HC, Fields AP, Murray NR, Wang QJ, Leitges M, and Storz P
- Subjects
- Acinar Cells drug effects, Animals, Carcinoma in Situ pathology, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic pathology, Mice, Inbred C57BL, Pancreatic Ducts drug effects, Pancreatic Ducts pathology, Pancreatic Neoplasms pathology, Phenotype, Proto-Oncogene Proteins p21(ras) metabolism, Receptors, Notch metabolism, Transforming Growth Factor alpha pharmacology, Up-Regulation drug effects, Acinar Cells enzymology, Acinar Cells pathology, Carcinoma in Situ enzymology, Cellular Reprogramming drug effects, Disease Progression, Pancreatic Neoplasms enzymology, Protein Kinase C metabolism
- Abstract
The transdifferentiation of pancreatic acinar cells to a ductal phenotype (acinar-to-ductal metaplasia, ADM) occurs after injury or inflammation of the pancreas and is a reversible process. However, in the presence of activating Kras mutations or persistent epidermal growth factor receptor (EGF-R) signalling, cells that underwent ADM can progress to pancreatic intraepithelial neoplasia (PanIN) and eventually pancreatic cancer. In transgenic animal models, ADM and PanINs are initiated by high-affinity ligands for EGF-R or activating Kras mutations, but the underlying signalling mechanisms are not well understood. Here, using a conditional knockout approach, we show that protein kinase D1 (PKD1) is sufficient to drive the reprogramming process to a ductal phenotype and progression to PanINs. Moreover, using 3D explant culture of primary pancreatic acinar cells, we show that PKD1 acts downstream of TGFα and Kras, to mediate formation of ductal structures through activation of the Notch pathway.
- Published
- 2015
- Full Text
- View/download PDF
35. Molecular pathways: novel approaches for improved therapeutic targeting of Hedgehog signaling in cancer stem cells.
- Author
-
Justilien V and Fields AP
- Subjects
- Animals, Humans, Molecular Targeted Therapy, Neoplasms drug therapy, Neoplastic Stem Cells drug effects, Translational Research, Biomedical, Hedgehog Proteins metabolism, Neoplasms metabolism, Neoplastic Stem Cells metabolism, Signal Transduction drug effects
- Abstract
The Hedgehog (Hh) signaling pathway is critical for embryonic development. In adult tissues, Hh signaling is relatively quiescent with the exception of roles in tissue maintenance and repair. Aberrant activation of Hh signaling is implicated in multiple aspects of transformation, including the maintenance of the cancer stem cell (CSC) phenotype. Preclinical studies indicate that CSCs from many tumor types are sensitive to Hh pathway inhibition and that Hh-targeted therapeutics block many aspects of transformation attributed to CSCs, including drug resistance, relapse, and metastasis. However, to date, Hh inhibitors, specifically those targeting Smoothened [such as vismodegib, BMS-833923, saridegib (IPI-926), sonidegib/erismodegib (LDE225), PF-04449913, LY2940680, LEQ 506, and TAK-441], have demonstrated good efficacy as monotherapy in patients with basal cell carcinoma and medulloblastoma, but have shown limited activity in other tumor types. This lack of success is likely due to many factors, including a lack of patient stratification in early trials, cross-talk between Hh and other oncogenic signaling pathways that can modulate therapeutic response, and a limited knowledge of Hh pathway activation mechanisms in CSCs from most tumor types. Here, we discuss Hh signaling mechanisms in the context of human cancer, particularly in the maintenance of the CSC phenotype, and consider new therapeutic strategies that hold the potential to expand considerably the scope and therapeutic efficacy of Hh-directed anticancer therapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
36. A mixed-methods feasibility trial of protein kinase C iota inhibition with auranofin in asymptomatic ovarian cancer patients.
- Author
-
Jatoi A, Radecki Breitkopf C, Foster NR, Block MS, Grudem M, Wahner Hendrickson A, Carlson RE, Barrette B, Karlin N, and Fields AP
- Subjects
- Administration, Oral, Aged, Aged, 80 and over, Biomarkers, Tumor metabolism, CA-125 Antigen metabolism, Drug Administration Schedule, Feasibility Studies, Female, Follow-Up Studies, Humans, Middle Aged, Neoplasm Staging, Ovarian Neoplasms enzymology, Ovarian Neoplasms pathology, Pilot Projects, Prognosis, Survival Rate, Antirheumatic Agents therapeutic use, Auranofin therapeutic use, Isoenzymes antagonists & inhibitors, Ovarian Neoplasms drug therapy, Protein Kinase C antagonists & inhibitors
- Abstract
Purpose: This trial was undertaken (1) to determine the feasibility of enrolling asymptomatic ovarian cancer patients with CA-125 elevation in a trial with the protein kinase C iota (PKCι) inhibitor auranofin and (2) to understand patients' perceptions of CA-125 monitoring., Methods: Asymptomatic ovarian cancer patients with CA-125 elevation received 3 mg auranofin orally twice per day and were evaluated. The patients participated in interviews about CA-125 monitoring., Results: Ten patients were enrolled in slightly over 6 months, exceeding our anticipated accrual rate. Four manifested stable CA-125 levels for 1 month or longer. The median progression-free survival was 2.8 months (95% CI: 1.3-3.8); auranofin was well tolerated. One patient had baseline and monthly CA-125 levels of 5,570, 6,085, 3,511, and 2,230 U/ml, respectively, stopped auranofin because of radiographic progression at 3 months, and manifested an increase in CA-125 to 7,168 U/ml approximately 3 months later. Patient interviews revealed (1) the important role of CA-125 in cancer monitoring, (2) ardent advocacy of CA-125 testing, and (3) an evolution toward CA-125 assuming a life of its own., Conclusions: This study showed the feasibility of enrolling asymptomatic ovarian cancer patients with CA-125 elevation in a trial with auranofin. One patient had a decline in CA-125, suggesting that PKCι inhibition merits further study in ovarian cancer., (© 2014 S. Karger AG, Basel.)
- Published
- 2015
- Full Text
- View/download PDF
37. Protein kinase Cα suppresses Kras-mediated lung tumor formation through activation of a p38 MAPK-TGFβ signaling axis.
- Author
-
Hill KS, Erdogan E, Khoor A, Walsh MP, Leitges M, Murray NR, and Fields AP
- Subjects
- Animals, Bronchioles metabolism, Bronchioles pathology, Cells, Cultured, Disease Models, Animal, Enzyme Activation, Female, Gene Expression Regulation, Neoplastic, Humans, Immunohistochemistry, Inhibitor of Differentiation Proteins genetics, Inhibitor of Differentiation Proteins metabolism, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Protein Kinase C-alpha metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Pulmonary Alveoli metabolism, Pulmonary Alveoli pathology, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells metabolism, Stem Cells pathology, Transforming Growth Factor beta metabolism, WT1 Proteins genetics, WT1 Proteins metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Lung Neoplasms genetics, Protein Kinase C-alpha genetics, Proto-Oncogene Proteins p21(ras) genetics, Signal Transduction genetics, Transforming Growth Factor beta genetics, p38 Mitogen-Activated Protein Kinases genetics
- Abstract
Protein kinase C alpha (PKCα) can activate both pro- and anti-tumorigenic signaling depending upon cellular context. Here, we investigated the role of PKCα in lung tumorigenesis in vivo. Gene expression data sets revealed that primary human non-small lung cancers (NSCLC) express significantly decreased PKCα levels, indicating that loss of PKCα expression is a recurrent event in NSCLC. We evaluated the functional relevance of PKCα loss during lung tumorigenesis in three murine lung adenocarcinoma models (LSL-Kras, LA2-Kras and urethane exposure). Genetic deletion of PKCα resulted in a significant increase in lung tumor number, size, burden and grade, bypass of oncogene-induced senescence, progression from adenoma to carcinoma and a significant decrease in survival in vivo. The tumor promoting effect of PKCα loss was reflected in enhanced Kras-mediated expansion of bronchio-alveolar stem cells (BASCs), putative tumor-initiating cells, both in vitro and in vivo. LSL-Kras/Prkca(-/-) mice exhibited a decrease in phospho-p38 MAPK in BASCs in vitro and in tumors in vivo, and treatment of LSL-Kras BASCs with a p38 inhibitor resulted in increased colony size indistinguishable from that observed in LSL-Kras/Prkca(-/-) BASCs. In addition, LSL-Kras/Prkca(-/-) BASCs exhibited a modest but reproducible increase in TGFβ1 mRNA, and addition of exogenous TGFβ1 to LSL-Kras BASCs results in enhanced growth similar to untreated BASCs from LSL-Kras/Prkca(-/-) mice. Conversely, a TGFβR1 inhibitor reversed the effects of PKCα loss in LSL-Kras/Prkca(-/-) BASCs. Finally, we identified the inhibitors of DNA binding (Id) Id1-3 and the Wilm's Tumor 1 as potential downstream targets of PKCα-dependent tumor suppressor activity in vitro and in vivo. We conclude that PKCα suppresses tumor initiation and progression, at least in part, through a PKCα-p38MAPK-TGFβ signaling axis that regulates tumor cell proliferation and Kras-induced senescence. Our results provide the first direct evidence that PKCα exhibits tumor suppressor activity in the lung in vivo.
- Published
- 2014
- Full Text
- View/download PDF
38. Atypical protein kinase Cι as a human oncogene and therapeutic target.
- Author
-
Parker PJ, Justilien V, Riou P, Linch M, and Fields AP
- Subjects
- Animals, Cell Survival drug effects, Humans, Molecular Targeted Therapy, Neoplasms enzymology, Neoplasms genetics, Neoplasms pathology, Signal Transduction drug effects, Isoenzymes antagonists & inhibitors, Isoenzymes genetics, Neoplasms drug therapy, Oncogenes drug effects, Protein Kinase C antagonists & inhibitors, Protein Kinase C genetics, Protein Kinase Inhibitors therapeutic use
- Abstract
Protein kinase inhibitors represent a major class of targeted therapeutics that has made a positive impact on treatment of cancer and other disease indications. Among the promising kinase targets for further therapeutic development are members of the Protein Kinase C (PKC) family. The PKCs are central components of many signaling pathways that regulate diverse cellular functions including proliferation, cell cycle, differentiation, survival, cell migration, and polarity. Genetic manipulation of individual PKC isozymes has demonstrated that they often fulfill distinct, nonredundant cellular functions. Participation of PKC members in different intracellular signaling pathways reflects responses to varying extracellular stimuli, intracellular localization, tissue distribution, phosphorylation status, and intermolecular interactions. PKC activity, localization, phosphorylation, and/or expression are often altered in human tumors, and PKC isozymes have been implicated in various aspects of transformation, including uncontrolled proliferation, migration, invasion, metastasis, angiogenesis, and resistance to apoptosis. Despite the strong relationship between PKC isozymes and cancer, to date only atypical PKCiota has been shown to function as a bona fide oncogene, and as such is a particularly attractive therapeutic target for cancer treatment. In this review, we discuss the role of PKCiota in transformation and describe mechanism-based approaches to therapeutically target oncogenic PKCiota signaling in cancer., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
39. The PRKCI and SOX2 oncogenes are coamplified and cooperate to activate Hedgehog signaling in lung squamous cell carcinoma.
- Author
-
Justilien V, Walsh MP, Ali SA, Thompson EA, Murray NR, and Fields AP
- Subjects
- Acyltransferases antagonists & inhibitors, Acyltransferases genetics, Animals, Apoptosis, Blotting, Western, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Cell Proliferation, Cell Transformation, Neoplastic genetics, High-Throughput Nucleotide Sequencing, Humans, Immunoenzyme Techniques, Isoenzymes antagonists & inhibitors, Isoenzymes genetics, Lung Neoplasms genetics, Lung Neoplasms metabolism, Mice, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Promoter Regions, Genetic genetics, Protein Kinase C antagonists & inhibitors, Protein Kinase C genetics, RNA, Messenger genetics, RNA, Small Interfering genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, SOXB1 Transcription Factors antagonists & inhibitors, SOXB1 Transcription Factors genetics, Signal Transduction, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Tumor Cells, Cultured, Acyltransferases metabolism, Carcinoma, Squamous Cell pathology, Cell Transformation, Neoplastic pathology, Isoenzymes metabolism, Lung Neoplasms pathology, Protein Kinase C metabolism, SOXB1 Transcription Factors metabolism
- Abstract
We report that two oncogenes coamplified on chromosome 3q26, PRKCI and SOX2, cooperate to drive a stem-like phenotype in lung squamous cell carcinoma (LSCC). Protein kinase Cι (PKCι) phosphorylates SOX2, a master transcriptional regulator of stemness, and recruits it to the promoter of Hedgehog (Hh) acyltransferase (HHAT) that catalyzes the rate-limiting step in Hh ligand production. PKCι-mediated SOX2 phosphorylation is required for HHAT promoter occupancy, HHAT expression, and maintenance of a stem-like phenotype. Primary LSCC tumors coordinately overexpress PKCι, SOX2, and HHAT and require PKCι-SOX2-HHAT signaling to maintain a stem-like phenotype. Thus, PKCι and SOX2 are genetically, biochemically, and functionally linked in LSCC, and together they drive tumorigenesis by establishing a cell-autonomous Hh signaling axis., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. PKCι maintains a tumor-initiating cell phenotype that is required for ovarian tumorigenesis.
- Author
-
Wang Y, Hill KS, and Fields AP
- Subjects
- Animals, Antineoplastic Agents pharmacology, Auranofin pharmacology, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Mice, Inbred C57BL, Mice, Nude, Neoplastic Stem Cells pathology, Phenotype, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Signal Transduction drug effects, Carcinogenesis, Isoenzymes metabolism, Neoplastic Stem Cells physiology, Ovarian Neoplasms enzymology, Ovarian Neoplasms pathology, Protein Kinase C metabolism
- Abstract
Unlabelled: Protein kinase Cι (PKCι) has oncogenic potential and is an attractive therapeutic target for treatment of lung cancer, particularly those tumors that express elevated PKCι. However, whether PKCι is a viable target in ovarian cancer is unknown, and virtually nothing is known about the mechanism by which PKCι drives ovarian tumorigenesis. Here, it is demonstrated that PKCι maintains a tumor-initiating cell (TIC) phenotype that drives ovarian tumorigenesis. A highly tumorigenic population of cells from human ovarian cancer cell lines exhibit cancer stem-like TIC properties, including self-renewal, clonal expansion, expression of stem-related genes, enhanced transformed growth in vitro, and aggressive tumor-initiating activity in vivo. Genetic disruption of PKCι inhibits the proliferation, clonal expansion, anchorage-independent growth, and enhanced tumorigenic properties of ovarian TICs. Biochemical analysis demonstrates that PKCι acts through its oncogenic partner Ect2 to activate a MEK/ERK signaling axis that drives the ovarian TIC phenotype. Genomic analysis reveals that PKCι and Ect2 are coordinately amplified and overexpressed in the majority of primary ovarian serous tumors, and these tumors exhibit evidence of an active PKCι-Ect2 signaling axis in vivo. Finally, this study reveals that auranofin, a potent and selective inhibitor of oncogenic PKCι signaling, inhibits the tumorigenic properties of ovarian TIC cells in vitro and in vivo. These data demonstrate that PKCι is required for a TIC phenotype in ovarian cancer, and that auranofin is an attractive therapeutic option to target deadly ovarian TICs in ovarian cancer patients., Implications: PKCι drives a tumor-initiating cell phenotype in ovarian cancer cells that can be therapeutically targeted with auranofin, a small molecule inhibitor of PKCι signaling.
- Published
- 2013
- Full Text
- View/download PDF
41. Euler buckling and nonlinear kinking of double-stranded DNA.
- Author
-
Fields AP, Meyer EA, and Cohen AE
- Subjects
- Base Pair Mismatch, Elasticity, Models, Molecular, Nucleic Acid Conformation, DNA chemistry
- Abstract
The bending stiffness of double-stranded DNA (dsDNA) at high curvatures is fundamental to its biological activity, yet this regime has been difficult to probe experimentally, and literature results have not been consistent. We created a 'molecular vise' in which base-pairing interactions generated a compressive force on sub-persistence length segments of dsDNA. Short dsDNA strands (<41 base pairs) resisted this force and remained straight; longer strands became bent, a phenomenon called 'Euler buckling'. We monitored the buckling transition via Förster Resonance Energy Transfer (FRET) between appended fluorophores. For low-to-moderate concentrations of monovalent salt (up to ∼150 mM), our results are in quantitative agreement with the worm-like chain (WLC) model of DNA elasticity, without the need to invoke any 'kinked' states. Greater concentrations of monovalent salts or 1 mM Mg(2+) induced an apparent softening of the dsDNA, which was best accounted for by a kink in the region of highest curvature. We tested the effects of all single-nucleotide mismatches on the DNA bending. Remarkably, the propensity to kink correlated with the thermodynamic destabilization of the mismatched DNA relative the perfectly complementary strand, suggesting that the kinked state is locally melted. The molecular vise is exquisitely sensitive to the sequence-dependent linear and nonlinear elastic properties of dsDNA.
- Published
- 2013
- Full Text
- View/download PDF
42. Phase I dose escalation study of the PKCι inhibitor aurothiomalate for advanced non-small-cell lung cancer, ovarian cancer, and pancreatic cancer.
- Author
-
Mansfield AS, Fields AP, Jatoi A, Qi Y, Adjei AA, Erlichman C, and Molina JR
- Subjects
- Aged, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Antineoplastic Agents toxicity, Carcinoma, Non-Small-Cell Lung enzymology, Dose-Response Relationship, Drug, Female, Gold blood, Gold Sodium Thiomalate pharmacokinetics, Gold Sodium Thiomalate therapeutic use, Gold Sodium Thiomalate toxicity, Humans, Injections, Intramuscular, Lung Neoplasms enzymology, Male, Middle Aged, Ovarian Neoplasms enzymology, Pancreatic Neoplasms enzymology, Antineoplastic Agents administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Gold Sodium Thiomalate administration & dosage, Isoenzymes antagonists & inhibitors, Lung Neoplasms drug therapy, Ovarian Neoplasms drug therapy, Pancreatic Neoplasms drug therapy, Protein Kinase C antagonists & inhibitors
- Abstract
Protein kinase C iota (PKCι) is overexpressed in non-small-cell lung cancer, ovarian, and pancreatic cancers, where it plays a critical role in oncogenesis. The gold compound aurothiomalate (ATM) has been shown to inhibit PKCι signaling and exerts potent antitumor activity in preclinical models. We sought to determine the maximum tolerated dose (MTD) of ATM. We conducted a phase I dose escalation trial of ATM in patients with non-small-cell lung cancer, ovarian or pancreatic cancer. Patients received ATM intramuscularly weekly for three cycles (cycle duration 4 weeks) at 25, 50, or 75 mg in a 3+3 design. The dose was not escalated for individual patients. Blood samples were analyzed for elemental gold levels. Patients were evaluated every 4 weeks for toxicity and every 8 weeks for response. Fifteen patients were enrolled in this study. Six patients were treated at 25 mg, seven at 50 mg, and two at 75 mg. There was one dose-limiting toxicity at 25 mg (hypokalemia), one at 50 mg (urinary tract infection), and none at 75 mg. There were three grade 3 hematologic toxicities. The recommended MTD of ATM is 50 mg. Patients received treatment for a median of two cycles (range 1-3). There appeared to be a dose-related accumulation of steady-state plasma concentrations of gold consistent with linear pharmacokinetics. In summary, this phase I study was successful in identifying ATM 50 mg intramuscularly weekly as the MTD. Future clinical investigations targeting PKCι are currently in progress.
- Published
- 2013
- Full Text
- View/download PDF
43. Utility and applications of orthotopic models of human non-small cell lung cancer (NSCLC) for the evaluation of novel and emerging cancer therapeutics.
- Author
-
Justilien V and Fields AP
- Subjects
- Animal Husbandry methods, Animals, Cell Line, Tumor, Humans, Medical Laboratory Science methods, Mice, Carcinoma, Non-Small-Cell Lung therapy, Disease Models, Animal, Lung Neoplasms therapy
- Abstract
Lung cancer is a leading cause of cancer deaths worldwide. Despite advances in chemotherapy, radiation therapy, and surgery, lung cancer continues to have a low 5-year survival rate, highlighting a dire need for more effective means of prevention, diagnosis, prognosis, and treatment. Mouse models that recapitulate the clinical features of advanced human lung cancer are critical for testing novel therapeutic approaches. This unit describes a highly reproducible, easy-to-establish orthotopic murine model of lung cancer, provides methods for in vivo imaging and monitoring of tumor growth, and discusses the usefulness of this model for translational lung cancer research and the development of therapeutic strategies., (Copyright © 2013 John Wiley & Sons, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
44. Protein kinase C zeta regulates human pancreatic cancer cell transformed growth and invasion through a STAT3-dependent mechanism.
- Author
-
Butler AM, Scotti Buzhardt ML, Li S, Smith KE, Fields AP, and Murray NR
- Subjects
- Adenocarcinoma pathology, Animals, Cell Line, Tumor, Cell Survival, Gene Knockdown Techniques, Humans, Male, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Transplantation, Pancreatic Neoplasms pathology, Phenotype, Phosphorylation, Protein Processing, Post-Translational, RNA, Small Interfering genetics, Tumor Burden, Adenocarcinoma enzymology, Pancreatic Neoplasms enzymology, Protein Kinase C physiology, STAT3 Transcription Factor metabolism
- Abstract
Pancreatic cancer is a very aggressive disease with few therapeutic options. In this study, we investigate the role of protein kinase C zeta (PKCζ) in pancreatic cancer cells. PKCζ has been shown to act as either a tumor suppressor or tumor promoter depending upon the cellular context. We find that PKCζ expression is either maintained or elevated in primary human pancreatic tumors, but is never lost, consistent with PKCζ playing a promotive role in the pancreatic cancer phenotype. Genetic inhibition of PKCζ reduced adherent growth, cell survival and anchorage-independent growth of human pancreatic cancer cells in vitro. Furthermore, PKCζ inhibition reduced orthotopic tumor size in vivo by inhibiting tumor cell proliferation and increasing tumor necrosis. In addition, PKCζ inhibition reduced tumor metastases in vivo, and caused a corresponding reduction in pancreatic cancer cell invasion in vitro. Signal transducer and activator of transcription 3 (STAT3) is often constitutively active in pancreatic cancer, and plays an important role in pancreatic cancer cell survival and metastasis. Interestingly, inhibition of PKCζ significantly reduced constitutive STAT3 activation in pancreatic cancer cells in vitro and in vivo. Pharmacologic inhibition of STAT3 mimicked the phenotype of PKCζ inhibition, and expression of a constitutively active STAT3 construct rescued the transformed phenotype in PKCζ-deficient cells. We conclude that PKCζ is required for pancreatic cancer cell transformed growth and invasion in vitro and tumorigenesis in vivo, and that STAT3 is an important downstream mediator of the pro-carcinogenic effects of PKCζ in pancreatic cancer cells.
- Published
- 2013
- Full Text
- View/download PDF
45. Overexpression of Evi-1 oncoprotein represses TGF-β signaling in colorectal cancer.
- Author
-
Deng X, Cao Y, Liu Y, Li F, Sambandam K, Rajaraman S, Perkins AS, Fields AP, Hellmich MR, Townsend CM Jr, Thompson EA, and Ko TC
- Subjects
- Animals, Base Sequence, Cell Line, Tumor, Colon metabolism, Colon pathology, Colorectal Neoplasms pathology, DNA-Binding Proteins metabolism, Exons, Gene Expression Regulation, Neoplastic, Humans, MDS1 and EVI1 Complex Locus Protein, Mice, Promoter Regions, Genetic, Rectum metabolism, Rectum pathology, Signal Transduction, Transcription Factors metabolism, Up-Regulation, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, DNA-Binding Proteins genetics, Proto-Oncogenes genetics, Transcription Factors genetics, Transforming Growth Factor beta metabolism
- Abstract
Human colorectal cancer (CRC) cells are resistant to the anti-proliferative effect of transforming growth factor-β (TGF-β), suggesting that disruption of TGF-β signaling plays an important role in colorectal carcinogenesis. Ecotropic virus integration site-1 (Evi-1) oncoprotein represses TGF-β signaling by interacting with Smads, but its role in CRC has not been established. The purpose of this study is to determine whether Evi-1 plays role(s) in CRCs and to characterize Evi-1 transcript(s) in CRCs. Evi-1 was overexpressed in 53% of human CRC samples, 100% of colon adenoma samples, and 100% of human colon cancer cell lines tested. Using 5' RACE, we cloned a novel Evi-1 transcript (Evi-1e) from a human CRC tissue and found that this novel transcript was expressed at a higher level in CRC tissues than in normal tissues and was the major Evi-1 transcript in CRCs. Transient Evi-1 transfection inhibited TGF-β-induced transcriptional activity and reversed the growth inhibitory effect of TGF-β in MC-26 mouse colon cancer cells. In conclusion, we have identified overexpression of Evi-1 oncoprotein as a novel mechanism by which a subset of human CRCs may escape TGF-β regulation. We have also identified a novel Evi-1 transcript, Evi-1e, as the major Evi-1 transcript expressed in human CRCs., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
46. Atypical protein kinase Cι is required for Wnt3a-dependent neurite outgrowth and binds to phosphorylated dishevelled 2.
- Author
-
Greer YE, Fields AP, Brown AM, and Rubin JS
- Subjects
- Casein Kinase I metabolism, Dishevelled Proteins, HEK293 Cells, Hippocampus metabolism, Humans, Isoenzymes metabolism, Microscopy, Fluorescence methods, Models, Biological, Neurons metabolism, Phosphorylation, Protein Kinase C metabolism, Protein Structure, Tertiary, RNA, Small Interfering metabolism, Signal Transduction, Adaptor Proteins, Signal Transducing metabolism, Neurites metabolism, Phosphoproteins metabolism, Protein Kinase C physiology, Wnt3A Protein metabolism
- Abstract
Previously we reported that Wnt3a-dependent neurite outgrowth in Ewing sarcoma family tumor cell lines was mediated by Frizzled3, Dishevelled (Dvl), and c-Jun N-terminal kinase (Endo, Y., Beauchamp, E., Woods, D., Taylor, W. G., Toretsky, J. A., Uren, A., and Rubin, J. S. (2008) Mol. Cell. Biol. 28, 2368-2379). Subsequently, we observed that Dvl2/3 phosphorylation correlated with neurite outgrowth and that casein kinase 1δ, one of the enzymes that mediate Wnt3a-dependent Dvl phosphorylation, was required for neurite extension (Greer, Y. E., and Rubin, J. S. (2011) J. Cell Biol. 192, 993-1004). However, the functional relevance of Dvl phosphorylation in neurite outgrowth was not established. Dvl1 has been shown by others to be important for axon specification in hippocampal neurons via an interaction with atypical PKCζ, but the role of Dvl phosphorylation was not evaluated. Here we report that Ewing sarcoma family tumor cells express PKCι but not PKCζ. Wnt3a stimulated PKCι activation and caused a punctate distribution of pPKCι in the neurites and cytoplasm, with a particularly intense signal at the centrosome. Knockdown of PKCι expression with siRNA reagents blocked neurite formation in response to Wnt3a. Aurothiomalate, a specific inhibitor of PKCι/Par6 binding, also suppressed neurite extension. Wnt3a enhanced the co-immunoprecipitation of endogenous PKCι and Dvl2. Although FLAG-tagged wild-type Dvl2 immunoprecipitated with PKCι, a phosphorylation-deficient Dvl2 derivative did not. This derivative also was unable to rescue neurite outgrowth when endogenous Dvl2/3 was suppressed by siRNA (González-Sancho, J. M., Greer, Y. E., Abrahams, C. L., Takigawa, Y., Baljinnyam, B., Lee, K. H., Lee, K. S., Rubin, J. S., and Brown, A. M. (2013) J. Biol. Chem. 288, 9428-9437). Taken together, these results suggest that site-specific Dvl2 phosphorylation is required for Dvl2 association with PKCι. This interaction is likely to be one of the mechanisms essential for Wnt3a-dependent neurite outgrowth.
- Published
- 2013
- Full Text
- View/download PDF
47. Protein kinase C iota as a therapeutic target in alveolar rhabdomyosarcoma.
- Author
-
Kikuchi K, Soundararajan A, Zarzabal LA, Weems CR, Nelon LD, Hampton ST, Michalek JE, Rubin BP, Fields AP, and Keller C
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Chemotherapy, Adjuvant, Drug Synergism, G2 Phase drug effects, Gene Expression Regulation, Neoplastic drug effects, Gold Sodium Thiomalate pharmacology, Gold Sodium Thiomalate therapeutic use, Humans, Isoenzymes deficiency, Isoenzymes genetics, Mice, Protein Kinase C deficiency, Protein Kinase C genetics, RNA Interference, RNA, Small Interfering genetics, Rhabdomyosarcoma, Alveolar metabolism, Rhabdomyosarcoma, Alveolar pathology, Vincristine pharmacology, Vincristine therapeutic use, Isoenzymes metabolism, Molecular Targeted Therapy methods, Protein Kinase C metabolism, Rhabdomyosarcoma, Alveolar drug therapy, Rhabdomyosarcoma, Alveolar enzymology
- Abstract
Alveolar rhabdomyosarcoma is an aggressive pediatric cancer exhibiting skeletal-muscle differentiation. New therapeutic targets are required to improve the dismal prognosis for invasive or metastatic alveolar rhabdomyosarcoma. Protein kinase C iota (PKCι) has been shown to have an important role in tumorigenesis of many cancers, but little is known about its role in rhabdomyosarcoma. Our gene-expression studies in human tumor samples revealed overexpression of PRKCI. We confirmed overexpression of PKCι at the mRNA and protein levels using our conditional mouse model that authentically recapitulates the progression of rhabdomyosarcoma in humans. Inhibition of Prkci by RNA interference resulted in a dramatic decrease in anchorage-independent colony formation. Interestingly, treatment of primary cell cultures using aurothiomalate (ATM), which is a gold-containing classical anti-rheumatic agent and a PKCι-specific inhibitor, resulted in decreased interaction between PKCι and Par6, decreased Rac1 activity and reduced cell viability at clinically relevant concentrations. Moreover, co-treatment with ATM and vincristine (VCR), a microtubule inhibitor currently used in rhabdomyosarcoma treatment regimens, resulted in a combination index of 0.470-0.793 through cooperative accumulation of non-proliferative multinuclear cells in the G2/M phase, indicating that these two drugs synergize. For in vivo tumor growth inhibition studies, ATM demonstrated a trend toward enhanced VCR sensitivity. Overall, these results suggest that PKCι is functionally important in alveolar rhabdomyosarcoma anchorage-independent growth and tumor-cell proliferation and that combination therapy with ATM and microtubule inhibitors holds promise for the treatment of alveolar rhabdomyosarcoma.
- Published
- 2013
- Full Text
- View/download PDF
48. Optimal tracking of a Brownian particle.
- Author
-
Fields AP and Cohen AE
- Subjects
- Computer Simulation, Diffusion, Image Enhancement methods, Image Interpretation, Computer-Assisted methods, Models, Statistical, Molecular Imaging methods, Particle Size
- Abstract
Optical tracking of a fluorescent particle in solution faces fundamental constraints due to Brownian motion, diffraction, and photon shot noise. Background photons and imperfect tracking apparatus further degrade tracking precision. Here we use a model of particle motion to combine information from multiple time-points to improve the localization precision. We derive successive approximations that enable real-time particle tracking with well controlled tradeoffs between precision and computational cost. We present the theory in the context of feedback electrokinetic trapping, though the results apply to optical tracking of any particle subject to diffusion and drift. We use numerical simulations and experimental data to validate the algorithms' performance.
- Published
- 2012
- Full Text
- View/download PDF
49. Matrix metalloproteinase induction of Rac1b, a key effector of lung cancer progression.
- Author
-
Stallings-Mann ML, Waldmann J, Zhang Y, Miller E, Gauthier ML, Visscher DW, Downey GP, Radisky ES, Fields AP, and Radisky DC
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma of Lung, Adenoma enzymology, Adenoma genetics, Adenoma pathology, Aging pathology, Animals, Cell Line, Tumor, Cells, Cultured, Epithelial-Mesenchymal Transition genetics, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms genetics, Mesoderm metabolism, Mice, Mice, Transgenic, Neuropeptides genetics, Oncogenes genetics, Vimentin metabolism, rac GTP-Binding Proteins genetics, rac1 GTP-Binding Protein genetics, Adenocarcinoma enzymology, Adenocarcinoma pathology, Disease Progression, Lung Neoplasms enzymology, Lung Neoplasms pathology, Matrix Metalloproteinase 3 metabolism, Neuropeptides metabolism, rac GTP-Binding Proteins metabolism, rac1 GTP-Binding Protein metabolism
- Abstract
Lung cancer is more deadly than colon, breast, and prostate cancers combined, and treatment improvements have failed to improve prognosis significantly. Here, we identify a critical mediator of lung cancer progression, Rac1b, a tumor-associated protein with cell-transforming properties that are linked to the matrix metalloproteinase (MMP)-induced epithelial-mesenchymal transition (EMT) in lung epithelial cells. We show that expression of mouse Rac1b in lung epithelial cells of transgenic mice stimulated EMT and spontaneous tumor development and that activation of EMT by MMP-induced expression of Rac1b gave rise to lung adenocarcinoma in the transgenic mice through bypassing oncogene-induced senescence. Rac1b is expressed abundantly in stages 1 and 2 of human lung adenocarcinomas and, hence, is an attractive molecular target for the development of new therapies that prevent progression to later-stage lung cancers.
- Published
- 2012
- Full Text
- View/download PDF
50. Matrix metalloproteinase-10 (MMP-10) interaction with tissue inhibitors of metalloproteinases TIMP-1 and TIMP-2: binding studies and crystal structure.
- Author
-
Batra J, Robinson J, Soares AS, Fields AP, Radisky DC, and Radisky ES
- Subjects
- Binding Sites genetics, Binding, Competitive, Catalytic Domain, Crystallography, X-Ray, HEK293 Cells, Humans, Kinetics, Matrix Metalloproteinase 10 genetics, Matrix Metalloproteinase 10 metabolism, Matrix Metalloproteinase 3 chemistry, Matrix Metalloproteinase 3 genetics, Matrix Metalloproteinase 3 metabolism, Models, Molecular, Mutation, Tissue Inhibitor of Metalloproteinase-1 genetics, Tissue Inhibitor of Metalloproteinase-1 metabolism, Tissue Inhibitor of Metalloproteinase-2 genetics, Tissue Inhibitor of Metalloproteinase-2 metabolism, Matrix Metalloproteinase 10 chemistry, Protein Structure, Tertiary, Tissue Inhibitor of Metalloproteinase-1 chemistry, Tissue Inhibitor of Metalloproteinase-2 chemistry
- Abstract
Matrix metalloproteinase 10 (MMP-10, stromelysin-2) is a secreted metalloproteinase with functions in skeletal development, wound healing, and vascular remodeling; its overexpression is also implicated in lung tumorigenesis and tumor progression. To understand the regulation of MMP-10 by tissue inhibitors of metalloproteinases (TIMPs), we have assessed equilibrium inhibition constants (K(i)) of putative physiological inhibitors TIMP-1 and TIMP-2 for the active catalytic domain of human MMP-10 (MMP-10cd) using multiple kinetic approaches. We find that TIMP-1 inhibits the MMP-10cd with a K(i) of 1.1 × 10(-9) M; this interaction is 10-fold weaker than the inhibition of the similar MMP-3 (stromelysin-1) catalytic domain (MMP-3cd) by TIMP-1. TIMP-2 inhibits the MMP-10cd with a K(i) of 5.8 × 10(-9) M, which is again 10-fold weaker than the inhibition of MMP-3cd by this inhibitor (K(i) = 5.5 × 10(-10) M). We solved the x-ray crystal structure of TIMP-1 bound to the MMP-10cd at 1.9 Å resolution; the structure was solved by molecular replacement and refined with an R-factor of 0.215 (R(free) = 0.266). Comparing our structure of MMP-10cd·TIMP-1 with the previously solved structure of MMP-3cd·TIMP-1 (Protein Data Bank entry 1UEA), we see substantial differences at the binding interface that provide insight into the differential binding of stromelysin family members to TIMP-1. This structural information may ultimately assist in the design of more selective TIMP-based inhibitors tailored for specificity toward individual members of the stromelysin family, with potential therapeutic applications.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.