Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 147: 107378, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643562

ABSTRACT

Alzheimer's disease (AD) is an enigmatic neurological illness that offers few treatment options. Recent exploration has highlighted the crucial connection of the Wnt signaling pathway in AD pathogenesis, shedding light on potential therapeutic targets. The present study focuses on the dual targeting of glycogen synthase kinase-3ß (GSK-3ß) and casein kinase-1δ (CK-1δ) within the framework of the Wnt signaling pathway as a possible technique for AD intervention. GSK-3ß and CK-1δ are multifunctional kinases known for their roles in tau hyperphosphorylation, amyloid processing, and synaptic dysfunction, all of which are major hallmarks of Alzheimer's disease. They are intricately linked to Wnt signaling, which plays a pivotal part in sustaining neuronal function and synaptic plasticity. Dysregulation of the Wnt pathway in AD contributes to cognitive decline and neurodegeneration. This review delves into the molecular mechanisms by which GSK-3ß and CK-1δ impact the Wnt signaling pathway, elucidating their roles in AD pathogenesis. We discuss the potential of small-molecule inhibitors along with their SAR studies along with the multi-targetd approach targeting GSK-3ß and CK-1δ to modulate Wnt signaling and mitigate AD-related pathology. In summary, the dual targeting of GSK-3ß and CK-1δ within the framework of the Wnt signaling pathway presents an innovative and promising avenue for future AD therapies, offering new hope for patients and caregivers in the quest to combat this challenging condition.


Subject(s)
Alzheimer Disease , Glycogen Synthase Kinase 3 beta , Wnt Signaling Pathway , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , Wnt Signaling Pathway/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Molecular Structure , Animals , Structure-Activity Relationship
2.
Int J Mol Sci ; 23(10)2022 May 10.
Article in English | MEDLINE | ID: mdl-35628143

ABSTRACT

Protein kinases are responsible for healthy cellular processes and signalling pathways, and their dysfunction is the basis of many pathologies. There are numerous small molecule inhibitors of protein kinases that systemically regulate dysfunctional signalling processes. However, attaining selectivity in kinase inhibition within the complex human kinome is still a challenge that inspires unconventional approaches. One of those approaches is photopharmacology, which uses light-controlled bioactive molecules to selectively activate drugs only at the intended space and time, thereby avoiding side effects outside of the irradiated area. Still, in the context of kinase inhibition, photopharmacology has thus far been rather unsuccessful in providing light-controlled drugs. Here, we present the discovery and optimisation of a photoswitchable inhibitor of casein kinase 1δ (CK1δ), important for the control of cell differentiation, circadian rhythm, DNA repair, apoptosis, and numerous other signalling processes. Varying the position at which the light-responsive azobenzene moiety has been introduced into a known CK1δ inhibitor, LH846, revealed the preferred regioisomer for efficient photo-modulation of inhibitory activity, but the photoswitchable inhibitor suffered from sub-optimal (photo)chemical properties. Replacement of the bis-phenyl azobenzene group with the arylazopyrazole moiety yielded a superior photoswitch with very high photostationary state distributions, increased solubility and a 10-fold difference in activity between irradiated and thermally adapted samples. The reasons behind those findings are explored with molecular docking and molecular dynamics simulations. Results described here show how the evaluation of privileged molecular architecture, followed by the optimisation of the photoswitchable unit, is a valuable strategy for the challenging design of the photoswitchable kinase inhibitors.


Subject(s)
Casein Kinase Idelta , Protein Kinase Inhibitors , Pyrazoles , Apoptosis/drug effects , Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/metabolism , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology
3.
Cell Rep ; 36(11): 109687, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525355

ABSTRACT

Mammalian cell cytoskeletal reorganization for efficient directional movement requires tight coordination of actomyosin and microtubule networks. In this study, we show that LRAP35a potentiates microtubule stabilization by promoting CLASP2/EB1 interaction besides its complex formation with MRCK/MYO18A for retrograde actin flow. The alternate regulation of these two networks by LRAP35a is tightly regulated by a series of phosphorylation events that dictated its specificity. Sequential phosphorylation of LRAP35a by Protein Kinase A (PKA) and Glycogen Synthase Kinase-3ß (GSK3ß) initiates the association of LRAP35a with CLASP2, while subsequent binding and further phosphorylation by Casein Kinase 1δ (CK1δ) induce their dissociation, which facilitates LRAP35a/MRCK association in driving lamellar actomyosin flow. Importantly, microtubule dynamics is directly moderated by CK1δ activity on CLASP2 to regulate GSK3ß phosphorylation of the SxIP motifs that blocks EB1 binding, an event countered by LRAP35a interaction and its competition for CK1δ activity. Overall this study reveals an essential role for LRAP35a in coordinating lamellar contractility and microtubule polarization in cell migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Casein Kinase Idelta/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Tumor Suppressor Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/genetics , Cell Line, Tumor , Cell Movement , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Microtubule-Associated Proteins/chemistry , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
4.
Int J Mol Sci ; 22(18)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34575906

ABSTRACT

Fragment-Based Drug Discovery (FBDD) has become, in recent years, a consolidated approach in the drug discovery process, leading to several drug candidates under investigation in clinical trials and some approved drugs. Among these successful applications of the FBDD approach, kinases represent a class of targets where this strategy has demonstrated its real potential with the approved kinase inhibitor Vemurafenib. In the Kinase family, protein kinase CK1 isoform δ (CK1δ) has become a promising target in the treatment of different neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In the present work, we set up and applied a computational workflow for the identification of putative fragment binders in large virtual databases. To validate the method, the selected compounds were tested in vitro to assess the CK1δ inhibition.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/chemistry , Drug Discovery/methods , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Binding Sites , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship , Workflow
5.
Nat Commun ; 12(1): 3164, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34039965

ABSTRACT

The circadian clock controls daily rhythms of physiological processes. The presence of the clock mechanism throughout the body is hampering its local regulation by small molecules. A photoresponsive clock modulator would enable precise and reversible regulation of circadian rhythms using light as a bio-orthogonal external stimulus. Here we show, through judicious molecular design and state-of-the-art photopharmacological tools, the development of a visible light-responsive inhibitor of casein kinase I (CKI) that controls the period and phase of cellular and tissue circadian rhythms in a reversible manner. The dark isomer of photoswitchable inhibitor 9 exhibits almost identical affinity towards the CKIα and CKIδ isoforms, while upon irradiation it becomes more selective towards CKIδ, revealing the higher importance of CKIδ in the period regulation. Our studies enable long-term regulation of CKI activity in cells for multiple days and show the reversible modulation of circadian rhythms with a several hour period and phase change through chronophotopharmacology.


Subject(s)
Casein Kinase Ialpha/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Circadian Rhythm/drug effects , Drug Chronotherapy , Protein Kinase Inhibitors/pharmacology , Animals , Casein Kinase Ialpha/metabolism , Casein Kinase Ialpha/ultrastructure , Casein Kinase Idelta/metabolism , Cell Line, Tumor , Chronobiology Disorders/drug therapy , Circadian Clocks/radiation effects , Drug Evaluation, Preclinical , Enzyme Assays , Humans , Light , Mice , Mice, Transgenic , Molecular Docking Simulation , Photoperiod , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/radiation effects , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Tissue Culture Techniques
6.
Eur J Med Chem ; 216: 113331, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33721670

