Network


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

Hotspot


Dive into the research topics where Heike Keilhack is active.

Publication


Featured researches published by Heike Keilhack.


Cancer Research | 2004

Activating mutations of the noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia.

Mohamed Bentires-Alj; J. Guillermo Paez; Frank S. David; Heike Keilhack; Balazs Halmos; Katsuhiko Naoki; John M. Maris; Andrea L. Richardson; Alberto Bardelli; David J. Sagarbaker; William G. Richards; Jinyan Du; Luc Girard; John D. Minna; Mignon L. Loh; David E. Fisher; Victor E. Velculescu; Bert Vogelstein; Matthew Meyerson; William R. Sellers; Benjamin G. Neel

The SH2 domain-containing protein-tyrosine phosphatase PTPN11 (Shp2) is required for normal development and is an essential component of signaling pathways initiated by growth factors, cytokines, and extracellular matrix. In many of these pathways, Shp2 acts upstream of Ras. About 50% of patients with Noonan syndrome have germ-line PTPN11 gain of function mutations. Associations between Noonan syndrome and an increased risk of some malignancies, notably leukemia and neuroblastoma, have been reported, and recent data indicate that somatic PTPN11 mutations occur in children with sporadic juvenile myelomonocytic leukemia, myelodysplasic syndrome, B-cell acute lymphoblastic leukemia, and acute myelogenous leukemia (AML). Juvenile myelomonocytic leukemia patients without PTPN11 mutations have either homozygotic NF-1 deletion or activating RAS mutations. Given the role of Shp2 in Ras activation and the frequent mutation of RAS in human tumors, these data raise the possibility that PTPN11 mutations play a broader role in cancer. We asked whether PTPN11 mutations occur in other malignancies in which activating RAS mutations occur at low but significant frequency. Sequencing of PTPN11 from 13 different human neoplasms including breast, lung, gastric, and neuroblastoma tumors and adult AML and acute lymphoblastic leukemia revealed 11 missense mutations. Five are known mutations predicted to result in an activated form of Shp2, whereas six are new mutations. Biochemical analysis confirmed that several of the new mutations result in increased Shp2 activity. Our data demonstrate that mutations in PTPN11 occur at low frequency in several human cancers, especially neuroblastoma and AML, and suggest that Shp2 may be a novel target for antineoplastic therapy.


Nature Biotechnology | 2009

Sensitive multiplexed analysis of kinase activities and activity-based kinase identification

Kazuishi Kubota; Rana Anjum; Yonghao Yu; Ryan C. Kunz; Jannik N. Andersen; Manfred Kraus; Heike Keilhack; Kumiko Nagashima; Stefan Krauss; Cloud P. Paweletz; Ronald C. Hendrickson; Adam S. Feldman; Chin-Lee Wu; John Rush; Judit Villén; Steven P. Gygi

Constitutive activation of one or more kinase signaling pathways is a hallmark of many cancers. Here we extend the previously described mass spectrometry–based KAYAK approach by monitoring kinase activities from multiple signaling pathways simultaneously. This improved single-reaction strategy, which quantifies the phosphorylation of 90 synthetic peptides in a single mass spectrometry run, is compatible with nanogram to microgram amounts of cell lysate. Furthermore, the approach enhances kinase monospecificity through substrate competition effects, faithfully reporting the signatures of many signaling pathways after mitogen stimulation or of basal pathway activation differences across a panel of well-studied cancer cell lines. Hierarchical clustering of activities from related experiments groups peptides phosphorylated by similar kinases together and, when combined with pathway alteration using pharmacological inhibitors, distinguishes underlying differences in potency, off-target effects and genetic backgrounds. Finally, we introduce a strategy to identify the kinase, and even associated protein complex members, responsible for phosphorylation events of interest.


Journal of Biological Chemistry | 2011

Differential Regulation of Endoplasmic Reticulum Stress by Protein Tyrosine Phosphatase 1B and T Cell Protein Tyrosine Phosphatase

Ahmed Bettaieb; Siming Liu; Yannan Xi; Naoto Nagata; Kosuke Matsuo; Izumi Matsuo; Samah Chahed; Jesse Bakke; Heike Keilhack; Tony Tiganis; Fawaz G. Haj

Protein-tyrosine phosphatase 1B (PTP1B) and T cell protein-tyrosine phosphatase (TCPTP) are closely related intracellular phosphatases implicated in the control of glucose homeostasis. PTP1B and TCPTP can function coordinately to regulate protein tyrosine kinase signaling, and PTP1B has been implicated previously in the regulation of endoplasmic reticulum (ER) stress. In this study, we assessed the roles of PTP1B and TCPTP in regulating ER stress in the endocrine pancreas. PTP1B and TCPTP expression was determined in pancreases from chow and high fat fed mice and the impact of PTP1B and TCPTP over- or underexpression on palmitate- or tunicamycin-induced ER stress signaling assessed in MIN6 insulinoma β cells. PTP1B expression was increased, and TCPTP expression decreased in pancreases of mice fed a high fat diet, as well as in MIN6 cells treated with palmitate. PTP1B overexpression or TCPTP knockdown in MIN6 cells mitigated palmitate- or tunicamycin-induced PERK/eIF2α ER stress signaling, whereas PTP1B deficiency enhanced ER stress. Moreover, PTP1B deficiency increased ER stress-induced cell death, whereas TCPTP deficiency protected MIN6 cells from ER stress-induced death. ER stress coincided with the inhibition of Src family kinases (SFKs), which was exacerbated by PTP1B overexpression and largely prevented by TCPTP knockdown. Pharmacological inhibition of SFKs ameliorated the protective effect of TCPTP deficiency on ER stress-induced cell death. These results demonstrate that PTP1B and TCPTP play nonredundant roles in modulating ER stress in pancreatic β cells and suggest that changes in PTP1B and TCPTP expression may serve as an adaptive response for the mitigation of chronic ER stress.


Cancer Research | 2011

PDK1 Attenuation Fails to Prevent Tumor Formation in PTEN-Deficient Transgenic Mouse Models

Katharine Ellwood-Yen; Heike Keilhack; Kaiko Kunii; Brian Dolinski; Yamicia Connor; Kun Hu; Kumiko Nagashima; Erin O'Hare; Yusuf Erkul; Alessandra Di Bacco; Diana Gargano; Nirah H. Shomer; Minilik Angagaw; Erica Leccese; Paula Andrade; Melissa S. Hurd; Myung K. Shin; Thomas F. Vogt; Alan B. Northrup; Ekaterina V. Bobkova; Shailaja Kasibhatla; Roderick T. Bronson; Martin L. Scott; Giulio Draetta; Victoria M. Richon; Nancy E. Kohl; Peter Blume-Jensen; Jannik N. Andersen; Manfred Kraus

PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target.


PLOS ONE | 2011

Regulation of Brown Fat Adipogenesis by Protein Tyrosine Phosphatase 1B

Kosuke Matsuo; Ahmed Bettaieb; Naoto Nagata; Izumi Matsuo; Heike Keilhack; Fawaz G. Haj

Background Protein-tyrosine phosphatase 1B (PTP1B) is a physiological regulator of insulin signaling and energy balance, but its role in brown fat adipogenesis requires additional investigation. Methodology/Principal Findings To precisely determine the role of PTP1B in adipogenesis, we established preadipocyte cell lines from wild type and PTP1B knockout (KO) mice. In addition, we reconstituted KO cells with wild type, substrate-trapping (D/A) and sumoylation-resistant (K/R) PTP1B mutants, then characterized differentiation and signaling in these cells. KO, D/A- and WT-reconstituted cells fully differentiated into mature adipocytes with KO and D/A cells exhibiting a trend for enhanced differentiation. In contrast, K/R cells exhibited marked attenuation in differentiation and lipid accumulation compared with WT cells. Expression of adipogenic markers PPARγ, C/EBPα, C/EBPδ, and PGC1α mirrored the differentiation pattern. In addition, the differentiation deficit in K/R cells could be reversed completely by the PPARγ activator troglitazone. PTP1B deficiency enhanced insulin receptor (IR) and insulin receptor substrate 1 (IRS1) tyrosyl phosphorylation, while K/R cells exhibited attenuated insulin-induced IR and IRS1 phosphorylation and glucose uptake compared with WT cells. In addition, substrate-trapping studies revealed that IRS1 is a substrate for PTP1B in brown adipocytes. Moreover, KO, D/A and K/R cells exhibited elevated AMPK and ACC phosphorylation compared with WT cells. Conclusions These data indicate that PTP1B is a modulator of brown fat adipogenesis and suggest that adipocyte differentiation requires regulated expression of PTP1B.


Cancer Research | 2017

EZH2 Modifies Sunitinib Resistance in Renal Cell Carcinoma by Kinome Reprogramming

Remi Adelaiye-Ogala; Justin Budka; Nur P. Damayanti; Justine V. Arrington; Mary W. Ferris; Chuan-Chih Hsu; Sreenivasulu Chintala; Ashley Orillion; Kiersten Marie Miles; Li Shen; May Elbanna; Eric Ciamporcero; Sreevani Arisa; Piergiorgio Pettazzoni; Giulio Draetta; Mukund Seshadri; Bradley A. Hancock; Milan Radovich; Janaiah Kota; Michael J. Buck; Heike Keilhack; Brian P. McCarthy; Scott Persohn; Paul R. Territo; Yong Zang; Joseph Irudayaraj; Andy Tao; Peter C. Hollenhorst; Roberto Pili

Acquired and intrinsic resistance to receptor tyrosine kinase inhibitors (RTKi) represents a major hurdle in improving the management of clear cell renal cell carcinoma (ccRCC). Recent reports suggest that drug resistance is driven by tumor adaptation via epigenetic mechanisms that activate alternative survival pathways. The histone methyl transferase EZH2 is frequently altered in many cancers, including ccRCC. To evaluate its role in ccRCC resistance to RTKi, we established and characterized a spontaneously metastatic, patient-derived xenograft model that is intrinsically resistant to the RTKi sunitinib, but not to the VEGF therapeutic antibody bevacizumab. Sunitinib maintained its antiangiogenic and antimetastatic activity but lost its direct antitumor effects due to kinome reprogramming, which resulted in suppression of proapoptotic and cell-cycle-regulatory target genes. Modulating EZH2 expression or activity suppressed phosphorylation of certain RTKs, restoring the antitumor effects of sunitinib in models of acquired or intrinsically resistant ccRCC. Overall, our results highlight EZH2 as a rational target for therapeutic intervention in sunitinib-resistant ccRCC as well as a predictive marker for RTKi response in this disease. Cancer Res; 77(23); 6651-66. ©2017 AACR.


Cancer Research | 2010

Abstract 4561: Quantitative phosphoproteomics of an AKT inhibitor in a PTEN-LOF breast model by label free phospho-dMS demonstrates modulation of protein transcription, protein translation, and motility

An Chi; Heike Keilhack; Hongbo Guo; Kumiko Nagashima; Jannik N. Andersen; Cloud P. Paweletz; Ronald C. Hendrickson

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Among the many types of molecular mechanisms that are currently understood, none is more important for the control of cellular faith than the network of protein kinases and phosphatases. The importance of understanding these signaling networks is furthermore highlighted by the fact that virtually all major pharmaceutical companies have significant projects focused on targeted interference against oncogenic kinases as potential anti-tumor therapies. Yet, a systems wide analysis of signaling phosphorylations by these inhibitors in in vivo (i.e. tissue or xenograft models) is cost prohibitive and rarely performed. The use of mass spectrometry is a powerful approach to study complex mixtures, and some techniques have used either chemical or metabolical isotopic labeling strategies to quantify protein differences between cohorts. While these techniques have been shown to be useful and are routinely applied for cell culture systems, they do not extend readily to in vivo studies as these require either combining of samples or are dependent on the duty cycle of the mass spectrometer. Here we report a generally applicable proteomic approach to identify global protein phosphorylation changes in in-vivo systems. This label-free discovery platform, named phospho-differential mass spectrometry (phospho-dMS) does not require complex mixing or pooling strategies or isotope labeling, and instead identifies statistically significant changes in full scan mass spectrometry data. Hence, in-vivo samples can be analyzed individually, allowing the use of longitudinal designs and within-subject data analysis. We demonstrate phospho-dMS on 27 breast tumor tissues samples (3 concentrations, 3 time points, 3 mice each) excised from a PTEN-LOF mouse model. We quantified in excess of 2000 high confidence phosphorylation events. The most prominent protein modules inhibited by compound treatment (P<0.0001) relate to cytoskeletal machinery such as cell polarity and cytoskeletal reorganization, transcription factors, co-activators, and protein translation with known relevance to cancer. For the first time, using quantitative MS, the topology and significance of phosphorylation networks may be investigated in xenograft, GEM models, and tumor tissue samples marking a new era of cancer signaling research Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4561.


Cancer Cell | 2005

Prognostic, therapeutic, and mechanistic implications of a mouse model of leukemia evoked by Shp2 (PTPN11) mutations

M. Golam Mohi; Ifor R. Williams; Charles R. Dearolf; Gordon Chan; Jeffery L. Kutok; Sarah L. Cohen; Kelly Morgan; Christina L. Boulton; Hirokazu Shigematsu; Heike Keilhack; Koichi Akashi; D. Gary Gilliland; Benjamin G. Neel


Journal of Biological Chemistry | 2005

Diverse Biochemical Properties of Shp2 Mutants IMPLICATIONS FOR DISEASE PHENOTYPES

Heike Keilhack; Frank S. David; Malcolm McGregor; Lewis C. Cantley; Benjamin G. Neel


Translational Oncology | 2012

Evidence of mTOR Activation by an AKT-Independent Mechanism Provides Support for the Combined Treatment of PTEN-Deficient Prostate Tumors with mTOR and AKT Inhibitors.

Weisheng Zhang; Brian B. Haines; Clay Efferson; Joe Zhu; Chris Ware; Kaiko Kunii; Jennifer Tammam; Minilik Angagaw; Marlene C. Hinton; Heike Keilhack; Cloud P. Paweletz; Theresa Zhang; Chris Winter; Sriram Sathyanarayanan; Jonathan D. Cheng; Leigh Zawel; Stephen Fawell; Gary Gilliland; Pradip K. Majumder

Collaboration


Dive into the Heike Keilhack's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmed Bettaieb

University of California

View shared research outputs
Top Co-Authors

Avatar

Christina L. Boulton

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Fawaz G. Haj

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giulio Draetta

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge