Network


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

Hotspot


Dive into the research topics where Philip J. Papst is active.

Publication


Featured researches published by Philip J. Papst.


Molecular and Cellular Biology | 2004

Protein kinases C and D mediate agonist-dependent cardiac hypertrophy through nuclear export of histone deacetylase 5.

Rick B. Vega; Brooke C. Harrison; Eric Meadows; Charles R. Roberts; Philip J. Papst; Eric N. Olson; Timothy A. McKinsey

ABSTRACT A variety of stress signals stimulate cardiac myocytes to undergo hypertrophy. Persistent cardiac hypertrophy is associated with elevated risk for the development of heart failure. Recently, we showed that class II histone deacetylases (HDACs) suppress cardiac hypertrophy and that stress signals neutralize this repressive function by triggering phosphorylation- and CRM1-dependent nuclear export of these chromatin-modifying enzymes. However, the identities of cardiac HDAC kinases have remained unclear. Here, we demonstrate that signaling by protein kinase C (PKC) is sufficient and, in some cases, necessary to drive nuclear export of class II HDAC5 in cardiomyocytes. Inhibition of PKC prevents nucleocytoplasmic shuttling of HDAC5 in response to a subset of hypertrophic agonists. Moreover, a nonphosphorylatable HDAC5 mutant is refractory to PKC signaling and blocks cardiomyocyte hypertrophy mediated by pharmacological activators of PKC. We also demonstrate that protein kinase D (PKD), a downstream effector of PKC, directly phosphorylates HDAC5 and stimulates its nuclear export. These findings reveal a novel function for the PKC/PKD axis in coupling extracellular cues to chromatin modifications that control cellular growth, and they suggest potential utility for small-molecule inhibitors of this pathway in the treatment of pathological cardiac gene expression.


FEBS Letters | 2001

Hepatic maturation in differentiating embryonic stem cells in vitro

Takashi Hamazaki; Yasuhiko Iiboshi; Masahiro Oka; Philip J. Papst; Amy Meacham; Leonard I. Zon; Naohiro Terada

We investigated the potential of mouse embryonic stem (ES) cells to differentiate into hepatocytes in vitro. Differentiating ES cells expressed endodermal‐specific genes, such as α‐fetoprotein, transthyretin, α 1‐anti‐trypsin and albumin, when cultured without additional growth factors and late differential markers of hepatic development, such as tyrosine aminotransferase (TAT) and glucose‐6‐phosphatase (G6P), when cultured in the presence of growth factors critical for late embryonic liver development. Further, induction of TAT and G6P expression was induced regardless of expression of the functional SEK1 gene, which is thought to provide a survival signal for hepatocytes during an early stage of liver morphogenesis. The data indicate that the in vitro ES differentiation system has a potential to generate mature hepatocytes. The system has also been found useful in analyzing the role of growth factors and intracellular signaling molecules in hepatic development.


Journal of Biological Chemistry | 2006

Canonical Transient Receptor Potential Channels Promote Cardiomyocyte Hypertrophy through Activation of Calcineurin Signaling

Erik Bush; David B. Hood; Philip J. Papst; Joseph A. Chapo; Wayne Minobe; Michael R. Bristow; Eric N. Olson; Timothy A. McKinsey

The calcium/calmodulin-dependent phosphatase calcineurin plays a central role in the control of cardiomyocyte hypertrophy in response to pathological stimuli. Although calcineurin is present at high levels in normal heart, its activity appears to be unaffected by calcium during the course of a cardiac cycle. The mechanism(s) whereby calcineurin is selectively activated by calcium under pathological conditions has remained unclear. Here, we demonstrate that diverse signals for cardiac hypertrophy stimulate expression of canonical transient receptor potential (TRPC) channels. TRPC consists of a family of seven membrane-spanning nonselective cation channels that have been implicated in the nonvoltage-gated influx of calcium in response to G protein-coupled receptor signaling, receptor tyrosine kinase signaling, and depletion of internal calcium stores. TRPC3 expression is up-regulated in multiple rodent models of pathological cardiac hypertrophy, whereas TRPC5 expression is induced in failing human heart. We demonstrate that TRPC promotes cardiomyocyte hypertrophy through activation of calcineurin and its downstream effector, the nuclear factor of activated T cells transcription factor. These results define a novel role for TRPC channels in the control of cardiac growth, and suggest that a TRPC-derived pool of calcium contributes to selective activation of calcineurin in diseased heart.


Journal of Biological Chemistry | 1999

Amino Acid-dependent Control of p70s6k INVOLVEMENT OF tRNA AMINOACYLATION IN THE REGULATION

Yasuhiko Iiboshi; Philip J. Papst; Hideki Kawasome; Hajime Hosoi; Robert T. Abraham; Peter J. Houghton; Naohiro Terada

In human T-lymphoblastoid cells, downstream signaling events of mammalian target of rapamycin (mTOR), including the activity of p70s6k and phosphorylation of eukaryotic initiation factor 4E-binding protein 1, were dependent on amino acid concentration in the culture media, whereas other growth-related protein kinases were not. Amino acid-induced p70s6kactivation was completely inhibited by rapamycin but only partially inhibited by wortmannin. Moreover, amino acid concentration similarly affected the p70s6k activity, which was dependent on a rapamycin-resistant mutant (S2035I) of mTOR. These data indicate that mTOR is required for amino acid-dependent activation of p70s6k. The mechanism by which amino acids regulate p70s6k activity was further explored: 1) amino acid alcohols, which inhibit aminoacylation of tRNA by their competitive binding to tRNA synthetases, suppressed p70s6k activity; 2) suppression of p70s6k by amino acid depletion was blocked by cycloheximide or puromycin, which inhibit utilization of aminoacylated tRNA in cells; and 3) in cells having a temperature-sensitive mutant of histidyl tRNA synthetase, p70s6k was suppressed by a transition of cells to a nonpermissible temperature, which was partially restored by addition of high concentrations of histidine. These results indicate that suppression of tRNA aminoacylation is able to inhibit p70s6k activity. Deacylated tRNA may be a factor negatively regulating p70s6k.


The EMBO Journal | 2000

MEKK2 gene disruption causes loss of cytokine production in response to IgE and c‐Kit ligand stimulation of ES cell‐derived mast cells

Timothy P. Garrington; Tamotsu Ishizuka; Philip J. Papst; Kosuke Chayama; Saiphone Webb; Toshiaki Yujiri; Weiyong Sun; Sue Sather; David Russell; Spencer B. Gibson; Gordon Keller; Erwin W. Gelfand; Gary L. Johnson

Ligation of the high‐affinity IgE receptor (FcϵRI) or of c‐Kit stimulates cytokine production in mast cells. We show that MEK kinase 2 (MEKK2), a MAPK kinase kinase (MAP3K) that regulates the JNK and ERK5 pathways, is required for cytokine production in embryonic stem (ES) cell‐derived mast cells (ESMC). Targeted disruption of the MEKK2 or MEKK1 gene was used to abolish expression of the respective kinases in ESMC. Transcription of specific cytokines in response to IgE or c‐Kit ligand was markedly reduced in MEKK2−/− ESMC relative to wild‐type ESMC. Cytokine production in MEKK1−/− ESMC was similar to that of wild‐type ESMC, demonstrating the specificity of MEKK2 in signaling cytokine gene regulation. MEKK2−/− ESMC also lost receptor‐mediated stimulation of JNK. In contrast, JNK activation in response to UV irradiation was normal, showing that MEKK2 is required for receptor signaling but not for cellular stress responses. MEKK2 is the first MAP3K shown to be required for mast cell tyrosine kinase receptor signaling controlling cytokine gene expression.


Circulation Research | 2001

Role of Phasic Dynamism of p38 Mitogen-Activated Protein Kinase Activation in Ischemic Preconditioning of the Canine Heart

Shoji Sanada; Masafumi Kitakaze; Philip J. Papst; Kazuhito Hatanaka; Hiroshi Asanuma; Toshihiko Aki; Yoshiro Shinozaki; Hisakazu Ogita; Koichi Node; Seiji Takashima; Masanori Asakura; Junko Yamada; Tomi Fukushima; Akiko Ogai; Tsunehiko Kuzuya; Hidezo Mori; Naohiro Terada; Kenichi Yoshida; Masatsugu Hori

Abstract — Although ischemic stress, including ischemic preconditioning (IP), activates p38 mitogen-activated protein kinase (MAPK), the relationship between p38 MAPK activation and the underlying cellular mechanisms of cardioprotection by IP is not verified in vivo. We examined the effects of the selective p38 MAPK inhibition on the cardioprotective effect of IP in the open-chest dogs. The coronary artery was occluded 4 times for 5 minutes, separated by 5 minutes of reperfusion (IP) followed by 90 minutes of occlusion and 6 hours of reperfusion. We infused SB203580 into the coronary artery during IP and 1 hour of reperfusion, during IP alone, and during sustained ischemia in the IP group. p38 MAPK activity markedly increased during IP but did not additionally increase at the onset of ischemia and was even attenuated at 15 minutes of sustained ischemia, and heat-shock protein (HSP) 27 was phosphorylated and translocated from cytosol to myofibril or nucleus without affecting total protein level at the onset of ischemia compared with the control group. SB203580 treatment (1 &mgr;mol/L) only during IP blunted the infarct size limitation by IP (37.3±6.3% versus 7.4±2.1% in the IP group, P <0.01) and attenuated either phosphorylation or translocation of HSP27 during IP. Although the SB203580 treatment throughout the preischemic and postischemic periods had no significant effect on infarct size (33.3±9.4%) in this model, treatment with SB203580 only during ischemia partially mimicked the infarct size limitation by IP (26.8±3.5%). Thus, transient p38 MAPK activation during ischemic preconditioning mainly mediates the cardioprotection followed by HSP27 phosphorylation and translocation in vivo in the canine heart.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Role of MEKK2-MEK5 in the regulation of TNF-α gene expression and MEKK2-MKK7 in the activation of c-Jun N-terminal kinase in mast cells

Kosuke Chayama; Philip J. Papst; Timothy P. Garrington; Joanne C. Pratt; Tamotsu Ishizuka; Saiphone Webb; Soula Ganiatsas; Leonard I. Zon; Weiyong Sun; Gary L. Johnson; Erwin W. Gelfand

Cross-linking of the high-affinity IgE receptor (FcɛRI) on mast cells with IgE and multivalent antigen triggers mitogen-activated protein (MAP) kinase activation and cytokine gene expression. We report here that MAP kinase kinase 4 (MKK4) gene disruption does not affect either MAP kinase activation or cytokine gene expression in response to cross-linking of FcɛRI in embryonic stem cell-derived mast cells. MKK7 is activated in response to cross-linking of FcɛRI, and this activation is inhibited by MAP/ERK kinase (MEK) kinase 2 (MEKK2) gene disruption. In addition, expression of kinase-inactive MKK7 in the murine mast cell line MC/9 inhibits c-Jun NH2-terminal kinase (JNK) activation in response to cross-linking of FcɛRI, whereas expression of kinase-inactive MKK4 does not affect JNK activation by this stimulus. However, FcɛRI-induced activation of the tumor necrosis factor-α (TNF-α) gene promoter is not affected by expression of kinase-inactive MKK7. We describe an alternative pathway by which MEKK2 activates MEK5 and big MAP kinase1/extracellular signal-regulated kinase 5 in addition to MKK7 and JNK, and interruption of this pathway inhibits TNF-α promoter activation. These findings suggest that JNK activation by antigen cross-linking is dependent on the MEKK2-MKK7 pathway, and cytokine production in mast cells is regulated in part by the signaling complex MEKK2-MEK5-ERK5.


Journal of Biological Chemistry | 1998

CDC2-CYCLIN B PHOSPHORYLATES P70 S6 KINASE ON SER411 AT MITOSIS

Philip J. Papst; Hirotaka Sugiyama; Masayuki Nagasawa; Joseph J. Lucas; James L. Maller; Naohiro Terada

The carboxyl terminus of p70 S6 kinase (p70s6k) has a set of Ser and Thr residues (Ser411, Ser418, Ser424, and Thr421) phosphorylated in vivo by an unidentified kinase(s). These Ser/Thr sites are immediately followed by proline, a motif that is commonly seen in the substrates of cyclin-dependent kinases (Cdk) and mitogen-activated protein kinases. A previous study has shown that Cdc2 (Cdk1) indeed phosphorylates these p70s6k Ser/Thr residues in vitro. Here, we demonstrate that Cdc2-cyclin B complex phosphorylates Ser411 in the KIRSPRR sequence, whereas other Cdk-cyclin complexes including those containing Cdk2, Cdk4, or Cdk6 do not. Additionally, Ser411 phosphorylationin vivo was increased at mitosis in parallel with Cdc2 activation, and it was suppressed by a dominant negative form of Cdc2. These data indicate that p70s6k is a physiological substrate of Cdc2-cyclin B in mitosis. Since the activity of p70s6k is low during mitosis, Cdc2-cyclin B may play a role in inactivating p70s6k during mitosis, where protein synthesis is suppressed.