ABSTRACT

Protein kinase CK1δ expression and activity is involved in different pathological situations that include neuroinflammatory and neurodegenerative diseases. For this reason, protein kinase CK1δ has become a possible therapeutic target for these conditions. 5,6-fused bicyclic heteroaromatic systems that resemble adenine of ATP represent optimal scaffolds for the development of a new class of ATP competitive CK1δ inhibitors. In particular, a new series of [1,2,4]triazolo[1,5-c]pyrimidines and [1,2,4]triazolo[1,5-a][1,3,5]triazines was developed. Some crucial interactors have been identified, such as the presence of a free amino group able to interact with the residues of the hinge region at the 5- and 7- positions of the [1,2,4]triazolo[1,5-c]pyrimidine and [1,2,4]triazolo[1,5-a][1,3,5]triazine scaffolds, respectively; or the presence of a 3-hydroxyphenyl or 3,5-dihydroxyphenyl moiety at the 2- position of both nuclei. Molecular modeling studies identified the key interactions involved in the inhibitor-protein recognition process that appropriately fit with the outlined structure-activity relationship. Considering the fact that the CK1 protein kinase is involved in various pathologies in particular of the central nervous system, the interest in the development of new inhibitors permeable to the blood-brain barrier represents today an important goal in the pharmaceutical field. The best potent compound of the series is the 5-(7-amino-5-(benzylamino)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-2-yl)benzen-1,3-diol (compound 51, IC50 = 0.18 µM) that was predicted to have an intermediate ability to cross the membrane in our in vitro assay and represents an optimal starting point to both studies the therapeutic value of protein kinase CK1δ inhibition and to develop new more potent derivatives.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Triazoles/chemistry , Binding Sites , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Casein Kinase Idelta/metabolism , Cell Line , Cell Survival/drug effects , Drug Design , Humans , Kinetics , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Static Electricity , Structure-Activity Relationship , Thermodynamics , Triazines/chemistry , Triazoles/metabolism , Triazoles/pharmacology
7.
Org Biomol Chem ; 18(41): 8402-8413, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33112339

ABSTRACT

Discovery of an anticancer medicine using a single target protein has often been unsuccessful due to the complexity of pathogenic mechanisms as well as the presence of redundant signaling pathways. In this work, we attempted to find promising anticancer drug candidates by simultaneously targeting casein kinase 1 delta (CK1δ) and muscarinic acetylcholine receptor M3 (M3R). Through the structure-based virtual screening and de novo design with the modified potential function for protein-ligand binding, a series of benzo[4,5]imidazo[1,2-a][1,3,5]triazine-2-amine (BITA) derivatives were identified as CK1δ inhibitors and also as M3R antagonists. The biochemical potencies of these bifunctional molecules reached the nanomolar and low-micromolar levels with respect to CK1δ and M3R, respectively. A common interaction feature in the calculated CK1δ-inhibitor and M3R-antagonist complexes is that the BITA moiety is well-stabilized in the orthosteric site of M3R and the hinge region of CK1δ through the establishment of the three hydrogen bonds and the hydrophobic contacts in the vicinity. The computational and experimental results found in this work exemplify the efficiency of kinase and GPCR polypharmacology in developing anticancer medicines.


Subject(s)
Antineoplastic Agents/pharmacology , Casein Kinase Idelta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptor, Muscarinic M3/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Casein Kinase Idelta/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Polypharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor, Muscarinic M3/metabolism , Structure-Activity Relationship
8.
Mol Cancer Ther ; 19(8): 1623-1635, 2020 08.
Article in English | MEDLINE | ID: mdl-32430484

ABSTRACT

Although gemcitabine is the cornerstone of care for pancreatic ductal adenocarcinoma (PDA), patients lack durable responses and relapse is inevitable. While the underlying mechanisms leading to gemcitabine resistance are likely to be multifactorial, there is a strong association between activating gemcitabine metabolism pathways and clinical outcome. This study evaluated casein kinase 1 delta (CK1δ) as a potential therapeutic target for PDA and bladder cancer, in which CK1δ is frequently overexpressed. We assessed the antitumor effects of genetically silencing or pharmacologically inhibiting CK1δ using our in-house CK1δ small-molecule inhibitor SR-3029, either alone or in combination with gemcitabine, on the proliferation and survival of pancreatic and bladder cancer cell lines and orthotopic mouse models. Genetic studies confirmed that silencing CK1δ or treatment with SR-3029 induced a significant upregulation of deoxycytidine kinase (dCK), a rate-limiting enzyme in gemcitabine metabolite activation. The combination of SR-3029 with gemcitabine induced synergistic antiproliferative activity and enhanced apoptosis in both pancreatic and bladder cancer cells. Furthermore, in an orthotopic pancreatic tumor model, we observed improved efficacy with combination treatment concomitant with increased dCK expression. This study demonstrates that CK1δ plays a role in gemcitabine metabolism, and that the combination of CK1δ inhibition with gemcitabine holds promise as a future therapeutic option for metastatic PDA as well as other cancers with upregulated CK1δ expression.


Subject(s)
Breast Neoplasms/drug therapy , Casein Kinase Idelta/antagonists & inhibitors , Deoxycytidine Kinase/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Pancreatic Neoplasms/drug therapy , Urinary Bladder Neoplasms/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Deoxycytidine/pharmacology , Deoxycytidine Kinase/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
9.
Aging (Albany NY) ; 12(7): 5764-5780, 2020 04 13.
Article in English | MEDLINE | ID: mdl-32282334

ABSTRACT

Bladder cancer is the second most common genitourinary malignancy in the world. However, only immune-checkpoint inhibitors and erdafitinib are available to treat advanced bladder cancer. Our previous study reported that 4-((4-(4-ethylpiperazin-1-yl) phenyl)amino)-N-(3,4,5-trichlorophenyl)-7H-pyrrolo-[2, 3-d]pyrimidine-7-carboxamide hydrochloride (13i HCl) is a potent CK1δ inhibitor showing significant anti-bladder cancer activity. In this study, we elucidated the pharmacological mechanisms underlying 13i HCl's inhibition of human bladder cancer. Our results demonstrate that expression of the CSNK1D gene, which codes for CK1δ, is upregulated in superficial and infiltrating bladder cancer patients in two independent datasets. CK1δ knockdown decreased ß-catenin expression in bladder cancer cells and inhibited their growth. Additionally, 13i HCl suppressed bladder cancer cell proliferation and increased apoptosis. We also observed that inhibition of CK1δ using 13i HCl or PF-670462 triggers necroptosis in bladder cancer cells. Finally, 13i HCl inhibited bladder cancer cell migration and reversed their mesenchymal characteristics. These findings suggest further development of 13i HCl as a potential therapeutic agent to treat bladder cancer is warranted.


Subject(s)
Apoptosis/drug effects , Casein Kinase Idelta/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Pyrimidines/pharmacology , Urinary Bladder Neoplasms/metabolism , Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Gene Knockdown Techniques , Humans , Up-Regulation/drug effects , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
10.
Sci Rep ; 10(1): 4449, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32157143

ABSTRACT

Pathogenesis of amyotrophic lateral sclerosis (ALS), a devastating disease where no treatment exists, involves the compartmentalization of the nuclear protein TDP-43 (TAR DNA-binding protein 43) in the cytoplasm which is promoted by its aberrant phosphorylation and others posttranslational modifications. Recently, it was reported that CK-1δ (protein casein kinase-1δ) is able to phosphorylate TDP-43. Here, the preclinical efficacy of a benzothiazole-based CK-1δ inhibitor IGS-2.7, both in a TDP-43 (A315T) transgenic mouse and in a human cell-based model of ALS, is shown. Treatment with IGS-2.7 produces a significant preservation of motor neurons in the anterior horn at lumbar level, a decrease in both astroglial and microglial reactivity in this area, and in TDP-43 phosphorylation in spinal cord samples. Furthermore, the recovery of TDP-43 homeostasis (phosphorylation and localization) in a human-based cell model from ALS patients after treatment with IGS-2.7 is also reported. Moreover, we have shown a trend to increase in CK-1δ mRNA in spinal cord and significantly in frontal cortex of sALS cases. All these data show for the first time the in vivo modulation of TDP-43 toxicity by CK-1δ inhibition with IGS-2.7, which may explain the benefits in the preservation of spinal motor neurons and point to the relevance of CK-1δ inhibitors in a future disease-modifying treatment for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Casein Kinase Idelta/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Motor Neurons/cytology , Protein Kinase Inhibitors/pharmacology , Spinal Cord/cytology , Aged , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Animals , Case-Control Studies , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Motor Neurons/drug effects , Motor Neurons/metabolism , Phosphorylation , Spinal Cord/drug effects , Spinal Cord/metabolism
11.
Int J Biol Sci ; 16(5): 882-892, 2020.
Article in English | MEDLINE | ID: mdl-32071557

ABSTRACT

Casein kinase 1δ (CK1δ) and casein kinase 1ε (CK1ε) have been proposed to be involved in DNA replication, differentiation and apoptosis, thus participating in the regulation of tumorigenesis. However, their functions in colon cancer and the underlying mechanism remain unclear. Here, we found that the expression of CK1ε and CK1δ increased significantly in cancer tissues and the upregulation of CK1ε and CK1δ were closely related to poor differentiation, advanced TNM stage and poor prognosis of colon cancer. CK1δ/ε inhibitor IC261 could induce a decrease in cell survival and proliferation, and an increase in apoptosis in colon cancer cells. Interestingly, IC261 increased the level of aerobic glycolysis in colon cancer cells. Meanwhile, IC261 caused the decrease of p53 protein level and the misregulation of glycolysis related genes (TIGAR, G6PD, GLUT1) which are closely related to the regulation of glycolysis by p53. Inhibiting p53 by siRNA or inhibitor could significantly attenuate the upregulation of aerobic glycolysis induced by IC261. Finally, inhibition of aerobic glycolysis can further increase the cytotoxicity induced by IC261. Collectively, our results revealed that IC261 could inhibit the growth of colon cancer cells and increase the level of aerobic glycolysis, which is regulated by p53-dependent manner. This result suggested that targeting CK1δ/ε and glycolysis might be a valuable strategy treatment and combination therapies for colon cancer.


Subject(s)
Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Colonic Neoplasms/metabolism , Indoles/pharmacology , Phloroglucinol/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Glycolysis/drug effects , Glycolysis/genetics , HCT116 Cells , Humans , Immunohistochemistry , Lactic Acid/metabolism , Phloroglucinol/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
12.
Biochem Biophys Res Commun ; 523(3): 809-815, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31954519

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most deadly cancers worldwide that responds poorly to existing therapies. The Casein kinase 1 (CK1) isoforms CK1δ and CK1ε are reported to be highly expressed in several tumor types, and both genetic and pharmacological inhibition of CK1δ/ε activity has deleterious effects on tumor cell growth. IC261, an CK1δ/ε selectively inhibitor, shows anti-tumor effect against pancreatic tumor and glioblastoma, but its role in HCC remains poorly characterized. In our research, IC261 displayed time- and dose-dependent inhibition of HCC cell proliferation, and induced G2/M arrest and cell apoptosis in vitro. However, the anti-tumor effects of IC261 was independent of CK1δ/ε. Additionally, IC261 was verified to induce centrosome fragmentation during mitosis independent of CK1δ status, and intraperitoneal injection of IC261 to HCCLM3 xenograft models inhibited tumor growth. Taken together, our data indicated that IC261 has therapeutic potential for HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Indoles/therapeutic use , Liver Neoplasms/drug therapy , Phloroglucinol/analogs & derivatives , Protein Kinase Inhibitors/therapeutic use , Animals , Carcinoma, Hepatocellular/metabolism , Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Humans , Indoles/pharmacology , Liver Neoplasms/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Phloroglucinol/pharmacology , Phloroglucinol/therapeutic use , Protein Kinase Inhibitors/pharmacology
13.
CPT Pharmacometrics Syst Pharmacol ; 9(1): 60-69, 2020 01.
Article in English | MEDLINE | ID: mdl-31749321

ABSTRACT

PF-05251749 is a dual inhibitor of casein kinase 1 δ/ε, key regulators of circadian rhythm. As a result of its mechanism of action, PF-05251749 may also change the heart rate corrected QT (QTc) circadian rhythm, which may confound detection of drug-induced QTc prolongation. In this analysis, a nonlinear mixed effect model including a multioscillator function was developed in addition to fitting the prespecified linear mixed effect concentration-QTc model, to identify QTc liability of PF-05251749 in the presence of potential circadian rhythm change. The modeling results suggested lack of clinically meaningful QTc prolongation (upper bound of 90% confidence interval for ∆∆QTc < 10 milliseconds) and that the drug-induced QTc circadian rhythm change was not present. However, simulation results indicated that inference of drug-induced QTc prolongation could be misleading if the drug effect on QTc circadian rhythm is not properly addressed. The modeling and simulation results suggest that prespecification of the concentration-QTc model should be reconsidered for drugs with circadian rhythm modulation potential.


Subject(s)
Circadian Rhythm/drug effects , Long QT Syndrome/chemically induced , Protein Kinase Inhibitors/adverse effects , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Electrocardiography , Humans , Linear Models , Nonlinear Dynamics
14.
Int J Mol Sci ; 20(24)2019 Dec 07.
Article in English | MEDLINE | ID: mdl-31817920

ABSTRACT

Protein kinases of the CK1 family can be involved in numerous physiological and pathophysiological processes. Dysregulated expression and/or activity as well as mutation of CK1 isoforms have previously been linked to tumorigenesis. Among all neoplastic diseases, colon and rectal cancer (CRC) represent the fourth leading cause of cancer related deaths. Since mutations in CK1δ previously found in CRC patients exhibited increased oncogenic features, inhibition of CK1δ is supposed to have promising therapeutic potential for tumors, which present overexpression or mutations of this CK1 isoform. Therefore, it is important to develop new small molecule inhibitors exhibiting higher affinity toward CK1δ mutants. In the present study, we first characterized the kinetic properties of CK1δ mutants, which were detected in different tumor entities. Subsequently, we characterized the ability of several newly developed IWP-based inhibitors to inhibit wild type and CK1δ mutants and we furthermore analyzed their effects on growth inhibition of various cultured colon cancer cell lines. Our results indicate, that these compounds represent a promising base for the development of novel CRC therapy concepts.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Drug Development , Mutant Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Casein Kinase Idelta/genetics , Casein Kinase Idelta/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Humans , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Phosphorylation , Tumor Cells, Cultured
15.
J Vet Med Sci ; 81(11): 1680-1684, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31582600

ABSTRACT

The molecular clock network in mast cells has been shown to be a factor responsible for circadian regulation of allergic inflammation. PF670462 is a selective inhibitor of casein kinase 1δ and ε (CK1δ/ε) that control the posttranslational modification of clock proteins. The aims of this study were to evaluate the effects of PF670462 on gene and protein expression of FcεRI, the high-affinity IgE receptor, in canine mast cells and on IgE-mediated immediate-type cutaneous reactions in dogs. PF670462 decreased mRNA expression of FcεRIα and ß, but not γ, and protein expression of FcεRI in a canine mast cell line. Furthermore, PF670462 suppressed IgE-mediated immediate-type cutaneous erythema in dogs. These findings indicate that CK1δ/ε function as regulators for FcεRI expression and IgE-mediated cutaneous reactions in dogs.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/antagonists & inhibitors , Dog Diseases/metabolism , Immunoglobulin E/metabolism , Pyrimidines/pharmacology , Receptors, IgE/metabolism , Anaphylaxis , Animals , Casein Kinase 1 epsilon/genetics , Dog Diseases/genetics , Dogs , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Mast Cells/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgE/genetics
16.
Gene ; 715: 144005, 2019 Oct 05.
Article in English | MEDLINE | ID: mdl-31376410

ABSTRACT

Members of the highly conserved pleiotropic CK1 family of serine/threonine-specific kinases are tightly regulated in the cell and play crucial regulatory roles in multiple cellular processes from protozoa to human. Since their dysregulation as well as mutations within their coding regions contribute to the development of various different pathologies, including cancer and neurodegenerative diseases, they have become interesting new drug targets within the last decade. However, to develop optimized CK1 isoform-specific therapeutics in personalized therapy concepts, a detailed knowledge of the regulation and functions of the different CK1 isoforms, their various splice variants and orthologs is mandatory. In this review we will focus on the stress-induced CK1 isoform delta (CK1δ), thereby addressing its regulation, physiological functions, the consequences of its deregulation for the development and progression of diseases, and its potential as therapeutic drug target.


Subject(s)
Casein Kinase Idelta/chemistry , Casein Kinase Idelta/metabolism , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Signal Transduction , Animals , Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/genetics , Drug Delivery Systems/methods , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Structure-Activity Relationship
17.
Mol Syst Biol ; 15(7): e8838, 2019 07.
Article in English | MEDLINE | ID: mdl-31353796

ABSTRACT

In mammals, the master circadian clock synchronizes daily rhythms of physiology and behavior with the day-night cycle. Failure of synchrony, which increases the risk for numerous chronic diseases, can be treated by phase adjustment of the circadian clock pharmacologically, for example, with melatonin, or a CK1δ/ε inhibitor. Here, using in silico experiments with a systems pharmacology model describing molecular interactions, and pharmacokinetic and behavioral experiments in cynomolgus monkeys, we find that the circadian phase delay caused by CK1δ/ε inhibition is more strongly attenuated by light in diurnal monkeys and humans than in nocturnal mice, which are common preclinical models. Furthermore, the effect of CK1δ/ε inhibition strongly depends on endogenous PER2 protein levels, which differs depending on both the molecular cause of the circadian disruption and the patient's lighting environment. To circumvent such large interindividual variations, we developed an adaptive chronotherapeutics to identify precise dosing regimens that could restore normal circadian phase under different conditions. Our results reveal the importance of photosensitivity in the clinical efficacy of clock-modulating drugs, and enable precision medicine for circadian disruption.


Subject(s)
Casein Kinase Idelta/genetics , Circadian Clocks/genetics , Circadian Rhythm/genetics , Light Signal Transduction/genetics , Period Circadian Proteins/genetics , Animals , Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/metabolism , Circadian Clocks/drug effects , Circadian Clocks/radiation effects , Circadian Rhythm/drug effects , Circadian Rhythm/radiation effects , Cryptochromes/genetics , Cryptochromes/metabolism , Drug Administration Schedule , Drug Chronotherapy , Gene Expression Regulation , Humans , Light , Macaca fascicularis , Mice , Period Circadian Proteins/metabolism , Photoperiod , Precision Medicine , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Species Specificity , Systems Biology/methods
18.
J Proteome Res ; 18(9): 3383-3393, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31334659

ABSTRACT

Sleep disturbances and memory impairment are common symptoms of Alzheimer's disease (AD). Given that the circadian clock regulates sleep, hippocampal function, and neurodegeneration, it represents a therapeutic target against AD. Casein kinase 1δ/ε (CK1δ/ε) are clock regulators and overexpressed in AD brains, making them viable targets to improve sleep and cognition. In this study, we evaluated the therapeutic potential of a small molecule CK1δ/ε inhibitor (PF-670462) in a triple transgenic mouse model of AD (3xTg-AD). Mass spectrometry-based proteomic analyses revealed that PF-670462 administration in 3xTg-AD mice reversed hippocampal proteomic alterations in several AD-related and clock-regulated pathways, including synaptic plasticity and amyloid precursor protein processing. Furthermore, PF-670462 administration rescued working memory deficits and normalized behavioral circadian rhythm disturbances in 3xTg-AD mice. Our study provides in vivo proof of concept for CK1δ/ε inhibition against AD-associated hippocampal proteomic changes, memory impairment, and circadian disturbances.


Subject(s)
Alzheimer Disease/therapy , Casein Kinase 1 epsilon/genetics , Casein Kinase Idelta/genetics , Memory Disorders/therapy , Sleep Wake Disorders/therapy , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Circadian Clocks/genetics , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/complications , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Transgenic , Nerve Degeneration/genetics , Neuronal Plasticity/drug effects , Proteomics/methods , Pyrimidines/pharmacology , Sleep Wake Disorders/complications , Sleep Wake Disorders/genetics , Sleep Wake Disorders/pathology
19.
Photochem Photobiol Sci ; 18(6): 1398-1407, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-30924488

ABSTRACT

In photopharmacology, photoswitchable compounds including azobenzene or other diarylazo moieties exhibit bioactivity against a target protein typically in the slender E-configuration, whereas the rather bulky Z-configuration usually is pharmacologically less potent. Herein we report the design, synthesis and photochemical/inhibitory characterization of new photoswitchable kinase inhibitors targeting p38α MAPK and CK1δ. A well characterized inhibitor scaffold was used to attach arylazo- and diazocine moieties. When the isolated isomers, or the photostationary state (PSS) of isomers, were tested in commonly used in vitro kinase assays, however, only small differences in activity were observed. X-ray analyses of ligand-bound p38α MAPK and CK1δ complexes revealed dynamic conformational adaptations of the protein with respect to both isomers. More importantly, irreversible reduction of the azo group to the corresponding hydrazine was observed. Independent experiments revealed that reducing agents such as DTT (dithiothreitol) and GSH (glutathione) that are typically used for protein stabilization in biological assays were responsible. Two further sources of error are the concentration dependence of the E-Z-switching efficiency and artefacts due to incomplete exclusion of light during testing. Our findings may also apply to a number of previously investigated azobenzene-based photoswitchable inhibitors.


Subject(s)
Azocines/pharmacology , Casein Kinase Idelta/antagonists & inhibitors , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Azocines/chemistry , Casein Kinase Idelta/metabolism , Dose-Response Relationship, Drug , Imidazoles/chemistry , Ligands , Mitogen-Activated Protein Kinase 14/metabolism , Models, Molecular , Molecular Structure , Photochemical Processes , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Thiazoles/chemistry
20.
Molecules ; 24(5)2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30832206

ABSTRACT

In this study, we report on the modification of a 3,4-diaryl-isoxazole-based CK1 inhibitor with chiral pyrrolidine scaffolds to develop potent and selective CK1 inhibitors. The pharmacophore of the lead structure was extended towards the ribose pocket of the adenosine triphosphate (ATP) binding site driven by structure-based drug design. For an upscale compatible multigram synthesis of the functionalized pyrrolidine scaffolds, we used a chiral pool synthetic route starting from methionine. Biological evaluation of key compounds in kinase and cellular assays revealed significant effects of the scaffolds towards activity and selectivity, however, the absolute configuration of the chiral moieties only exhibited a limited effect on inhibitory activity. X-ray crystallographic analysis of ligand-CK1δ complexes confirmed the expected binding mode of the 3,4-diaryl-isoxazole inhibitors. Surprisingly, the original compounds underwent spontaneous Pictet-Spengler cyclization with traces of formaldehyde during the co-crystallization process to form highly potent new ligands. Our data suggests chiral "ribose-like" pyrrolidine scaffolds have interesting potential for modifications of pharmacologically active compounds.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Isoxazoles/chemistry , Adenosine Triphosphate/chemistry , Binding Sites , Casein Kinase Idelta/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Isoxazoles/chemical synthesis , Isoxazoles/pharmacology , Ligands , Multiprotein Complexes/chemistry , Pyrrolidines/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...