Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Molecules ; 26(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34443486

ABSTRACT

Phosphorylation events catalyzed by protein kinases represent one of the most prevalent as well as important regulatory posttranslational modifications, and dysregulation of protein kinases is associated with the pathogenesis of different diseases. Therefore, interest in developing potent small molecule kinase inhibitors has increased enormously within the last two decades. A critical step in the development of new inhibitors is cell-free in vitro testing with the intention to determine comparable parameters like the commonly used IC50 value. However, values described in the literature are often biased as experimental setups used for determination of kinase activity lack comparability due to different readout parameters, insufficient normalization or the sheer number of experimental approaches. Here, we would like to hold a brief for highly sensitive, radioactive-based in vitro kinase assays especially suitable for kinases exhibiting autophosphorylation activity. Therefore, we demonstrate a systematic workflow for complementing and validating results from high-throughput screening as well as increasing the comparability of enzyme-specific inhibitor parameters for radiometric as well as non-radiometric assays. Using members of the CK1 family of serine/threonine-specific protein kinases and established CK1-specific inhibitors as examples, we clearly demonstrate the power of our proposed workflow, which has the potential to support the generation of more comparable data for biological characterization of kinase inhibitors.


Subject(s)
Casein Kinase I/antagonists & inhibitors , Enzyme Assays/methods , Protein Kinase Inhibitors/chemistry , In Vitro Techniques , Inhibitory Concentration 50 , Kinetics , Phosphorylation
2.
PLoS One ; 16(4): e0249908, 2021.
Article in English | MEDLINE | ID: mdl-33861760

ABSTRACT

The single mitochondrial nucleoid (kinetoplast) of Trypanosoma brucei is found proximal to a basal body (mature (mBB)/probasal body (pBB) pair). Kinetoplast inheritance requires synthesis of, and scission of kinetoplast DNA (kDNA) generating two kinetoplasts that segregate with basal bodies into daughter cells. Molecular details of kinetoplast scission and the extent to which basal body separation influences the process are unavailable. To address this topic, we followed basal body movements in bloodstream trypanosomes following depletion of protein kinase TbCK1.2 which promotes kinetoplast division. In control cells we found that pBBs are positioned 0.4 um from mBBs in G1, and they mature after separating from mBBs by at least 0.8 um: mBB separation reaches ~2.2 um. These data indicate that current models of basal body biogenesis in which pBBs mature in close proximity to mBBs may need to be revisited. Knockdown of TbCK1.2 produced trypanosomes containing one kinetoplast and two nuclei (1K2N), increased the percentage of cells with uncleaved kDNA 400%, decreased mBB spacing by 15%, and inhibited cytokinesis 300%. We conclude that (a) separation of mBBs beyond a threshold of 1.8 um correlates with division of kDNA, and (b) TbCK1.2 regulates kDNA scission. We propose a Kinetoplast Division Factor hypothesis that integrates these data into a pathway for biogenesis of two daughter mitochondrial nucleoids.


Subject(s)
Basal Bodies/physiology , Casein Kinase I/metabolism , DNA, Kinetoplast/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/genetics , Cytokinesis/physiology , Cytoplasm/metabolism , DNA Cleavage , DNA Replication , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
3.
Molecules ; 26(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33579052

ABSTRACT

Microtubules composed of α/ß tubulin heterodimers are an essential part of the cytoskeleton of eukaryotic cells and are widely regarded as targets for cancer chemotherapy. IC261, which is discovered as an ATP-competitive inhibitor of serine/threonine-specific casein kinase 1 (CK1), has shown its inhibitory activity on microtubule polymerization in recent studies. However, the structural information of the interaction between tubulin and IC261 is still unclear. Here, we provided a high-resolution (2.85 Å) crystal structure of tubulin and IC261 complex, revealed the intermolecular interaction between tubulin and IC261, and analyzed the structure-activity relationship (SAR). Subsequently, the structure of tubulin-IC261 complex was compared with tubulin-colchicine complex to further elucidate the novelty of IC261. Furthermore, eight optimal candidate compounds of new IC261-based microtubule inhibitors were obtained through molecular docking studies. In conclusion, the co-crystal structure of tubulin-IC261 complex paves a way for the design and development of microtubule inhibitor drugs.


Subject(s)
Casein Kinase I/antagonists & inhibitors , Drug Design , Indoles/chemistry , Microtubules/drug effects , Phloroglucinol/analogs & derivatives , Tubulin/chemistry , Animals , Binding Sites , Colchicine/chemistry , Colchicine/metabolism , Crystallography, X-Ray , Indoles/metabolism , Molecular Docking Simulation , Phloroglucinol/chemistry , Phloroglucinol/metabolism , Protein Conformation , Structure-Activity Relationship , Swine , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/metabolism
4.
Future Med Chem ; 13(5): 505-528, 2021 03.
Article in English | MEDLINE | ID: mdl-33438471

ABSTRACT

Casein kinase 1 (CK1) is an extensively expressed serine/threonine kinase family, with six highly conserved isoforms of human CK1. Due to its involvement in many biological processes, CK1 is a promising target for several pathological states, including circadian sleep disorder, neurodegenerative diseases, cancer and inflammation. However, due to the structural similarities between the six CK1 members, the design of CK1 inhibitors is intricate. So far, no effective CK1 inhibitors are reported to reach clinical trials; thus, approaches to obtaining both selective and effective CK1 inhibitors are in great demand. Here we analyze several CK1 inhibitors that provide successful experience for structure-based drug design and rational structure modification, which could provide references for further drug design.


Subject(s)
Casein Kinase I/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Binding Sites , Casein Kinase I/metabolism , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/therapeutic use , Molecular Dynamics Simulation , Neoplasms/drug therapy , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Oxindoles/chemistry , Oxindoles/metabolism , Oxindoles/therapeutic use , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/therapeutic use
5.
Eur J Med Chem ; 210: 113088, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33316691

ABSTRACT

The family with sequence similarity 20, member C (Fam20C), a Golgi casein kinase, has been recently regarded as a potential therapeutic target for the treatment of triple negative breast cancer (TNBC). Lacking enzyme activity center has been becoming an obstacle to the development of small-molecule inhibitors of Fam20C. Herein, we combined in silico high-throughput screening with chemical synthesis methods to obtain a new small-molecule Fam20C inhibitor 3r, which exhibited desired anti-proliferative activities against MDA-MB-231 cells and also inhibited migration. Subsequently, the enzymatic assay, molecular docking, and molecular dynamics (MD) simulations were carried out for validating that 3r could bind to Fam20C. In addition, 3r was found to induce apoptosis via the mitochondrial pathway in MDA-MB-231 cells as well as to inhibit cell migration. Moreover, we demonstrated that 3r inhibited tumor growth in vivo and thereby having a good therapeutic potential on TNBC. Taken together, these results suggest that 3r may be a novel Fam20C inhibitor with anti-proliferative and apoptosis-inducing activities, which would shed light on discovering more small-molecule drugs for the future TNBC therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Casein Kinase I/antagonists & inhibitors , Drug Discovery , Extracellular Matrix Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Casein Kinase I/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Extracellular Matrix Proteins/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
6.
Eur J Med Chem ; 210: 112956, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33148491

ABSTRACT

Leishmaniasis constitutes a severe public health problem, with an estimated prevalence of 12 million cases. This potentially fatal disease has a worldwide distribution and in 2012, the fatal Visceral Leishmaniasis (VL) was declared as new emerging disease in Europe, mainly due to global warming, with expected important public health impact. The available treatments are toxic, costly or lead to parasite resistance, thus there is an urgent need for new drugs with new mechanism of action. Previously, we reported the discovery of CTN1122, a potent imidazo[1,2-a]pyrazine-based antileishmanial hit compound targeting L-CK1.2 at low micromolar ranges. Here, we described structurally related, safe and selective compounds endowed with antiparasitic properties, better than miltefosine, the reference therapy by oral route. L-CK1.2 homology model gave the first structural explanations of the role of 4-pyridyl (CTN1122) and 2-aminopyrimidin-4-yl (compound 21) moieties, at the position 3 of the central core, in the low micromolar to nanomolar L-CK1.2 inhibition, whereas N-methylpyrazole derivative 11 remained inactive against the parasite kinase.


Subject(s)
Casein Kinase I/antagonists & inhibitors , Imidazoles/pharmacology , Leishmania major/enzymology , Pyrazines/pharmacology , Trypanocidal Agents/pharmacology , Casein Kinase I/metabolism , Humans , Imidazoles/chemistry , Leishmania major/drug effects , Leishmania major/metabolism , Leishmaniasis/drug therapy , Leishmaniasis/parasitology , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazines/chemistry , Trypanocidal Agents/chemistry
7.
Int J Mol Sci ; 21(23)2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33261128

ABSTRACT

The casein kinase 1 enzymes (CK1) form a family of serine/threonine kinases with seven CK1 isoforms identified in humans. The most important substrates of CK1 kinases are proteins that act in the regulatory nodes essential for tumorigenesis of hematological malignancies. Among those, the most important are the functions of CK1s in the regulation of Wnt pathways, cell proliferation, apoptosis and autophagy. In this review we summarize the recent developments in the understanding of biology and therapeutic potential of the inhibition of CK1 isoforms in the pathogenesis of chronic lymphocytic leukemia (CLL), other non-Hodgkin lymphomas (NHL), myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and multiple myeloma (MM). CK1δ/ε inhibitors block CLL development in preclinical models via inhibition of WNT-5A/ROR1-driven non-canonical Wnt pathway. While no selective CK1 inhibitors have reached clinical stage to date, one dual PI3Kδ and CK1ε inhibitor, umbralisib, is currently in clinical trials for CLL and NHL patients. In MDS, AML and MM, inhibition of CK1α, acting via activation of p53 pathway, showed promising preclinical activities and the first CK1α inhibitor has now entered the clinical trials.


