1
|
Su Q, Zhang J, Lin W, Zhang JF, Newton AC, Mehta S, Yang J, Zhang J. Sensitive fluorescent biosensor reveals differential subcellular regulation of PKC. Nat Chem Biol 2024:10.1038/s41589-024-01758-3. [PMID: 39394268 DOI: 10.1038/s41589-024-01758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
The protein kinase C (PKC) family of serine and threonine kinases, consisting of three distinctly regulated subfamilies, has been established as critical for various cellular functions. However, how PKC enzymes are regulated at different subcellular locations, particularly at emerging signaling hubs, is unclear. Here we present a sensitive excitation ratiometric C kinase activity reporter (ExRai-CKAR2) that enables the detection of minute changes (equivalent to 0.2% of maximum stimulation) in subcellular PKC activity. Using ExRai-CKAR2 with an enhanced diacylglycerol (DAG) biosensor, we uncover that G-protein-coupled receptor stimulation triggers sustained PKC activity at the endoplasmic reticulum and lysosomes, differentially mediated by Ca2+-sensitive conventional PKC and DAG-sensitive novel PKC, respectively. The high sensitivity of ExRai-CKAR2, targeted to either the cytosol or partitioning defective complexes, further enabled us to detect previously inaccessible endogenous atypical PKC activity in three-dimensional organoids. Taken together, ExRai-CKAR2 is a powerful tool for interrogating PKC regulation in response to physiological stimuli.
Collapse
Affiliation(s)
- Qi Su
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jing Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Wei Lin
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jin-Fan Zhang
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Alexandra C Newton
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sohum Mehta
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jing Yang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Jin Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
2
|
Su Q, Zhang J, Lin W, Zhang JF, Newton AC, Mehta S, Yang J, Zhang J. Sensitive Fluorescent Biosensor Reveals Differential Subcellular Regulation of PKC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587373. [PMID: 38586003 PMCID: PMC10996667 DOI: 10.1101/2024.03.29.587373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The protein kinase C (PKC) family of serine/threonine kinases, which consist of three distinctly regulated subfamilies, have long been established as critical for a variety of cellular functions. However, how PKC enzymes are regulated at different subcellular locations, particularly at emerging signaling hubs such as the ER, lysosome, and Par signaling complexes, is unclear. Here, we present a sensitive Excitation Ratiometric (ExRai) C Kinase Activity Reporter (ExRai-CKAR2) that enables the detection of minute changes in subcellular PKC activity. Using ExRai-CKAR2 in conjunction with an enhanced diacylglycerol (DAG) biosensor capable of detecting intracellular DAG dynamics, we uncover the differential regulation of PKC isoforms at distinct subcellular locations. We find that G-protein coupled receptor (GPCR) stimulation triggers sustained PKC activity at the ER and lysosomes, primarily mediated by Ca2+ sensitive conventional PKC (cPKC) and novel PKC (nPKC), respectively, with nPKC showing high basal activity due to elevated basal DAG levels on lysosome membranes. The high sensitivity of ExRai-CKAR2, targeted to either the cytosol or Par-complexes, further enabled us to detect previously inaccessible endogenous atypical PKC (aPKC) activity in 3D organoids. Taken together, ExRai-CKAR2 is a powerful tool for interrogating PKC regulation in response to physiological stimuli.
Collapse
Affiliation(s)
- Qi Su
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Zhang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Wei Lin
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jin-Fan Zhang
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Alexandra C Newton
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sohum Mehta
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Yang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jin Zhang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
3
|
Kulkarni MV, Jadhav CK, Nipate AS, Bhutada SV, Gill CH, Magar BK. An Efficient One-Pot Green Protocol for the Synthesis of Dihydrochromeno[4,3- b]Pyrazolo[4,3- e]Pyridin-6(7 H)-Ones Mediated by Diisopropyl Ethyl Ammonium Acetate at Room Temperature. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2131852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
| | - Chetan K. Jadhav
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad, India
| | - Amol S. Nipate
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad, India
| | | | - Charansingh H. Gill
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad, India
| | - Bhausaheb K. Magar
- Department of Chemistry, Shivaji Art, Commerce and Science College, Aurangabad, India
| |
Collapse
|
4
|
Chen XZ, Huang Q, Yu XY, Dai C, Shen Y, Lin ZH. Insights into the structural requirements of PKCζ inhibitors as potential anti-arthritis agents based on 3D-QSAR, homology modeling and docking approach. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.131148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
5
|
Pavithra T, Devi ES, Maheswari CU. Recent Advances in N‐Heterocyclic Carbene Catalyzed Oxidative Cyclization for the Formation of Heterocycles. ASIAN J ORG CHEM 2021. [DOI: 10.1002/ajoc.202100289] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- T. Pavithra
- Department of Chemistry, School of Chemical and Biotechnology SASTRA Deemed University Thanjavur 613401 India
| | - E. Sankari Devi
- Department of Chemistry, School of Chemical and Biotechnology SASTRA Deemed University Thanjavur 613401 India
| | - C. Uma Maheswari
- Department of Chemistry, School of Chemical and Biotechnology SASTRA Deemed University Thanjavur 613401 India
| |
Collapse
|
6
|
Atobe M, Serizawa T, Yamakawa N, Takaba K, Nagano Y, Yamaura T, Tanaka E, Tazumi A, Bito S, Ishiguro M, Kawanishi M. Discovery of 4,6- and 5,7-Disubstituted Isoquinoline Derivatives as a Novel Class of Protein Kinase C ζ Inhibitors with Fragment-Merging Strategy. J Med Chem 2020; 63:7143-7162. [DOI: 10.1021/acs.jmedchem.0c00449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Masakazu Atobe
- Laboratory for Medicinal Chemistry, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Takayuki Serizawa
- Laboratory for Medicinal Chemistry, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Natsumi Yamakawa
- Laboratory for Medicinal Chemistry, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Kenichiro Takaba
- Laboratory for Medicinal Chemistry, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Yukiko Nagano
- Research Coordination, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Toshiaki Yamaura
- Laboratory for Drug Discovery, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Eiichi Tanaka
- Laboratory for Drug Discovery, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Atsutoshi Tazumi
- Laboratory for Pharmacology, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Shino Bito
- Laboratory for Safety Assessment & ADME, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni Shizuoka 410-2321, Japan
| | - Masashi Ishiguro
- Laboratory for Safety Assessment & ADME, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni Shizuoka 410-2321, Japan
| | - Masashi Kawanishi
- Laboratory for Medicinal Chemistry, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| |
Collapse
|
7
|
Liu X, Wilson MW, Liu K, Lee P, Yeomans L, Hagen SE, Lin CM, Wen B, Sun D, White AD, Showalter HD, Antonetti DA. Synthesis and structure-activity relationships of thieno[2,3-d]pyrimidines as atypical protein kinase C inhibitors to control retinal vascular permeability and cytokine-induced edema. Bioorg Med Chem 2020; 28:115480. [PMID: 32327351 DOI: 10.1016/j.bmc.2020.115480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 11/28/2022]
Abstract
Studies demonstrate that small molecule targeting of atypical protein kinase C (aPKC) may provide an effective means to control vascular permeability, prevent edema, and reduce inflammation providing novel and important alternatives to anti-VEGF therapies for certain blinding eye diseases. Based on a literature tricyclic thieno[2,3-d]pyrimidine lead (1), an ATP-competitive inhibitor of the aPKC iota (ι) and aPKC zeta (ζ) isoforms, we have synthesized a small series of compounds in 1-2 steps from a readily available chloro intermediate. A single pyridine congener was also made using 2D NMR to assign regiochemistry. Within the parent pyrimidine series, a range of potencies was observed against aPKCζ whereas the pyridine congener was inactive. Selected compounds were also tested for their effect toward VEGF-induced permeability in BREC cells. The most potent of these (7l) was further assayed against the aPKCι isoform and showed a favorable selectivity profile against a panel of 31 kinases, including kinases from the AGC superfamily, with a focus on PKC isoforms and kinases previously shown to affect permeability. Further testing of 7l in a luciferase assay in HEK293 cells showed an ability to prevent TNF-α induced NFκB activation while not having any effect on cell survival. Intravitreal administration of 7l to the eye yielded a complete reduction in permeability in a test to determine whether the compound could block VEGF- and TNFα-induced permeability across the retinal vasculature in a rat model. The compound in mice displayed good microsomal stability and in plasma moderate exposure (AUC and Cmax), low clearance, a long half-life and high oral bioavailability. With IV dosing, higher levels were observed in the brain and eye relative to plasma, with highest levels in the eye by either IV or PO dosing. With a slow oral absorption profile, 7l accumulates in the eye to maintain a high concentration after dosing with higher levels than in plasma. Compound 7l may represent a class of aPKC inhibitors for further investigation.
Collapse
Affiliation(s)
- Xuwen Liu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michael W Wilson
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kun Liu
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pil Lee
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Larisa Yeomans
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Susan E Hagen
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew D White
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hollis D Showalter
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
8
|
aPKC in neuronal differentiation, maturation and function. Neuronal Signal 2019; 3:NS20190019. [PMID: 32269838 PMCID: PMC7104321 DOI: 10.1042/ns20190019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
The atypical Protein Kinase Cs (aPKCs)—PRKCI, PRKCZ and PKMζ—form a subfamily within the Protein Kinase C (PKC) family. These kinases are expressed in the nervous system, including during its development and in adulthood. One of the aPKCs, PKMζ, appears to be restricted to the nervous system. aPKCs are known to play a role in a variety of cellular responses such as proliferation, differentiation, polarity, migration, survival and key metabolic functions such as glucose uptake, that are critical for nervous system development and function. Therefore, these kinases have garnered a lot of interest in terms of their functional role in the nervous system. Here we review the expression and function of aPKCs in neural development and in neuronal maturation and function. Despite seemingly paradoxical findings with genetic deletion versus gene silencing approaches, we posit that aPKCs are likely candidates for regulating many important neurodevelopmental and neuronal functions, and may be associated with a number of human neuropsychiatric diseases.
Collapse
|
9
|
Reina-Campos M, Diaz-Meco MT, Moscat J. The Dual Roles of the Atypical Protein Kinase Cs in Cancer. Cancer Cell 2019; 36:218-235. [PMID: 31474570 PMCID: PMC6751000 DOI: 10.1016/j.ccell.2019.07.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/24/2019] [Accepted: 07/30/2019] [Indexed: 02/08/2023]
Abstract
Atypical protein kinase C (aPKC) isozymes, PKCλ/ι and PKCζ, are now considered fundamental regulators of tumorigenesis. However, the specific separation of functions that determine their different roles in cancer is still being unraveled. Both aPKCs have pleiotropic context-dependent functions that can translate into tumor-promoter or -suppressive functions. Here, we review early and more recent literature to discuss how the different tumor types, and their microenvironments, might account for the selective signaling of each aPKC isotype. This is of clinical relevance because a better understanding of the roles of these kinases is essential for the design of new anti-cancer treatments.
Collapse
Affiliation(s)
- Miguel Reina-Campos
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maria T Diaz-Meco
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jorge Moscat
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
10
|
Construction of substituted imidazoles from aryl methyl ketones and benzylamines via N-heterocyclic carbene-catalysis. CATAL COMMUN 2019. [DOI: 10.1016/j.catcom.2019.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
11
|
Abstract
Protein kinase C (PKC) isozymes belong to a family of Ser/Thr kinases whose activity is governed by reversible release of an autoinhibitory pseudosubstrate. For conventional and novel isozymes, this is effected by binding the lipid second messenger, diacylglycerol, but for atypical PKC isozymes, this is effected by binding protein scaffolds. PKC shot into the limelight following the discovery in the 1980s that the diacylglycerol-sensitive isozymes are "receptors" for the potent tumor-promoting phorbol esters. This set in place a concept that PKC isozymes are oncoproteins. Yet three decades of cancer clinical trials targeting PKC with inhibitors failed and, in some cases, worsened patient outcome. Emerging evidence from cancer-associated mutations and protein expression levels provide a reason: PKC isozymes generally function as tumor suppressors and their activity should be restored, not inhibited, in cancer therapies. And whereas not enough activity is associated with cancer, variants with enhanced activity are associated with degenerative diseases such as Alzheimer's disease. This review describes the tightly controlled mechanisms that ensure PKC activity is perfectly balanced and what happens when these controls are deregulated. PKC isozymes serve as a paradigm for the wisdom of Confucius: "to go beyond is as wrong as to fall short."
Collapse
Affiliation(s)
- Alexandra C Newton
- a Department of Pharmacology , University of California at San Diego , La Jolla , CA , USA
| |
Collapse
|
12
|
Kajimoto T, Caliman AD, Tobias IS, Okada T, Pilo CA, Van AAN, Andrew McCammon J, Nakamura SI, Newton AC. Activation of atypical protein kinase C by sphingosine 1-phosphate revealed by an aPKC-specific activity reporter. Sci Signal 2019; 12:eaat6662. [PMID: 30600259 PMCID: PMC6657501 DOI: 10.1126/scisignal.aat6662] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Atypical protein kinase C (aPKC) isozymes are unique in the PKC superfamily in that they are not regulated by the lipid second messenger diacylglycerol, which has led to speculation about whether a different second messenger acutely controls their function. Here, using a genetically encoded reporter that we designed, aPKC-specific C kinase activity reporter (aCKAR), we found that the lipid mediator sphingosine 1-phosphate (S1P) promoted the cellular activity of aPKC. Intracellular S1P directly bound to the purified kinase domain of aPKC and relieved autoinhibitory constraints, thereby activating the kinase. In silico studies identified potential binding sites on the kinase domain, one of which was validated biochemically. In HeLa cells, S1P-dependent activation of aPKC suppressed apoptosis. Together, our findings identify a previously undescribed molecular mechanism of aPKC regulation, a molecular target for S1P in cell survival regulation, and a tool to further explore the biochemical and biological functions of aPKC.
Collapse
Affiliation(s)
- Taketoshi Kajimoto
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA.
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Alisha D Caliman
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA
| | - Irene S Tobias
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA
| | - Taro Okada
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Caila A Pilo
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA
| | - An-Angela N Van
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA
| | - J Andrew McCammon
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA
| | - Shun-Ichi Nakamura
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Alexandra C Newton
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Li G, Hao WJ, Hu JY, Gao YY, Zhang WW, Zhang KY, Li WL, Jiang B. Diastereoselective Synthesis of Poly-Substituted syn-Imidazolidine-2-thiones via Microwave-Assisted Three-Component [2+2+1] Heterocyclizations. HETEROCYCLES 2019. [DOI: 10.3987/com-18-s(f)20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
14
|
Abdel-Halim M, Abadi AH, Engel M. Design and synthesis of novel 1,3,5-triphenyl pyrazolines as potential anti-inflammatory agents through allosteric inhibition of protein kinase Czeta (PKCζ). MEDCHEMCOMM 2018; 9:1076-1082. [PMID: 30108997 PMCID: PMC6072096 DOI: 10.1039/c8md00100f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/25/2018] [Indexed: 01/01/2023]
Abstract
Much light has been shed on the vital role of protein kinase Czeta (PKCζ) in NF-κB activation and the potential use of PKCζ inhibitors as anti-inflammatory agents. We previously reported a series of 1,3,5-trisubstituted pyrazolines as potent and selective allosteric inhibitors of PKCζ; in that series of compounds, the phenolic OH at the 5-phenyl was essential for binding to the PKCζ PIF pocket. In the present study, we surprisingly found that replacing it by a halogen and at the same time moving the OH to the 3-phenyl still resulted in active compounds. An extension of this class of compounds with a new focused library is presented herein, where the phenolic OH at the 5-phenyl, which was reported to be an irreplaceable feature for activity, was moved to the 3-phenyl and replaced by halogen. The new set of compounds maintained the same level of potency against PKCζ and selectivity against PKC isoforms, and showed reduced potency against the PIF pocket mutant PKCζ[Val297Leu]. Of note, the repositioning of the key functional groups resulted in a marked enhancement of cellular potency. One of the most potent new PKCζ inhibitors, 2h, was able to suppress NO production in RAW 264.7 macrophage cells with 8 times higher efficacy than the previous series, and inhibited the NF-κB transcriptional activity in U937 cells with a sub-micromolar IC50.
Collapse
Affiliation(s)
- Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry , Faculty of Pharmacy and Biotechnology , German University in Cairo , Cairo 11835 , Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry , Faculty of Pharmacy and Biotechnology , German University in Cairo , Cairo 11835 , Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry , Saarland University , Campus C2.3 , D-66123 Saarbrücken , Germany . ; http://www.pharmmedchem.de ; ; Tel: +49 681 302 70312
| |
Collapse
|
15
|
Catalyst-free synthesis of 1,2,4,5-tetrasubstituted imidazoles from arylamins, benzonitriles, arylglyoxals, and Meldrum’s acid. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.03.093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Chevalier A, Ouahrouch A, Arnaud A, Gallavardin T, Franck X. An optimized procedure for direct access to 1H-indazole-3-carboxaldehyde derivatives by nitrosation of indoles. RSC Adv 2018; 8:13121-13128. [PMID: 35542511 PMCID: PMC9079728 DOI: 10.1039/c8ra01546e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/31/2018] [Indexed: 12/24/2022] Open
Abstract
The nitrosation of indoles under slightly acidic conditions and reverse addition conditions leads to the preparation of the corresponding indazole-3-carboxaldehydes in high yields and greatly minimizes side reactions.
Collapse
Affiliation(s)
- Arnaud Chevalier
- Normandie Univ
- CNRS
- UNIROUEN
- INSA Rouen
- COBRA (UMR 6014 and FR 3038)
| | | | - Alexandre Arnaud
- Normandie Univ
- CNRS
- UNIROUEN
- INSA Rouen
- COBRA (UMR 6014 and FR 3038)
| | | | - Xavier Franck
- Normandie Univ
- CNRS
- UNIROUEN
- INSA Rouen
- COBRA (UMR 6014 and FR 3038)
| |
Collapse
|
17
|
Harashima H, Dissmeyer N, Hammann P, Nomura Y, Kramer K, Nakagami H, Schnittger A. Modulation of plant growth in vivo and identification of kinase substrates using an analog-sensitive variant of CYCLIN-DEPENDENT KINASE A;1. BMC PLANT BIOLOGY 2016; 16:209. [PMID: 27669979 PMCID: PMC5037886 DOI: 10.1186/s12870-016-0900-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/16/2016] [Indexed: 05/12/2023]
Abstract
BACKGROUND Modulation of protein activity by phosphorylation through kinases and subsequent de-phosphorylation by phosphatases is one of the most prominent cellular control mechanisms. Thus, identification of kinase substrates is pivotal for the understanding of many - if not all - molecular biological processes. Equally, the possibility to deliberately tune kinase activity is of great value to analyze the biological process controlled by a particular kinase. RESULTS Here we have applied a chemical genetic approach and generated an analog-sensitive version of CDKA;1, the central cell-cycle regulator in Arabidopsis and homolog of the yeast Cdc2/CDC28 kinases. This variant could largely rescue a cdka;1 mutant and is biochemically active, albeit less than the wild type. Applying bulky kinase inhibitors allowed the reduction of kinase activity in an organismic context in vivo and the modulation of plant growth. To isolate CDK substrates, we have adopted a two-dimensional differential gel electrophoresis strategy, and searched for proteins that showed mobility changes in fluorescently labeled extracts from plants expressing the analog-sensitive version of CDKA;1 with and without adding a bulky ATP variant. A pilot set of five proteins involved in a range of different processes could be confirmed in independent kinase assays to be phosphorylated by CDKA;1 approving the applicability of the here-developed method to identify substrates. CONCLUSION The here presented generation of an analog-sensitive CDKA;1 version is functional and represent a novel tool to modulate kinase activity in vivo and identify kinase substrates. Our here performed pilot screen led to the identification of CDK targets that link cell proliferation control to sugar metabolism, proline proteolysis, and glucosinolate production providing a hint how cell proliferation and growth are integrated with plant development and physiology.
Collapse
Affiliation(s)
- Hirofumi Harashima
- Department of Molecular Mechanisms of Phenotypic Plasticity, Institut de Biologie Moléculaire des Plantes du CNRS, IBMP-CNRS - UPR2357, Université de Strasbourg, F-67084 Strasbourg, France
- Trinationales Institut für Pflanzenforschung, F-67084 Strasbourg Cedex, France
- Present address: RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045 Japan
| | - Nico Dissmeyer
- Department of Molecular Mechanisms of Phenotypic Plasticity, Institut de Biologie Moléculaire des Plantes du CNRS, IBMP-CNRS - UPR2357, Université de Strasbourg, F-67084 Strasbourg, France
- Trinationales Institut für Pflanzenforschung, F-67084 Strasbourg Cedex, France
- Present address: Leibniz Institute of Plant Biochemistry (IPB), Independent Junior Research Group on Protein Recognition and Degradation, Weinberg 3, D-06120 Halle, (Saale) Germany
| | - Philippe Hammann
- Plateforme protéomique Strasbourg Esplanade, Institut de Biologie Moléculaire et Cellulaire FRC1589-CNRS, F-67084 Strasbourg, France
| | - Yuko Nomura
- Plant Proteomics Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi Yokohama, 230-0045 Japan
| | - Katharina Kramer
- Max Planck Institute for Plant Breeding Research, Basic Immune System of Plants / Protein Mass Spectrometry, Carl-von-Linne-Weg 10, 50829 Cologne, Germany
| | - Hirofumi Nakagami
- Plant Proteomics Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi Yokohama, 230-0045 Japan
- Max Planck Institute for Plant Breeding Research, Basic Immune System of Plants / Protein Mass Spectrometry, Carl-von-Linne-Weg 10, 50829 Cologne, Germany
| | - Arp Schnittger
- Department of Molecular Mechanisms of Phenotypic Plasticity, Institut de Biologie Moléculaire des Plantes du CNRS, IBMP-CNRS - UPR2357, Université de Strasbourg, F-67084 Strasbourg, France
- Trinationales Institut für Pflanzenforschung, F-67084 Strasbourg Cedex, France
- Department of Developmental Biology, University of Hamburg, Biozentrum Klein Flottbek, Ohnhorststr. 18, D-22609 Hamburg, Germany
| |
Collapse
|
18
|
Tobias IS, Newton AC. Protein Scaffolds Control Localized Protein Kinase Cζ Activity. J Biol Chem 2016; 291:13809-22. [PMID: 27143478 DOI: 10.1074/jbc.m116.729483] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Indexed: 11/06/2022] Open
Abstract
Atypical protein kinase C (aPKC) isozymes modulate insulin signaling and cell polarity, but how their activity is controlled in cells is not well understood. These enzymes are constitutively phosphorylated, insensitive to second messengers, and have relatively low activity. Here we show that protein scaffolds not only localize but also differentially control the catalytic activity of the aPKC PKCζ, thus promoting activity toward localized substrates and restricting activity toward global substrates. Using cellular substrate readouts and scaffolded activity reporters in live cell imaging, we show that PKCζ has highly localized and differentially controlled activity on the scaffolds p62 and Par6. Both scaffolds tether aPKC in an active conformation as assessed through pharmacological inhibition of basal activity, monitored using a genetically encoded reporter for PKC activity. However, binding to Par6 is of higher affinity and is more effective in locking PKCζ in an active conformation. FRET-based translocation assays reveal that insulin promotes the association of both p62 and aPKC with the insulin-regulated scaffold IRS-1. Using the aPKC substrate MARK2 as another readout for activity, we show that overexpression of IRS-1 reduces the phosphorylation of MARK2 and enhances its plasma membrane localization, indicating sequestration of aPKC by IRS-1 away from MARK2. These results are consistent with scaffolds serving as allosteric activators of aPKCs, tethering them in an active conformation near specific substrates. Thus, signaling of these intrinsically low activity kinases is kept at a minimum in the absence of scaffolding interactions, which position the enzymes for stoichiometric phosphorylation of substrates co-localized on the same protein scaffold.
Collapse
Affiliation(s)
- Irene S Tobias
- From the Department of Pharmacology and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California 92093
| | | |
Collapse
|
19
|
Grigoroudis AI, Kontopidis G. Preparation of CDK/Cyclin Inhibitor Complexes for Structural Determination. Methods Mol Biol 2016; 1336:29-45. [PMID: 26231706 DOI: 10.1007/978-1-4939-2926-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The abundance of biochemical and structural knowledge on the Cyclin-Dependent Kinases (CDKs) has provided a comprehensive but not exhaustive insight into the molecular determinants that govern their function mechanisms. The implementation of structural and functional CDK models towards developing novel anticancer strategies that will specifically target individual or multiple CDKs remains a critical need.More than 250 CDKs crystal structures are available to-date, including truncated or whole, modified or not, active or inactive forms, co-crystallized with the cyclins and/or their respective putative inhibitors, though, to our knowledge, there is no NMR solved structure available to date. We hitherto attempt to provide a useful guide from protein production to crystallization for CDK/Inhibitors complexes based on an overview of the already elucidated CDK structures, constructs and the preferable expression vectors in each case, in order to yield the respective crystals.
Collapse
Affiliation(s)
- Asterios I Grigoroudis
- Department of Biochemistry, Veterinary School, University of Thessaly, Trikalon 224 Str., Karditsa, 43100, Greece
| | | |
Collapse
|
20
|
Protein kinase Cζ exhibits constitutive phosphorylation and phosphatidylinositol-3,4,5-triphosphate-independent regulation. Biochem J 2015; 473:509-23. [PMID: 26635352 DOI: 10.1042/bj20151013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/03/2015] [Indexed: 12/21/2022]
Abstract
Atypical protein kinase C (aPKC) isoenzymes are key modulators of insulin signalling, and their dysfunction correlates with insulin-resistant states in both mice and humans. Despite the engaged interest in the importance of aPKCs to type 2 diabetes, much less is known about the molecular mechanisms that govern their cellular functions than for the conventional and novel PKC isoenzymes and the functionally-related protein kinase B (Akt) family of kinases. Here we show that aPKC is constitutively phosphorylated and, using a genetically-encoded reporter for PKC activity, basally active in cells. Specifically, we show that phosphorylation at two key regulatory sites, the activation loop and turn motif, of the aPKC PKCζ in multiple cultured cell types is constitutive and independently regulated by separate kinases: ribosome-associated mammalian target of rapamycin complex 2 (mTORC2) mediates co-translational phosphorylation of the turn motif, followed by phosphorylation at the activation loop by phosphoinositide-dependent kinase-1 (PDK1). Live cell imaging reveals that global aPKC activity is constitutive and insulin unresponsive, in marked contrast to the insulin-dependent activation of Akt monitored by an Akt-specific reporter. Nor does forced recruitment to phosphoinositides by fusing the pleckstrin homology (PH) domain of Akt to the kinase domain of PKCζ alter either the phosphorylation or activity of PKCζ. Thus, insulin stimulation does not activate PKCζ through the canonical phosphatidylinositol-3,4,5-triphosphate-mediated pathway that activates Akt, contrasting with previous literature on PKCζ activation. These studies support a model wherein an alternative mechanism regulates PKCζ-mediated insulin signalling that does not utilize conventional activation via agonist-evoked phosphorylation at the activation loop. Rather, we propose that scaffolding near substrates drives the function of PKCζ.
Collapse
|
21
|
Kumar PPS, Suchetan PA, Sreenivasa S, Naveen S, Lokanath NK, Kumar DBA. Molecular and crystal structures of two 1,2,4-benzothiadiazine derivatives. J STRUCT CHEM+ 2015. [DOI: 10.1134/s0022476615070215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Tsai LCL, Xie L, Dore K, Xie L, Del Rio JC, King CC, Martinez-Ariza G, Hulme C, Malinow R, Bourne PE, Newton AC. Zeta Inhibitory Peptide Disrupts Electrostatic Interactions That Maintain Atypical Protein Kinase C in Its Active Conformation on the Scaffold p62. J Biol Chem 2015; 290:21845-56. [PMID: 26187466 PMCID: PMC4571940 DOI: 10.1074/jbc.m115.676221] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 07/17/2015] [Indexed: 11/06/2022] Open
Abstract
Atypical protein kinase C (aPKC) enzymes signal on protein scaffolds, yet how they are maintained in an active conformation on scaffolds is unclear. A myristoylated peptide based on the autoinhibitory pseudosubstrate fragment of the atypical PKCζ, zeta inhibitory peptide (ZIP), has been extensively used to inhibit aPKC activity; however, we have previously shown that ZIP does not inhibit the catalytic activity of aPKC isozymes in cells (Wu-Zhang, A. X., Schramm, C. L., Nabavi, S., Malinow, R., and Newton, A. C. (2012) J. Biol. Chem. 287, 12879-12885). Here we sought to identify a bona fide target of ZIP and, in so doing, unveiled a novel mechanism by which aPKCs are maintained in an active conformation on a protein scaffold. Specifically, we used protein-protein interaction network analysis, structural modeling, and protein-protein docking to predict that ZIP binds an acidic surface on the Phox and Bem1 (PB1) domain of p62, an interaction validated by peptide array analysis. Using a genetically encoded reporter for PKC activity fused to the p62 scaffold, we show that ZIP inhibits the activity of wild-type aPKC, but not a construct lacking the pseudosubstrate. These data support a model in which the pseudosubstrate of aPKCs is tethered to the acidic surface on p62, locking aPKC in an open, signaling-competent conformation. ZIP competes for binding to the acidic surface, resulting in displacement of the pseudosubstrate of aPKC and re-engagement in the substrate-binding cavity. This study not only identifies a cellular target for ZIP, but also unveils a novel mechanism by which scaffolded aPKC is maintained in an active conformation.
Collapse
Affiliation(s)
| | - Lei Xie
- the Department of Computer Science, Hunter College, the City University of New York, New York, New York 10065
| | | | - Li Xie
- Skaggs School of Pharmacy, and
| | | | - Charles C King
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California 92093
| | - Guillermo Martinez-Ariza
- the Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, and
| | - Christopher Hulme
- the Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, and
| | | | - Philip E Bourne
- the Office of the Director, the National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|
23
|
Maleki A, Alrezvani Z, Maleki S. Design, preparation and characterization of urea-functionalized Fe3O4/SiO2 magnetic nanocatalyst and application for the one-pot multicomponent synthesis of substituted imidazole derivatives. CATAL COMMUN 2015. [DOI: 10.1016/j.catcom.2015.05.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
24
|
Abstract
In preclinical studies, protein kinase C (PKC) enzymes have been implicated in regulating many aspects of pancreatic cancer development and progression. However, clinical Phase I or Phase II trials with compounds targeting classical PKC isoforms were not successful. Recent studies implicate that mainly atypical and novel PKC enzymes regulate oncogenic signaling pathways in pancreatic cancer. Members of these two subgroups converge signaling induced by mutant Kras, growth factors and inflammatory cytokines. Different approaches for the development of inhibitors for atypical PKC and novel PKC have been described; and new compounds include allosteric inhibitors and inhibitors that block ATP binding.
Collapse
Affiliation(s)
- Peter Storz
- Department of Cancer Biology, Mayo Clinic, Griffin Building, Room 306, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
25
|
Rescifina A, Scala A, Sciortino MT, Colao I, Siracusano G, Mazzaglia A, Chiacchio U, Grassi G. Decorated 6,6′,7,7′-tetrahydro-1H,1′H-2,3′-biindole scaffold as promising candidate for recognition of the CDK2 allosteric site. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00364k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Decorated 6,6′,7,7′-tetrahydro-1H,1′H-2,3′-biindoles, such as DPIT, targeting CDK2 seem to be an attractive scaffold for development of useful anticancer drugs.
Collapse
Affiliation(s)
- Antonio Rescifina
- Dipartimento di Scienze del Farmaco
- Università di Catania
- 95125 Catania
- Italy
| | - Angela Scala
- Dipartimento di Scienze Chimiche
- Università di Messina
- 98166 Messina
- Italy
| | | | - Ivana Colao
- Dipartimento di Scienze Biologiche ed Ambientali
- Università di Messina
- 98166 Messina
- Italy
| | - Gabriel Siracusano
- Dipartimento di Scienze Biologiche ed Ambientali
- Università di Messina
- 98166 Messina
- Italy
| | - Antonino Mazzaglia
- CNR-ISMN Istituto per lo Studio dei Materiali Nanostrutturati c/o Dipartimento di Scienze Chimiche dell'Università di Messina
- 98166 Messina
- Italy
| | - Ugo Chiacchio
- Dipartimento di Scienze del Farmaco
- Università di Catania
- 95125 Catania
- Italy
| | - Giovanni Grassi
- Dipartimento di Scienze Chimiche
- Università di Messina
- 98166 Messina
- Italy
| |
Collapse
|
26
|
Shinoj Kumar PP, Suchetan PA, Sreenivasa S, Naveen S, Lokanath NK, Aruna Kumar DB. Crystal structure of (R)-6'-bromo-3,3-dimethyl-3',4'-di-hydro-2'H-spiro-[cyclo-hexane-1,3'-1,2,4-benzo-thia-diazine] 1',1'-dioxide. Acta Crystallogr Sect E Struct Rep Online 2014; 70:359-61. [PMID: 25484745 PMCID: PMC4257282 DOI: 10.1107/s1600536814022417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 10/12/2014] [Indexed: 11/10/2022]
Abstract
In the title compound, C14H19BrN2O2S, the 1,2,4-thia-diazinane ring adopts an envelope conformation with the N atom (attached to the sulfonyl group) as the flap, while the cyclo-hexane ring adopts a chair conformation. The mean plane of the cyclo-hexane ring is almost normal to the benzene ring and the mean plane of the 1,2,4-thia-diazinane ring, making dihedral angles of 70.4 (2) and 71.43 (19)°, respectively. Furthermore, the dihedral angle between the benzene ring and the mean plane of the 1,2,4-thia-diazinane ring is 4.91 (18)°. The mol-ecular structure is stabilized by an intra-molecular C-H⋯O hydrogen bond, which encloses an S(6) ring motif. In the crystal, mol-ecules are linked by N-H⋯O hydrogen bonds into chains along [10-1], forming a C(6) graph-set motif. These chains are inter-connected via C-H⋯π inter-actions, leading to chains along [-101], so finally forming sheets parallel to (010).
Collapse
Affiliation(s)
- P P Shinoj Kumar
- Department of Studies and Research in Chemistry, Tumkur University, Tumkur 572 103, India
| | - P A Suchetan
- Department of Studies and Research in Chemistry, U.C.S., Tumkur University, Tumkur 572 013, India
| | - S Sreenivasa
- Department of Studies and Research in Chemistry, Tumkur University, Tumkur 572 103, India
| | - S Naveen
- Institution of Excellence, Vijnana Bhavan, University of Mysore, Manasagangotri, India
| | - N K Lokanath
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore, India
| | - D B Aruna Kumar
- Department of Studies and Research in Chemistry, Tumkur University, Tumkur 572 103, India
| |
Collapse
|
27
|
Kusne Y, Carrera-Silva EA, Perry AS, Rushing EJ, Mandell EK, Dietrich JD, Errasti AE, Gibbs D, Berens ME, Loftus JC, Hulme C, Yang W, Lu Z, Aldape K, Sanai N, Rothlin CV, Ghosh S. Targeting aPKC disables oncogenic signaling by both the EGFR and the proinflammatory cytokine TNFα in glioblastoma. Sci Signal 2014; 7:ra75. [PMID: 25118327 PMCID: PMC4486020 DOI: 10.1126/scisignal.2005196] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Grade IV glioblastoma is characterized by increased kinase activity of epidermal growth factor receptor (EGFR); however, EGFR kinase inhibitors have failed to improve survival in individuals with this cancer because resistance to these drugs often develops. We showed that tumor necrosis factor-α (TNFα) produced in the glioblastoma microenvironment activated atypical protein kinase C (aPKC), thereby producing resistance to EGFR kinase inhibitors. Additionally, we identified that aPKC was required both for paracrine TNFα-dependent activation of the transcription factor nuclear factor κB (NF-κB) and for tumor cell-intrinsic receptor tyrosine kinase signaling. Targeting aPKC decreased tumor growth in mouse models of glioblastoma, including models of EGFR kinase inhibitor-resistant glioblastoma. Furthermore, aPKC abundance and activity were increased in human glioblastoma tumor cells, and high aPKC abundance correlated with poor prognosis. Thus, targeting aPKC might provide an improved molecular approach for glioblastoma therapy.
Collapse
Affiliation(s)
- Yael Kusne
- Neuroscience Graduate Program, Arizona State University, Phoenix, AZ 85287, USA
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | | | - Anthony S. Perry
- Department of Pathology, Banner MD Anderson Cancer Center, Gilbert, AZ 85234, USA
| | | | - Edward K. Mandell
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Andrea E. Errasti
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daniel Gibbs
- Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Michael E. Berens
- Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA
| | | | | | - Weiwei Yang
- MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhimin Lu
- MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Nader Sanai
- Neuroscience Graduate Program, Arizona State University, Phoenix, AZ 85287, USA
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Carla V. Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sourav Ghosh
- Neuroscience Graduate Program, Arizona State University, Phoenix, AZ 85287, USA
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06511, USA
- Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA
| |
Collapse
|
28
|
Abdel-Halim M, Diesel B, Kiemer AK, Abadi AH, Hartmann RW, Engel M. Discovery and optimization of 1,3,5-trisubstituted pyrazolines as potent and highly selective allosteric inhibitors of protein kinase C-ζ. J Med Chem 2014; 57:6513-30. [PMID: 25058929 DOI: 10.1021/jm500521n] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is increasing evidence that the atypical protein kinase C, PKCζ, might be a therapeutic target in pulmonary and hepatic inflammatory diseases. However, targeting the highly conserved ATP-binding pocket in the catalytic domain held little promise to achieve selective inhibition. In the present study, we introduce 1,3,5-trisubstituted pyrazolines as potent and selective allosteric PKCζ inhibitors. The rigid scaffold offered many sites for modification, all acting as hot spots for improving activity, and gave rise to sharp structure-activity relationships. Targeting of PKCζ in cells was confirmed by reporter gene assay, transfection assays, and Western blotting. The strongly reduced cell-free and cellular activities toward a PIF-pocket mutant of PKCζ suggested that the inhibitors most likely bound to the PIF-pocket on the kinase catalytic domain. Thus, using a rigidification strategy and by establishing and optimizing multiple molecular interactions with the binding site, we were able to significantly improve the potency of the previously reported PKCζ inhibitors.
Collapse
Affiliation(s)
- Mohammad Abdel-Halim
- Pharmaceutical and Medicinal Chemistry, Saarland University , Campus C2.3, D-66123 Saarbrücken, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Synthesis of dihydrochromeno[4,3-b]pyrazolo[4,3-e]pyridin-6(7H)-ones involving one-pot three-component tandem Knoevenagel–Michael reaction catalyzed by n-tetrabutylammonium tribromide (TBATB). Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
30
|
Singla P, Luxami V, Paul K. Benzimidazole-biologically attractive scaffold for protein kinase inhibitors. RSC Adv 2014. [DOI: 10.1039/c3ra46304d] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
31
|
Selective kinase inhibitors as tools for neuroscience research. Neuropharmacology 2012; 63:1227-37. [DOI: 10.1016/j.neuropharm.2012.07.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 07/06/2012] [Accepted: 07/11/2012] [Indexed: 01/02/2023]
|
32
|
Novel atypical PKC inhibitors prevent vascular endothelial growth factor-induced blood-retinal barrier dysfunction. Biochem J 2012; 446:455-67. [PMID: 22721706 DOI: 10.1042/bj20111961] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pro-inflammatory cytokines and growth factors such as VEGF (vascular endothelial growth factor) contribute to the loss of the BRB (blood-retinal barrier) and subsequent macular oedema in various retinal pathologies. VEGF signalling requires PKCβ [conventional PKC (protein kinase C)] activity; however, PKCβ inhibition only partially prevents VEGF-induced endothelial permeability and does not affect pro-inflammatory cytokine-induced permeability, suggesting the involvement of alternative signalling pathways. In the present study, we provide evidence for the involvement of aPKC (atypical PKC) signalling in VEGF-induced endothelial permeability and identify a novel class of inhibitors of aPKC that prevent BRB breakdown in vivo. Genetic and pharmacological manipulations of aPKC isoforms were used to assess their contribution to endothelial permeability in culture. A chemical library was screened using an in vitro kinase assay to identify novel small-molecule inhibitors, and further medicinal chemistry was performed to delineate a novel pharmacophore. We demonstrate that aPKC isoforms are both sufficient and required for VEGF-induced endothelial permeability. Furthermore, these specific, potent, non-competitive, small-molecule inhibitors prevented VEGF-induced tight junction internalization and retinal endothelial permeability in response to VEGF in both primary culture and in rodent retina. The results of the present study suggest that aPKC inhibition with 2-amino-4-phenyl-thiophene derivatives may be developed to preserve the BRB in retinal diseases such as diabetic retinopathy or uveitis, and the BBB (blood-brain barrier) in the presence of brain tumours.
Collapse
|
33
|
Migliorini A, Oliviero C, Gasperi T, Loreto MA. The Suzuki reaction applied to the synthesis of novel pyrrolyl and thiophenyl indazoles. Molecules 2012; 17:4508-21. [PMID: 22508331 PMCID: PMC6268423 DOI: 10.3390/molecules17044508] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/05/2012] [Accepted: 04/09/2012] [Indexed: 11/30/2022] Open
Abstract
The paper describes the Suzuki cross-coupling of a variety of N and C-3 substituted 5-bromoindazoles with N-Boc-2-pyrrole and 2-thiopheneboronic acids. The reactions, performed in the presence of K(2)CO(3), dimethoxyethane and Pd(dppf)Cl(2) as catalyst, gave the corresponding adducts in good yields. The methodology allows the facile production of indazole-based heteroaryl compounds, a unique architectural motif that is ubiquitous in biologically active molecules.
Collapse
Affiliation(s)
- Antonella Migliorini
- Department of Chemistry, “Sapienza” University of Rome, P.le A. Moro 5, I-00185 Roma, Italy; (A.M.); (C.O.)
| | - Chiara Oliviero
- Department of Chemistry, “Sapienza” University of Rome, P.le A. Moro 5, I-00185 Roma, Italy; (A.M.); (C.O.)
| | - Tecla Gasperi
- Department of Mechanical and Industrial Engineering and CISDiC, University of Studies “Roma Tre”, via della Vasca Navale 79, I-00146 Roma, Italy;
| | - Maria Antonietta Loreto
- Department of Chemistry, “Sapienza” University of Rome, P.le A. Moro 5, I-00185 Roma, Italy; (A.M.); (C.O.)
| |
Collapse
|
34
|
Lopez-Garcia LA, Schulze JO, Fröhner W, Zhang H, Süss E, Weber N, Navratil J, Amon S, Hindie V, Zeuzem S, Jørgensen TJD, Alzari PM, Neimanis S, Engel M, Biondi RM. Allosteric regulation of protein kinase PKCζ by the N-terminal C1 domain and small compounds to the PIF-pocket. ACTA ACUST UNITED AC 2012; 18:1463-73. [PMID: 22118680 DOI: 10.1016/j.chembiol.2011.08.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 07/28/2011] [Accepted: 08/05/2011] [Indexed: 02/08/2023]
Abstract
Protein kinases are key mediators of cellular signaling, and therefore, their activities are tightly controlled. AGC kinases are regulated by phosphorylation and by N- and C-terminal regions. Here, we studied the molecular mechanism of inhibition of atypical PKCζ and found that the inhibition by the N-terminal region cannot be explained by a simple pseudosubstrate inhibitory mechanism. Notably, we found that the C1 domain allosterically inhibits PKCζ activity and verified an allosteric communication between the PIF-pocket of atypical PKCs and the binding site of the C1 domain. Finally, we developed low-molecular-weight compounds that bind to the PIF-pocket and allosterically inhibit PKCζ activity. This work establishes a central role for the PIF-pocket on the regulation of PKCζ and allows us to envisage development of drugs targeting the PIF-pocket that can either activate or inhibit AGC kinases.
Collapse
Affiliation(s)
- Laura A Lopez-Garcia
- Research Group PhosphoSites, Department of Internal Medicine I, Universitätsklinikum Frankfurt, 60590 Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Fröhner W, Lopez-Garcia LA, Neimanis S, Weber N, Navratil J, Maurer F, Stroba A, Zhang H, Biondi RM, Engel M. 4-benzimidazolyl-3-phenylbutanoic acids as novel PIF-pocket-targeting allosteric inhibitors of protein kinase PKCζ. J Med Chem 2011; 54:6714-23. [PMID: 21863889 DOI: 10.1021/jm2005892] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Protein kinase inhibitors with an allosteric mode of action are expected to reach, in many cases, higher selectivity for the target enzyme than ATP-competitive compounds. Therefore, basic research is aiming at identifying and establishing novel sites on the catalytic domain of protein kinases which might be targeted by allosteric inhibitors. We previously published the first structure-activity relationships (SARs) for allosteric activators of protein kinase PDK1. Here, we present the design, synthesis, and SAR data on a series of novel compounds, 4-benzimidazolyl-3-phenylbutanoic acids, that inhibit the atypical protein kinace C (PKC) ζ via binding to the PIF-pocket. Key positions were identified in the compounds that can be modified to increase potency and selectivity. Some congeners showed a high selectivity toward PKCζ, lacking inhibition of the most closely related isoform, PKCι, and of further AGC kinases. Furthermore, evidence is provided that these compounds are also active toward cellular PKCζ without loss of potency compared to the cell-free assay.
Collapse
Affiliation(s)
- Wolfgang Fröhner
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 151150, D-66041 Saarbrücken, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Djung JF, Mears RJ, Montalbetti CA, Coulter TS, Golebiowski A, Carr AN, Barker O, Greis KD, Zhou S, Dolan E, Davis GF. The synthesis and evaluation of indolylureas as PKCα inhibitors. Bioorg Med Chem 2011; 19:2742-50. [DOI: 10.1016/j.bmc.2011.02.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 02/17/2011] [Accepted: 02/21/2011] [Indexed: 10/18/2022]
|
38
|
Design, synthesis, and testing of an 6-O-linked series of benzimidazole based inhibitors of CDK5/p25. Bioorg Med Chem 2010; 19:359-73. [PMID: 21144757 DOI: 10.1016/j.bmc.2010.11.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/01/2010] [Accepted: 11/06/2010] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease resulting in cognitive and behavioral impairment. The two classic pathological hallmarks of AD include extraneuronal deposition of amyloid β (Aβ) and intraneuronal formation of neurofibrillary tangles (NFTs). NFTs contain hyperphosphorylated tau. Tau is the major microtubule-associated protein in neurons and stabilizes microtubules (MTs). Cyclin dependent kinase 5 (CDK5), when activated by the regulatory binding protein p25, phosphorylates tau at a number of proline-directed serine/threonine residues, resulting in formation of phosphorylated tau as paired helical filaments (PHFs) then in subsequent deposition of PHFs as NFTs. Beginning with the structure of Roscovitine, a moderately selective CDK5 inhibitor, we sought to conduct structural modifications to increase inhibitory potency of CDK5 and increase selectivity over a similar enzyme, cyclin dependent kinase 2 (CDK2). The design, synthesis, and testing of a series of 1-isopropyl-4-aminobenzyl-6-ether-linked benzimidazoles is presented.
Collapse
|
39
|
Dolle RE, Bourdonnec BL, Worm K, Morales GA, Thomas CJ, Zhang W. Comprehensive survey of chemical libraries for drug discovery and chemical biology: 2009. JOURNAL OF COMBINATORIAL CHEMISTRY 2010; 12:765-806. [PMID: 20923157 PMCID: PMC4140011 DOI: 10.1021/cc100128w] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Roland E Dolle
- Adolor Corporation, 700 Pennsylvania Drive, Exton, Pennsylvania 19341, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Ma XH, Wang R, Tan CY, Jiang YY, Lu T, Rao HB, Li XY, Go ML, Low BC, Chen YZ. Virtual screening of selective multitarget kinase inhibitors by combinatorial support vector machines. Mol Pharm 2010; 7:1545-60. [PMID: 20712327 DOI: 10.1021/mp100179t] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Multitarget agents have been increasingly explored for enhancing efficacy and reducing countertarget activities and toxicities. Efficient virtual screening (VS) tools for searching selective multitarget agents are desired. Combinatorial support vector machines (C-SVM) were tested as VS tools for searching dual-inhibitors of 11 combinations of 9 anticancer kinase targets (EGFR, VEGFR, PDGFR, Src, FGFR, Lck, CDK1, CDK2, GSK3). C-SVM trained on 233-1,316 non-dual-inhibitors correctly identified 26.8%-57.3% (majority >36%) of the 56-230 intra-kinase-group dual-inhibitors (equivalent to the 50-70% yields of two independent individual target VS tools), and 12.2% of the 41 inter-kinase-group dual-inhibitors. C-SVM were fairly selective in misidentifying as dual-inhibitors 3.7%-48.1% (majority <20%) of the 233-1,316 non-dual-inhibitors of the same kinase pairs and 0.98%-4.77% of the 3,971-5,180 inhibitors of other kinases. C-SVM produced low false-hit rates in misidentifying as dual-inhibitors 1,746-4,817 (0.013%-0.036%) of the 13.56 M PubChem compounds, 12-175 (0.007%-0.104%) of the 168 K MDDR compounds, and 0-84 (0.0%-2.9%) of the 19,495-38,483 MDDR compounds similar to the known dual-inhibitors. C-SVM was compared to other VS methods Surflex-Dock, DOCK Blaster, kNN and PNN against the same sets of kinase inhibitors and the full set or subset of the 1.02 M Zinc clean-leads data set. C-SVM produced comparable dual-inhibitor yields, slightly better false-hit rates for kinase inhibitors, and significantly lower false-hit rates for the Zinc clean-leads data set. Combinatorial SVM showed promising potential for searching selective multitarget agents against intra-kinase-group kinases without explicit knowledge of multitarget agents.
Collapse
Affiliation(s)
- X H Ma
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore 117543
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yuan L, Seo JS, Kang NS, Keinan S, Steele SE, Michelotti GA, Wetsel WC, Beratan DN, Gong YD, Lee TH, Hong J. Identification of 3-hydroxy-2-(3-hydroxyphenyl)-4H-1-benzopyran-4-ones as isoform-selective PKC-zeta inhibitors and potential therapeutics for psychostimulant abuse. MOLECULAR BIOSYSTEMS 2009; 5:927-30. [PMID: 19668856 DOI: 10.1039/b903036k] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
From a screen of small molecule libraries to identify potential therapeutics for psychostimulant abuse, 3-hydroxy-2-(3-hydroxyphenyl)-4H-1-benzopyran-4-ones were shown to be isoform-selective PKC-zeta inhibitors.
Collapse
Affiliation(s)
- Langtian Yuan
- Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|