Biochimica et Biophysica Acta | 2009

Suppression of HDAC nuclear export and cardiomyocyte hypertrophy by novel irreversible inhibitors of CRM1.

Lauren G. Monovich; Keith A. Koch; Robin Burgis; Ekundayo Osimboni; Thierry Mann; Daniel Wall; Jinhai Gao; Yan Feng; Richard B. Vega; Benjamin A. Turner; David B. Hood; Andy Law; Philip J. Papst; David Koditek; Joseph A. Chapo; Brian G. Reid; Lawrence Melvin; Nikos Pagratis; Timothy A. McKinsey

Histone deacetylase 5 (HDAC5) represses expression of nuclear genes that promote cardiac hypertrophy. Agonism of a variety of G protein coupled receptors (GPCRs) triggers phosphorylation-dependent nuclear export of HDAC5 via the CRM1 nuclear export receptor, resulting in derepression of pro-hypertrophic genes. A cell-based high-throughput screen of a commercial compound collection was employed to identify compounds with the ability to preserve the nuclear fraction of GFP-HDAC5 in primary cardiomyocytes exposed to GPCR agonists. A hit compound potently inhibited agonist-induced GFP-HDAC5 nuclear export in cultured neonatal rat ventricular myocytes (NRVMs). A small set of related compounds was designed and synthesized to evaluate structure-activity relationship (SAR). The results demonstrated that inhibition of HDAC5 nuclear export was a result of compounds irreversibly reacting with a key cysteine residue in CRM1 that is required for its function. CRM1 inhibition by the compounds also resulted in potent suppression of cardiomyocyte hypertrophy. These studies define a novel class of anti-hypertrophic compounds that function through irreversible inhibition of CRM1-dependent nuclear export.


Cardiovascular Drugs and Therapy | 2000

Inhibition of Nitric Oxide Synthesis Induces Coronary Vascular Remodeling and Cardiac Hypertrophy Associated with the Activation of p70 S6 Kinase in Rats

Tetsuo Minamino; Masafumi Kitakaze; Philip J. Papst; Yasunori Ueda; Yasuhiko Sakata; Hiroshi Asanuma; Akiko Ogai; Tsunehiko Kuzuya; Naohiro Terada; Masatsugu Hori

Chronic inhibition of nitric oxide (NO) synthesis is reported to induce the thickening of coronary artery walls and cardiac hypertrophy in vivo via angiotensin II receptors. Increased protein synthesis is the main feature of these structural changes. Activation of 70 kD S6 kinase (p70S6K) phosphorylates the 40S ribosomal protein S6 that regulates protein synthesis. We examined the role of p70S6K in the vascular and myocardial structural changes induced by the chronic inhibition of NO synthesis. The following 5 groups were studied: untreated Wister-Kyoto rats, those treated with an inhibitor of NO synthase, Nω-nitro-L-arginine methyl ester (L-NAME), those treated with L-NAME and an angiotensin I converting enzyme inhibitor (imidapril), those treated with L-NAME and hydralazine, and those treated with L-NAME and an inhibitor of p70S6K (rapamycin). After 8 weeks, wall-to-lumen ratio in myocardium and cardiomyocyte cross-sectional areas were quantified. L-NAME increased systolic blood pressure, wall-to-lumen ratio, and cardiomyocyte cross-sectional area compared with control animals. Imidapril or rapamycin, but not hydralazine, markedly reduced these structural changes. L-NAME increased p70S6K activity in myocardium compared with control rats. Imidapril or rapamycin prevented the activation of p70S6K activity in myocardium induced by L-NAME. These results suggest that activation of p70S6K plays an important role in coronary vascular remodeling and cardiac hypertrophy induced by the chronic inhibition of nitric oxide synthesis in vivo.

Collaboration


Dive into the Philip J. Papst's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erwin W. Gelfand

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Gary L. Johnson

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Timothy A. McKinsey

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasuhiko Iiboshi

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge