Network


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

Hotspot


Dive into the research topics where John M. Koomen is active.

Publication


Featured researches published by John M. Koomen.


Cancer Research | 2011

PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression

Kim H. T. Paraiso; Yun Xiang; Vito W. Rebecca; Ethan V. Abel; Y. Ann Chen; A. Cecilia Munko; Elizabeth R. Wood; Inna V. Fedorenko; Vernon K. Sondak; Alexander R. A. Anderson; Antoni Ribas; Maurizia Dalla Palma; Katherine L. Nathanson; John M. Koomen; Jane L. Messina; Keiran S.M. Smalley

This study addresses the role of PTEN loss in intrinsic resistance to the BRAF inhibitor PLX4720. Immunohistochemical staining of a tissue array covering all stages of melanocytic neoplasia (n = 192) revealed PTEN expression to be lost in >10% of all melanoma cases. Although PTEN expression status did not predict for sensitivity to the growth inhibitory effects of PLX4720, it was predictive for apoptosis, with only limited cell death observed in melanomas lacking PTEN expression (PTEN-). Mechanistically, PLX4720 was found to stimulate AKT signaling in the PTEN- but not the PTEN+ cell lines. Liquid chromatography multiple reaction monitoring mass spectrometry (LC-MRM) was performed to identify differences in apoptosis signaling between the two cell line groups. PLX4720 treatment significantly increased BIM expression in the PTEN+ (>14-fold) compared with the PTEN- cell lines (four-fold). A role for PTEN in the regulation of PLX4720-mediated BIM expression was confirmed by siRNA knockdown of PTEN and through reintroduction of PTEN into cells that were PTEN-. Further studies showed that siRNA knockdown of BIM significantly blunted the apoptotic response in PTEN+ melanoma cells. Dual treatment of PTEN- cells with PLX4720 and a PI3K inhibitor enhanced BIM expression at both the mRNA and protein level and increased the level of apoptosis through a mechanism involving AKT3 and the activation of FOXO3a. In conclusion, we have shown for the first time that loss of PTEN contributes to intrinsic BRAF inhibitor resistance via the suppression of BIM-mediated apoptosis.


Bioinformatics | 2005

Feature extraction and quantification for mass spectrometry in biomedical applications using the mean spectrum

Jeffrey S. Morris; Kevin R. Coombes; John M. Koomen; Keith A. Baggerly; Ryuji Kobayashi

MOTIVATION Mass spectrometry yields complex functional data for which the features of scientific interest are peaks. A common two-step approach to analyzing these data involves first extracting and quantifying the peaks, then analyzing the resulting matrix of peak quantifications. Feature extraction and quantification involves a number of interrelated steps. It is important to perform these steps well, since subsequent analyses condition on these determinations. Also, it is difficult to compare the performance of competing methods for analyzing mass spectrometry data since the true expression levels of the proteins in the population are generally not known. RESULTS In this paper, we introduce a new method for feature extraction in mass spectrometry data that uses translation-invariant wavelet transforms and performs peak detection using the mean spectrum. We examine the methods performance through examples and simulation, and demonstrate the advantages of using the mean spectrum to detect peaks. We also describe a new physics-based computer model of mass spectrometry and demonstrate how one may design simulation studies based on this tool to systematically compare competing methods. AVAILABILITY MATLAB scripts to implement the methods described in this paper and R code for the virtual mass spectrometer are available at http://bioinformatics.mdanderson.org/software.html SUPPLEMENTARY INFORMATION http://bioinformatics.mdanderson.org/supplements.html.


Molecular & Cellular Proteomics | 2014

Targeted Peptide Measurements in Biology and Medicine: Best Practices for Mass Spectrometry-based Assay Development Using a Fit-for-Purpose Approach

Steven A. Carr; Susan E. Abbatiello; Bradley L. Ackermann; Christoph H. Borchers; Bruno Domon; Eric W. Deutsch; Russell P. Grant; Andrew N. Hoofnagle; Ruth Hüttenhain; John M. Koomen; Daniel C. Liebler; Tao Liu; Brendan MacLean; D. R. Mani; Elizabeth Mansfield; Hendrik Neubert; Amanda G. Paulovich; Lukas Reiter; Olga Vitek; Ruedi Aebersold; Leigh Anderson; Robert Bethem; Josip Blonder; Emily S. Boja; Julianne Cook Botelho; Michael T. Boyne; Ralph A. Bradshaw; Alma L. Burlingame; Daniel W. Chan; Hasmik Keshishian

Adoption of targeted mass spectrometry (MS) approaches such as multiple reaction monitoring (MRM) to study biological and biomedical questions is well underway in the proteomics community. Successful application depends on the ability to generate reliable assays that uniquely and confidently identify target peptides in a sample. Unfortunately, there is a wide range of criteria being applied to say that an assay has been successfully developed. There is no consensus on what criteria are acceptable and little understanding of the impact of variable criteria on the quality of the results generated. Publications describing targeted MS assays for peptides frequently do not contain sufficient information for readers to establish confidence that the tests work as intended or to be able to apply the tests described in their own labs. Guidance must be developed so that targeted MS assays with established performance can be made widely distributed and applied by many labs worldwide. To begin to address the problems and their solutions, a workshop was held at the National Institutes of Health with representatives from the multiple communities developing and employing targeted MS assays. Participants discussed the analytical goals of their experiments and the experimental evidence needed to establish that the assays they develop work as intended and are achieving the required levels of performance. Using this “fit-for-purpose” approach, the group defined three tiers of assays distinguished by their performance and extent of analytical characterization. Computational and statistical tools useful for the analysis of targeted MS results were described. Participants also detailed the information that authors need to provide in their manuscripts to enable reviewers and readers to clearly understand what procedures were performed and to evaluate the reliability of the peptide or protein quantification measurements reported. This paper presents a summary of the meeting and recommendations.


Nature Chemical Biology | 2010

A chemical and phosphoproteomic characterization of dasatinib action in lung cancer

Jiannong Li; Uwe Rix; Bin Fang; Yun Bai; Arthur Edwards; Jacques Colinge; Keiryn L. Bennett; Jingchun Gao; Lanxi Song; Steven Eschrich; Giulio Superti-Furga; John M. Koomen; Eric B. Haura

We describe a strategy to comprehend signaling pathways active in lung cancer cells and targeted by dasatinib employing chemical proteomics to identify direct interacting proteins combined with immunoaffinity purification of tyrosine phosphorylated peptides corresponding to activated tyrosine kinases. We identified nearly 40 different kinase targets of dasatinib. These include SFK members (LYN, SRC, FYN, LCK, YES), non-receptor tyrosine kinases (FRK, BRK, ACK), and receptor tyrosine kinases (Ephrin receptors, DDR1, EGFR). Using quantitative phosphoproteomics we identified peptides corresponding to autophosphorylation sites of these tyrosine kinases that are inhibited in a concentration-dependent manner by dasatinib. Using drug resistant gatekeeper mutants, we show that SFK kinases, particularly SRC and FYN, as well as EGFR are relevant targets for dasatinib action. The combined mass spectrometry based approach described here provides a system-level view of dasatinib action in cancer cells and suggests both functional targets and rationale combinatorial therapeutic strategies.


Oncogene | 2005

Induction of p21 by p53 following DNA damage inhibits both Cdk4 and Cdk2 activities.

Guangan He; Zahid H. Siddik; Zaifeng Huang; Ruoning Wang; John M. Koomen; Ryuji Kobayashi; Abdul R. Khokhar; Jian Kuang

DNA damage often activates the p53–p21 pathway and causes G1-phase arrest in mammalian cells. Although there is ample evidence that p21 induction by p53 leads to Cdk2 inhibition, it is unclear whether this checkpoint event also leads to Cdk4 inhibition. Diaminocyclohexane(trans-diacetato)(dichloro) platinum(IV) (DAP), a platinum-based coordination complex, is a DNA-damaging agent that is effective against a variety of tumor cells resistant to the parental drug cisplatin. Our previous studies established that treatment of human cancer cells with low effective concentrations of DAP specifically activates the G1-phase checkpoint and simultaneously inhibit Cdk4 and Cdk2 activities. Here we demonstrate that DAP treatment of human cancer cells activates the p53–p21 pathway without activating other known mechanisms that inhibit Cdk4 and Cdk2 activities. The induced p21 binds to both the Cdk4/cyclin D and Cdk2/cyclin E complexes and inhibits both of their kinase activities. Conversely, inhibition of p21 induction by cycloheximide or by p21 gene deletion prevents DAP-induced inhibition of Cdk4 and Cdk2 activities. Attenuated p53 expression and p21 induction also eliminates DAP-induced G1-phase arrest and inhibition of Cdk4 and Cdk2 activities. Together, these findings establish that activation of the p53–p21 pathway is responsible for the DAP-induced G1-phase checkpoint response and provide the first solid evidence that p21 induction by p53 during a DNA damage-induced G1-phase checkpoint response inhibits both Cdk4 and Cdk2 activities.


Journal of Biological Chemistry | 2004

Comparative Analyses of the Three-dimensional Structures and Enzymatic Properties of α, β, γ, and δ Isoforms of Ca2+-Calmodulin-dependent Protein Kinase II

Tara R. Gaertner; Steven J. Kolodziej; Dan Wang; Ryuji Kobayashi; John M. Koomen; James K. Stoops; M. Neal Waxham

Ca2+-calmodulin-dependent protein kinase II (CaM-kinase II) is a ubiquitous Ser/Thr-directed protein kinase that is expressed from a family of four genes (α, β, γ, and δ) in mammalian cells. We have documented the three-dimensional structures and the biophysical and enzymatic properties of the four gene products. Biophysical analyses showed that each isoform assembles into oligomeric forms and their three-dimensional structures at 21–25 Å revealed that all four isoforms were dodecamers with similar but highly unusual architecture. A gear-shaped core comprising the association domain has the catalytic domains tethered on appendages, six of which extend from both ends of the core. At this level of resolution, we can discern no isoform-dependent differences in ultrastructure of the holoenzymes. Enzymatic analyses showed that the isoforms were similar in their Km for ATP and the peptide substrate syntide, but showed significant differences in their interactions with Ca2+-calmodulin as assessed by binding, substrate phosphorylation, and autophosphorylation. Interestingly, the rank order of CaM binding affinity (γ > β > δ > α) does not directly correlate with the rank order of their CaM dependence for autophosphorylation (β > γ > δ > α). Simulations utilizing this data revealed that the measured differences in CaM binding affinities play a minor role in the autophosphorylation of the enzyme, which is largely dictated by the rate of autophosphorylation for each isoform.


Cancer | 2004

Pharmacoproteomic analysis of prechemotherapy and postchemotherapy plasma samples from patients receiving neoadjuvant or adjuvant chemotherapy for breast carcinoma.

Lajos Pusztai; Betsy Gregory; Keith A. Baggerly; Bo Peng; John M. Koomen; Henry M. Kuerer; Francisco J. Esteva; W. Fraser Symmans; Peter Wagner; Gabriel N. Hortobagyi; Christine Laronga; O. John Semmes; George L. Wright; Richard R. Drake; Antonia Vlahou

In this study, proteomic changes were examined in response to paclitaxel chemotherapy or 5‐fluorouracil, doxorubicin, and cyclophosphamide (FAC) chemotherapy in plasma from patients with Stage I–III breast carcinoma. The authors also compared the plasma profiles of patients with cancer with the plasma profiles of healthy women to identify breast carcinoma–associated protein markers.


Clinical Cancer Research | 2012

The HSP90 Inhibitor XL888 Overcomes BRAF Inhibitor Resistance Mediated through Diverse Mechanisms

Kim H. T. Paraiso; H. E. Haarberg; Elizabeth R. Wood; Vito W. Rebecca; Yian A. Chen; Yun Xiang; Antoni Ribas; Roger S. Lo; Jeffrey S. Weber; Vernon K. Sondak; Jobin K. John; Amod A. Sarnaik; John M. Koomen; Keiran S.M. Smalley

Purpose: The clinical use of BRAF inhibitors is being hampered by the acquisition of drug resistance. This study shows the potential therapeutic use of the HSP90 inhibitor (XL888) in six different models of vemurafenib resistance. Experimental Design: The ability of XL888 to inhibit growth and to induce apoptosis and tumor regression of vemurafenib-resistant melanoma cell lines was shown in vitro and in vivo. A novel mass spectrometry–based pharmacodynamic assay was developed to measure intratumoral HSP70 levels following HSP90 inhibition in melanoma cell lines, xenografts, and melanoma biopsies. Mechanistic studies were carried out to determine the mechanism of XL888-induced apoptosis. Results: XL888 potently inhibited cell growth, induced apoptosis, and prevented the growth of vemurafenib-resistant melanoma cell lines in 3-dimensional cell culture, long-term colony formation assays, and human melanoma mouse xenografts. The reversal of the resistance phenotype was associated with the degradation of PDGFRβ, COT, IGFR1, CRAF, ARAF, S6, cyclin D1, and AKT, which in turn led to the nuclear accumulation of FOXO3a, an increase in BIM (Bcl-2 interacting mediator of cell death) expression, and the downregulation of Mcl-1. In most resistance models, XL888 treatment increased BIM expression, decreased Mcl-1 expression, and induced apoptosis more effectively than dual mitogen-activated protein–extracellular signal–regulated kinase/phosphoinositide 3-kinase (MEK/PI3K) inhibition. Conclusions: HSP90 inhibition may be a highly effective strategy at managing the diverse array of resistance mechanisms being reported to BRAF inhibitors and appears to be more effective at restoring BIM expression and downregulating Mcl-1 expression than combined MEK/PI3K inhibitor therapy. Clin Cancer Res; 18(9); 2502–14. ©2012 AACR.


Molecular and Cellular Biology | 2011

SIRT1 deacetylates the DNA methyltransferase 1 (DNMT1) protein and alters its activities.

Lirong Peng; Zhigang Yuan; Hongbo Ling; Kenji Fukasawa; Keith D. Robertson; Nancy Olashaw; John M. Koomen; Jiandong Chen; William S. Lane; Edward Seto

ABSTRACT DNA methylation and histone acetylation/deacetylation are distinct biochemical processes that control gene expression. While DNA methylation is a common epigenetic signal that inhibits gene transcription, histone deacetylation similarly represses transcription but can be both an epigenetic and nonepigenetic phenomenon. Here we report that the histone deacetylase SIRT1 regulates the activities of DNMT1, a key enzyme responsible for DNA methylation. In mass spectrometry analysis, 12 new acetylated lysine sites were identified in DNMT1. SIRT1 physically associates with DNMT1 and can deacetylate acetylated DNMT1 in vitro and in vivo. Interestingly, deacetylation of different lysines on DNMT1 has different effects on the functions of DNMT1. For example, deacetylation of Lys1349 and Lys1415 in the catalytic domain of DNMT1 enhances DNMT1s methyltransferase activity, while deacetylation of lysine residues in the GK linker decreases DNMT1s methyltransferase-independent transcriptional repression function. Furthermore, deacetylation of all identified acetylated lysine sites in DNMT1 abrogates its binding to SIRT1 and impairs its capability to regulate cell cycle G2/M transition. Finally, inhibition of SIRT1 strengthens the silencing effects of DNMT1 on the expression of tumor suppressor genes ER-α and CDH1 in MDA-MB-231 breast cancer cells. Together, these results suggest that SIRT1-mediated deacetylation of DNMT1 is crucial for DNMT1s multiple effects in gene silencing.


PLOS ONE | 2010

Ack1 Mediated AKT/PKB Tyrosine 176 Phosphorylation Regulates Its Activation

Kiran Mahajan; Domenico Coppola; Sridevi Challa; Bin Fang; Y. Ann Chen; Weiwei Zhu; Alexis S. Lopez; John M. Koomen; Robert W. Engelman; Charlene Rivera; Rebecca S. Muraoka-Cook; Jin Q. Cheng; Ernst Schönbrunn; Said M. Sebti; H. Shelton Earp; Nupam P. Mahajan

The AKT/PKB kinase is a key signaling component of one of the most frequently activated pathways in cancer and is a major target of cancer drug development. Most studies have focused on its activation by Receptor Tyrosine Kinase (RTK) mediated Phosphatidylinositol-3-OH kinase (PI3K) activation or loss of Phosphatase and Tensin homolog (PTEN). We have uncovered that growth factors binding to RTKs lead to activation of a non-receptor tyrosine kinase, Ack1 (also known as ACK or TNK2), which directly phosphorylates AKT at an evolutionarily conserved tyrosine 176 in the kinase domain. Tyr176-phosphorylated AKT localizes to the plasma membrane and promotes Thr308/Ser473-phosphorylation leading to AKT activation. Mice expressing activated Ack1 specifically in the prostate exhibit AKT Tyr176-phosphorylation and develop murine prostatic intraepithelial neoplasia (mPINs). Further, expression levels of Tyr176-phosphorylated-AKT and Tyr284-phosphorylated-Ack1 were positively correlated with the severity of disease progression, and inversely correlated with the survival of breast cancer patients. Thus, RTK/Ack1/AKT pathway provides a novel target for drug discovery.

Collaboration


Dive into the John M. Koomen's collaboration.

Top Co-Authors

Avatar

Bin Fang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Eric B. Haura

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Steven Eschrich

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiannong Li

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Eric A. Welsh

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Fumi Kinose

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Uwe Rix

Austrian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Elizabeth R. Wood

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Guolin Zhang

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge