scholarly journals The Kinase Specificity of Protein Kinase Inhibitor Peptide

2021 ◽  
Vol 12 ◽  
Author(s):  
Yao Chen ◽  
Bernardo L. Sabatini

G-protein-coupled-receptor (GPCR) signaling is exquisitely controlled to achieve spatial and temporal specificity. The endogenous protein kinase inhibitor peptide (PKI) confines the spatial and temporal spread of the activity of protein kinase A (PKA), which integrates inputs from three major types of GPCRs. Despite its wide usage as a pharmaceutical inhibitor of PKA, it was unclear whether PKI only inhibits PKA activity. Here, the effects of PKI on 55 mouse kinases were tested in in vitro assays. We found that in addition to inhibiting PKA activity, both PKI (6–22) amide and full-length PKIα facilitated the activation of multiple isoforms of protein kinase C (PKC), albeit at much higher concentrations than necessary to inhibit PKA. Thus, our results call for appropriate interpretation of experimental results using PKI as a pharmaceutical agent. Furthermore, our study lays the foundation to explore the potential functions of PKI in regulating PKC activity and in coordinating PKC and PKA activities.

2020 ◽  
Author(s):  
Yao Chen ◽  
Bernardo L. Sabatini

AbstractG-protein-coupled-receptor (GPCR) signaling is exquisitely controlled to achieve spatial and temporal specificity. The endogenous protein kinase inhibitor peptide (PKI) confines the spatial and temporal spread of the activity of protein kinase A (PKA), which integrates inputs from three major types of GPCRs. Despite its wide usage as a pharmaceutical inhibitor of PKA, it was unclear whether PKI only inhibits PKA activity. Here, the effects of PKI on 55 mouse kinases were tested in in vitro assays. We found that in addition to inhibiting PKA activity, both PKI (6-22) amide and full-length PKIα facilitated the activation of multiple isoforms of protein kinase C (PKC), albeit at much higher concentrations than necessary to inhibit PKA. Thus, our results call for appropriate interpretation of experimental results using PKI as a pharmaceutical agent. Furthermore, our study lays the foundation to explore the potential functions of PKI in regulating PKC activity and in coordinating PKC and PKA activities.


2002 ◽  
Vol 282 (3) ◽  
pp. G461-G469 ◽  
Author(s):  
Ya-Ping Fan ◽  
Rajinder N. Puri ◽  
Satish Rattan

Effect of ANG II was investigated in in vitro smooth muscle strips and in isolated smooth muscle cells (SMC). Among different species, rat internal and sphincter (IAS) smooth muscle showed significant and reproducible contraction that remained unmodified by different neurohumoral inhibitors. The AT1antagonist losartan but not AT2 antagonist PD-123319 antagonized ANG II-induced contraction of the IAS smooth muscle and SMC. ANG II-induced contraction of rat IAS smooth muscle and SMC was attenuated by tyrosine kinase inhibitors genistein and tyrphostin, protein kinase C (PKC) inhibitor H-7, Ca2+ channel blocker nicardipine, Rho kinase inhibitor Y-27632 or p44/42mitogen-activating protein kinase (MAPK44/42) inhibitor PD-98059. Combinations of nicardipine and H-7, Y-27632, and PD-98059 caused further attenuation of the ANG II effects. Western blot analyses revealed the presence of both AT1 and AT2receptors. We conclude that ANG II causes contraction of rat IAS smooth muscle by the activation of AT1 receptors at the SMC and involves multiple intracellular pathways, influx of Ca2+, and activation of PKC, Rho kinase, and MAPK44/42.


2001 ◽  
Vol 2 (3) ◽  
pp. 233-244 ◽  
Author(s):  
Dietmar Böcker ◽  
Eugen J. Verspohl

MAP (mitogen-activated protein) kinase (also called Erk 1/2) plays a crucial role in cell proliferation and differentiation. Its impact on secretory events is less well established. The interplay of protein kinase C (PKC), PI3-kinase nd cellular tyrosine kinase with MAP kinase activity using inhibitors and compounds such as glucose, phorbol 12-myristate 13-acetate (PMA) and agonists of G-protein coupled receptors like gastrin releasing peptide (GRP), oxytocin (OT) and glucose-dependent insulinotropic peptide (GIP) was investigated in INS-1 cells, an insulin secreting cell line. MAP kinase activity was determined by using a peptide derived from the EGF receptor as a MAP kinase substrate and [P32]ATP. Glucose as well as GRP, OT and GIP exhibited a time-dependent increase in MAP kinase activity with a maximum at time point 2.5 min. All further experiments were performed using 2.5 min incubations. The flavone PD 098059 is known to bind to the inactive forms of MEK1 (MAPK/ERK-Kinase) thus preventing activation by upstream activators. 20 μM PD 098059 (IC50=51 μM) inhibited MAP kinase stimulated by either glucose, GRP, OT, GIP or PMA. Inhibiton (“downregulation”) of PKC by a long term (22h) pretreatment with 1 μM PMA did not influence MAP kinase activity when augmented by either of the above mentioned compound. To investigate whether PI3-kinase and cellular tyrosine kinase are involved in G-protein mediated effects on MAP kinase, inhibitors were used: 100 nM wortmannin (PI3-kinase inhibitor) reduced the effects of GRP, OT and GIP but not that of PMA; 100 μM genistein (tyrosine kinase inhibitor) inhibited the stimulatory effect of either above mentioned compound on MAP kinase activation. Inhibition of MAP kinase by 20 μM PD 098059 did not influence insulin secretion modulated by either compound (glucose, GRP, OT or GIP). [H3]Thymidine incorporation, however, was severely inhibited by PD 098059. Thus MAP kinase is important for INS-1 cell proliferation but not for its insulin secretory response with respect to major initiators and modulators of insulin release. The data indicate that MAP kinase is active and under the control of MAP kinase. PKC is upstream of a genisteinsensitive tyrosine kinase and probably downstream of a PI3-kinase in INS-1 cells.


2020 ◽  
Vol 11 ◽  
Author(s):  
Chong Liu ◽  
Ping Ke ◽  
Jingjing Zhang ◽  
Xiaoying Zhang ◽  
Xiongwen Chen

The protein kinase enzyme family plays a pivotal role in almost every aspect of cellular function, including cellular metabolism, division, proliferation, transcription, movement, and survival. Protein kinase A (PKA), whose activation is triggered by cyclic adenosine monophosphate (cAMP), is widely distributed in various systems and tissues throughout the body and highly related to pathogenesis and progression of various kinds of diseases. The inhibition of PKA activation is essential for the study of PKA functions. Protein kinase inhibitor peptide (PKI) is a potent, heat-stable, and specific PKA inhibitor. It has been demonstrated that PKI can block PKA-mediated phosphorylase activation. Since then, researchers have a lot of knowledge about PKI. PKI is considered to be the most effective and specific method to inhibit PKA and is widely used in related research. In this review, we will first introduce the knowledge on the activation of PKA and mechanisms related on the inhibitory effects of PKI on PKA. Then, we will compare PKI-mediated PKA inhibition vs. several popular methods of PKA inhibition.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 474-474 ◽  
Author(s):  
Neil P. Shah ◽  
Corynn Kasap ◽  
Ronald Paquette ◽  
Jorge Cortes ◽  
Javier Pinilla ◽  
...  

Abstract The fusion protein BCR-ABL is a hallmark of chronic myelogenous leukemia (CML) and Philadelphia-positive acute lymphocytic leukemia (Ph+-ALL), and has been demonstrated as the primary driver of these diseases. Control of CML for considerable periods of time has been achieved through use of selective ABL kinase inhibitors, particularly imatinib. Once patients fail imatinib therapy, they are commonly found to harbor a mutated and activated form of BCR-ABL which is unable to bind the inhibitor. A more recent ABL inhibitor, dasatinib, can block growth of cells harboring most of the imatinib-resistant mutations, but T315I and F317L mutations are often seen in patients relapsing after dasatinib therapy. Thus once a patient develops CML harboring these mutations, there are few therapeutic options available. XL228 is a potent multi-targeted protein kinase inhibitor with activity against IGF1R, src, and Abl. It displays low nanomolar biochemical activity against wild type Abl kinase (Ki = 5 nM), as well as the T315I form of Abl resistant to imatinib and dasatinib (Ki = 1.4 nM). XL228 also inhibits Aurora A with an IC50 of approximately 3 nM, demonstrating a more balanced inhibition profile compared to other dual Abl /Aurora inhibitors. CML and ALL cell lines were evaluated for sensitivity to XL228, and in each case the IC50 for inhibition of proliferation was less than 100 nM. XL228 inhibits phosphorylation of BCR-ABL and its substrate STAT5 in K562 cells in vitro with IC50s of 33 and 43 nM, respectively. Single-dose pharmacodynamics studies demonstrate a potent effect of XL228 on BCR-ABL signaling in K562 xenograft tumors. Phosphorylation of BCR-ABL was decreased by 50% at XL228 plasma concentrations of 3.5 μM; a similar decrease in phospho-STAT5 occurred at 0.8 μM plasma concentration. XL228 showed clear superiority to MK-0457, imatinib, and dasatinib in downregulating BCR-ABL phosphorylation in BaF3 cells expressing the T315I form of BCR-ABL in vitro (406 nM, 6912 nM, >10,000 nM, >10,000 nM, respectively), and in xenograft experiments in vivo. These results indicate that XL228 potently inhibits wild type and T315I forms of BCR-ABL, and provide a rationale for the clinical development of this agent for the treatment of patients with drug-resistant disease. A phase I dose escalation study of XL228 in subjects with CML or Ph+-ALL who have failed prior imatinib and dasatinib therapy has been initiated, focusing on safety/tolerability, pharmacodynamics, and pharmacokinetics. Pharmacodynamic assessments include a flow cytometry-based phospho-CrkL assay, quantitative PCR for BCR-ABL, and plasma markers of XL228 activity. An update on our clinical experience with XL228 in subjects resistant or intolerant to imatinib and dasatinib will be presented.


Sign in / Sign up

Export Citation Format

Share Document