Publications

2023
C. Nardone, B. A. Palanski, D. C. Scott, R. T. Timms, K. W. Barber, X. Gu, A. Mao, Y. Leng, E. V. Watson, B. A. Schulman, P. A. Cole, and S. J. Elledge. 2023. “A central role for regulated protein stability in the control of TFE3 and MITF by nutrients.” Mol. Cell, 83, Pp. 57-73. Publisher's Version
Z. A. Wang, S. D. Whedon, M. Wu, E.A. Brown, A. Anmangandla, L. Regan, K. Lee, J. Du, J.Y. Hong, L. Fairall, T. Kay, H. Lin, Y. Zhao, J.W.R. Schwabe, and P. A. Cole. 2023. “Histone H2B Deacylation Selectivity: Exploring Chromatin’s Dark Matter with an Engineered Sortase .” J. Am. Chem. Soc., 144, Pp. 3360-3364. Publisher's Version
R. Iwase, D. R. Dempsey, S. D. Whedon, H. Jiang, B. A. Palanski, B. Deng, and P. A. Cole. 2023. “Semisynthetic Approach to the Analysis of Tumor Suppressor PTEN Ubiquitination.” J. Am. Chem. Soc., 145, Pp. 6039–6044. Publisher's Version
2022
K. Lee, S. D. Whedon, Z. A. Wang, and P. A. Cole. 2022. “Distinct biochemical properties of the class I histone deacetylase complexes.” Curr Opin Chem Biol, 70, Pp. 102179.
H. Jiang, C. Chiang, Z. Chen, S. Nathan, G. D’Agostino, J. Paulo, G. Song, H. Zhu, S.B. Gabelli, and P. A. Cole. 2022. “Enzymatic Analysis of WWP2 E3 Ubiquitin Ligase Using Protein Microarrays Identifies Autophagy-Related Substrates.” J. Biol. Chem. , 298, Pp. 101854.
H. Bae, T. Viennet, E. Park, N. Chu, A. Salguero, M. J. Eck, H. Arthanari, and P. A. Cole. 2022. “PH domain-mediated autoinhibition and oncogenic activation of Akt.” eLife, 11, Pp. e80148.
D. Min, J. Byun, E.H. Lee, A.A. Khan, C. Liu, O. Loudig, W. Hu, Y. Zhao, M. Herlyn, B. Tycko, P. A. Cole, and B. Ryu. 2022. “Epigenetic Silencing of BMP6 by Sin3A-HDAC1/2 Repressor Complex Drives Melanoma Metastasis via FAM83G/PAWS1.” Mol. Cancer Res., 20, Pp. 217-230.
Z. A. Wang, S. D. Whedon, M. Wu, S. Weng, E.A. Brown, A. Anmangandla, L. Regan, K. Lee, J. Du, J.Y. Hong, L. Fairall, T. Kay, H. Lin, Y. Zhao, J.W.R. Schwabe, and P. A. Cole. 2022. “Histone H2B Deacylation Selectivity: Exploring Chromatin’s Dark Matter with an Engineered Sortase.” J. Am. Chem. Soc., 144, Pp. 3360-3364.
S. Whedon and P. A. Cole. 2022. “KATs off: Biomedical insights from lysine acetyltransferase inhibitors.” Curr. Opin. Chem. Biol., 72, Pp. 102255. Publisher's Version
Antonieta Salguero, Maggie Chen, Aaron Balana, Nam Chu, Hanjie Jiang, Hwan Bae, Brad Palanski, Katharine Wright, Sara Nathan, Heng Zhu, Sandra Gabelli, Matthew Pratt, and Philip A Cole. 2022. “Multifaceted Regulation of Akt by Diverse C-terminal Post-translational Modifications.” ACS Chem. Biol., 17, Pp. 68-76.
2021
Hitoshi Shiota, Artyom A Alekseyenko, Zhipeng A Wang, Ivona Filic, Tatiana M Knox, Nhi M Luong, Yeying Huang, David A Scott, Kristen L Jones, Prafulla C Gokhale, Madeleine E Lemieux, Philip A Cole, Mitzi I Kuroda, and Christopher A French. 2021. “Chemical Screen Identifies Diverse and Novel Histone Deacetylase Inhibitors as Repressors of NUT Function: Implications for NUT Carcinoma Pathogenesis and Treatment.” Mol Cancer Res, 19, 11, Pp. 1818-1830.Abstract
NUT carcinoma (NC), characterized most commonly by the BRD4-NUTM1 fusion, is a rare, aggressive variant of squamous carcinoma with no effective treatment. BRD4-NUT drives growth and maintains the poorly differentiated state of NC by activating pro-growth genes such as MYC, through the formation of massive, hyperacetylated, superenhancer-like domains termed megadomains. BRD4-NUT-mediated hyperacetylation of chromatin is facilitated by the chromatin-targeting tandem bromodomains of BRD4, combined with NUT, which recruits the histone acetyltransferase, p300. Here, we developed a high-throughput small-molecule screen to identify inhibitors of transcriptional activation by NUT. In this dCAS9-based GFP-reporter assay, the strongest hits were diverse histone deacetylase (HDAC) inhibitors. Two structurally unrelated HDAC inhibitors, panobinostat and the novel compound, IRBM6, both repressed growth and induced differentiation of NC cells in proportion to their inhibition of NUT transcriptional activity. These two compounds repressed transcription of megadomain-associated oncogenic genes, such as MYC and SOX2, while upregulating pro-differentiation, non-megadomain-associated genes, including JUN, FOS, and key cell-cycle regulators, such as CDKN1A. The transcriptional changes correlate with depletion of BRD4-NUT from megadomains, and redistribution of the p300/CBP-associated chromatin acetylation mark, H3K27ac, away from megadomains toward regular enhancer regions previously populated by H3K27ac. In NC xenograft models, we demonstrated that suppression of tumor growth by panobinostat was comparable with that of bromodomain inhibition, and when combined they improved both survival and growth suppression. IMPLICATIONS: The findings provide mechanistic and preclinical rationale for the use of HDAC inhibitors, alone or combined with other agents, in the treatment of NUT carcinoma.
Zhiqin Ji, Richard F Clark, Vikram Bhat, T Matthew Hansen, Loren M Lasko, Kenneth D Bromberg, Vlasios Manaves, Mikkel Algire, Ruth Martin, Wei Qiu, Maricel Torrent, Clarissa G Jakob, Hong Liu, Philip A Cole, Ronen Marmorstein, Edward A Kesicki, Albert Lai, and Michael R Michaelides. 2021. “Discovery of spirohydantoins as selective, orally bioavailable inhibitors of p300/CBP histone acetyltransferases.” Bioorg Med Chem Lett, 39, Pp. 127854.Abstract
p300 and CREB-binding protein (CBP) are essential for a multitude of cellular processes. Dysregulation of p300/CBP histone acetyltransferase activity is linked to a broad spectrum of human diseases including cancers. A novel drug-like spirohydantoin (21) has been discovered as a selective orally bioavailable inhibitor of p300/CBP histone acetyltransferase. Lead compound 21 is more potent than the first-in-class lead A-485 in both enzymatic and cellular assays and lacks the off-target inhibition of dopamine and serotonin transporters, that was observed with A-485.
David D Aufhauser, Paul Hernandez, Seth J Concors, Ciaran O'Brien, Zhonglin Wang, Douglas R Murken, Arabinda Samanta, Ulf H Beier, Lauren Krumeich, Tricia R Bhatti, Yanfeng Wang, Guanghui Ge, Liqing Wang, Shayan Cheraghlou, Florence F Wagner, Edward B Holson, Jay H Kalin, Philip A Cole, Wayne W Hancock, and Matthew H Levine. 2021. “HDAC2 targeting stabilizes the CoREST complex in renal tubular cells and protects against renal ischemia/reperfusion injury.” Sci Rep, 11, 1, Pp. 9018.Abstract
Histone/protein deacetylases (HDAC) 1 and 2 are typically viewed as structurally and functionally similar enzymes present within various co-regulatory complexes. We tested differential effects of these isoforms in renal ischemia reperfusion injury (IRI) using inducible knockout mice and found no significant change in ischemic tolerance with HDAC1 deletion, but mitigation of ischemic injury with HDAC2 deletion. Restriction of HDAC2 deletion to the kidney via transplantation or PAX8-controlled proximal renal tubule-specific Cre resulted in renal IRI protection. Pharmacologic inhibition of HDAC2 increased histone acetylation in the kidney but did not extend renal protection. Protein analysis demonstrated increased HDAC1-associated CoREST protein in HDAC2-/- versus WT cells, suggesting that in the absence of HDAC2, increased CoREST complex occupancy of HDAC1 can stabilize this complex. In vivo administration of a CoREST inhibitor exacerbated renal injury in WT mice and eliminated the benefit of HDAC2 deletion. Gene expression analysis of endothelin showed decreased endothelin levels in HDAC2 deletion. These data demonstrate that contrasting effects of HDAC1 and 2 on CoREST complex stability within renal tubules can affect outcomes of renal IRI and implicate endothelin as a potential downstream mediator.
He Huang, Di Zhang, Yejing Weng, Kyle Delaney, Zhanyun Tang, Cong Yan, Shankang Qi, Chao Peng, Philip A Cole, Robert G Roeder, and Yingming Zhao. 2021. “The regulatory enzymes and protein substrates for the lysine β-hydroxybutyrylation pathway.” Sci Adv, 7, 9.Abstract
Metabolism-mediated epigenetic changes represent an adapted mechanism for cellular signaling, in which lysine acetylation and methylation have been the historical focus of interest. We recently discovered a β-hydroxybutyrate-mediated epigenetic pathway that couples metabolism to gene expression. However, its regulatory enzymes and substrate proteins remain unknown, hindering its functional study. Here, we report that the acyltransferase p300 can catalyze the enzymatic addition of β-hydroxybutyrate to lysine (Kbhb), while histone deacetylase 1 (HDAC1) and HDAC2 enzymatically remove Kbhb. We demonstrate that p300-dependent histone Kbhb can directly mediate in vitro transcription. Moreover, a comprehensive analysis of Kbhb substrates in mammalian cells has identified 3248 Kbhb sites on 1397 substrate proteins. The dependence of histone Kbhb on p300 argues that enzyme-catalyzed acylation is the major mechanism for nuclear Kbhb. Our study thus reveals key regulatory elements for the Kbhb pathway, laying a foundation for studying its roles in diverse cellular processes.
Daniel R Dempsey, Thibault Viennet, Reina Iwase, Eun Young Park, Stephanie Henriquez, Zan Chen, Jeliazko R Jeliazkov, Brad A Palanski, Kim L Phan, Paul Coote, Jeffrey J Gray, Michael J Eck, Sandra B Gabelli, Haribabu Arthanari, and Philip A Cole. 2021. “The structural basis of PTEN regulation by multi-site phosphorylation.” Nat Struct Mol Biol, 28, 10, Pp. 858-868.Abstract
Phosphatase and tensin homolog (PTEN) is a phosphatidylinositol-3,4,5-triphosphate (PIP3) phospholipid phosphatase that is commonly mutated or silenced in cancer. PTEN's catalytic activity, cellular membrane localization and stability are orchestrated by a cluster of C-terminal phosphorylation (phospho-C-tail) events on Ser380, Thr382, Thr383 and Ser385, but the molecular details of this multi-faceted regulation have remained uncertain. Here we use a combination of protein semisynthesis, biochemical analysis, NMR, X-ray crystallography and computational simulations on human PTEN and its sea squirt homolog, VSP, to obtain a detailed picture of how the phospho-C-tail forms a belt around the C2 and phosphatase domains of PTEN. We also visualize a previously proposed dynamic N-terminal α-helix and show that it is key for PTEN catalysis but disordered upon phospho-C-tail interaction. This structural model provides a comprehensive framework for how C-tail phosphorylation can impact PTEN's cellular functions.
Hanjie Jiang, Daniel R Dempsey, and Philip A Cole. 2021. “Ubiquitin Ligase Activities of WWP1 Germline Variants K740N and N745S.” Biochemistry.Abstract
WWP1 is an E3 ubiquitin ligase that has been reported to target the tumor suppressor lipid phosphatase PTEN. K740N and N745S are recently identified germline variants of WWP1 that have been linked to PTEN-associated cancers [Lee, Y. R., et al. (2020) ]. These WWP1 variants have been suggested to release WWP1 from its native autoinhibited state, thereby promoting enhanced PTEN ubiquitination as a mechanism for driving cancer. Using purified proteins and enzymatic assays, we investigate the possibility that K740N and N745S WWP1 possess enhanced ubiquitin ligase activity and demonstrate that these variants are similar to the wild type (WT) in both autoubiquitination and PTEN ubiquitination. Furthermore, K740N and N745S WWP1 show dependencies similar to those of WT in terms of allosteric activation by an engineered ubiquitin variant, upstream E2 concentration, and substrate ubiquitin concentration. Transfected WWP1 WT and mutants demonstrate comparable effects on cellular PTEN levels. These findings challenge the idea that K740N and N745S WWP1 variants promote cancer by enhanced PTEN ubiquitination.
Hanjie Jiang and Philip A Cole. 2021. “N-Terminal Protein Labeling with N-Hydroxysuccinimide Esters and Microscale Thermophoresis Measurements of Protein-Protein Interactions Using Labeled Protein.” Curr Protoc Mol Bio, Pp. e14.Abstract
Protein labeling strategies have been explored for decades to study protein structure, function, and regulation. Fluorescent labeling of a protein enables the study of protein-protein interactions through biophysical methods such as microscale thermophoresis (MST). MST measures the directed motion of a fluorescently labeled protein in response to microscopic temperature gradients, and the protein's thermal mobility can be used to determine binding affinity. However, the stoichiometry and site specificity of fluorescent labeling are hard to control, and heterogeneous labeling can generate inaccuracies in binding measurements. Here, we describe an easy-to-apply protocol for high-stoichiometric, site-specific labeling of a protein at its N-terminus with N-hydroxysuccinimide (NHS) esters as a means to measure protein-protein interaction affinity by MST. This protocol includes guidelines for NHS ester labeling, fluorescent-labeled protein purification, and MST measurement using a labeled protein. As an example of the entire workflow, we additionally provide a protocol for labeling a ubiquitin E3 enzyme and testing ubiquitin E2-E3 enzyme binding affinity. These methods are highly adaptable and can be extended for protein interaction studies in various biological and biochemical circumstances. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Labeling a protein of interest at its N-terminus with NHS esters through stepwise reaction Alternate Protocol: Labeling a protein of interest at its N-terminus with NHS esters through a one-pot reaction Basic Protocol 2: Purifying the N-terminal fluorescent-labeled protein and determining its concentration and labeling efficiency Basic Protocol 3: Using MST to determine the binding affinity of an N-terminal fluorescent-labeled protein to a binding partner. Basic Protocol 4: NHS ester labeling of ubiquitin E3 ligase WWP2 and measurement of the binding affinity between WWP2 and an E2 conjugating enzyme by the MST binding assay.
M. Na, G.M. Martin, D.L. Karl, J. Sun, M. Torres-Martin, H. Itonaga, C. Martinez, S. Chen, Y. Xu, S. Duffort, P.-J. Hamard, C. Chen, L. Cimmino, F.-C. Yang, B. Zucconi, P. A. Cole, M. Xu, M.E. Figueroa, and S.D. Nimer. 2021. “P300 Suppresses the Transition of Myelodysplastic Syndrome to Acute Myeloid Leukemia.” JCI Insights, 6, Pp. e138478.
S.S Pujari, M. Wu, J. Thomforde, Z. A. Wang, C. Chao, N. Olson, L. Erber, W. Pomerantz, P. A. Cole, and N. Tretyakova. 2021. “Site-specific 5-Formyl Cytosine Mediated DNA-Histone Crosslinks: Synthesis and Polymerase Bypass by Human DNA Polymerase η.” Angewandte Chemie, 60, Pp. 26489-26494.
2020
Z. A. Wang and P. A. Cole. 2020. “The Chemical Biology of Reversible Lysine Post-translational Modifications.” Cell Chem Biol.Abstract
Lysine (Lys) residues in proteins undergo a wide range of reversible post-translational modifications (PTMs), which can regulate enzyme activities, chromatin structure, protein-protein interactions, protein stability, and cellular localization. Here we discuss the "writers," "erasers," and "readers" of some of the common protein Lys PTMs and summarize examples of their major biological impacts. We also review chemical biology approaches, from small-molecule probes to protein chemistry technologies, that have helped to delineate Lys PTM functions and show promise for a diverse set of biomedical applications.

Pages