Subject(s)
Casein Kinase I/metabolism , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/enzymology , Molecular Targeted Therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/chemistry , Hematologic Neoplasms/pathology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Wnt Signaling Pathway
8.
Int J Mol Sci ; 21(24)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333737

ABSTRACT

Protein kinases (PKs) are enzymes that catalyze the transfer of the terminal phosphate group from ATP to a protein acceptor, mainly to serine, threonine, and tyrosine residues. PK catalyzed phosphorylation is critical to the regulation of cellular signaling pathways that affect crucial cell processes, such as growth, differentiation, and metabolism. PKs represent attractive targets for drugs against a wide spectrum of diseases, including viral infections. Two different approaches are being applied in the search for antivirals: compounds directed against viral targets (direct-acting antivirals, DAAs), or against cellular components essential for the viral life cycle (host-directed antivirals, HDAs). One of the main drawbacks of DAAs is the rapid emergence of drug-resistant viruses. In contrast, HDAs present a higher barrier to resistance development. This work reviews the use of chemicals that target cellular PKs as HDAs against virus of the Flaviviridae family (Flavivirus and Hepacivirus), thus being potentially valuable therapeutic targets in the control of these pathogens.


Subject(s)
Flaviviridae Infections/drug therapy , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Antiviral Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Flaviviridae/drug effects , Flaviviridae/enzymology , Flaviviridae Infections/enzymology , Hepacivirus/enzymology , Hepacivirus/metabolism , Hepatitis C, Chronic/metabolism , Humans , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism
9.
Cell Physiol Biochem ; 54(4): 605-614, 2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32543797

ABSTRACT

BACKGROUND/AIMS: Suicidal erythrocyte death (eryptosis) is characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface following a Ca2+ entry in the cell. Eryptosis is stimulated by increased cytosolic Ca2+ ([Ca2+]i), oxidative stress, energy depletion, or high osmotic shock. Eryptosis signaling includes p38 mitogen-activated protein kinase (MAPK), caspases, casein kinase 1 (CK1), janus kinase 3 (JAK3), and protein kinase C (PKC). Dog and human erythrocytes have different characteristics, for example, dog erythrocytes lack Na+/K+- ATPase activity. Whether eryptosis occurs in dog erythrocytes in an analogous way as that in humans remains unclear. Eryptosis in dogs has not been investigated. This study aimed to explore which stimulator and signaling molecules are involved in eryptosis in healthy dog erythrocytes. METHODS: Erythrocytes were isolated from 10 dogs, and eryptosis was stimulated by oxidative stress with tert-butyl hydroperoxide (tBOOH), high osmotic shock with excessive sucrose condition, energy depletion with minus glucose condition, and high [Ca2+]i with ionomycin. Phosphatidylserine exposure was estimated using annexin V binding. Erythrocyte volume and [Ca2+]i were measured by forward scatter and Fluo3-fluorescence, respectively. In addition, the role of certain mediators was assessed using the following inhibitors to determine the detailed mechanisms of eryptosis in dog erythrocytes: p38MAPK, caspase family, CK1, JAK3, and PKC inhibitors. RESULTS: All eryptosis-inducing factors resulted in phosphatidylserine exposures, except for ionomycin. In addition, the erythrocyte volume increased with ionomycin and tBOOH but decreased with excessive sucrose and minus glucose condition. All treatments increased [Ca2+]i. Furthermore, WH1-P154 and chelerythrine significantly blunted the increase of annexin V binding erythrocytes following the tBOOH treatment. CONCLUSION: Eryptosis in dogs is triggered by oxidative stress, hyperosmotic shock, and energy depletion. It is suggested that JAK3 and PKC play an important role in eryptosis following an oxidative stress in dog erythrocytes.


Subject(s)
Calcium/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Phosphatidylserines/metabolism , tert-Butylhydroperoxide/pharmacology , Animals , Annexin A5/metabolism , Benzophenanthridines/pharmacology , Casein Kinase I/antagonists & inhibitors , Caspase Inhibitors , Caspases/metabolism , Cell Size/drug effects , Dogs , Eryptosis , Glucose/metabolism , Ionomycin/pharmacology , Janus Kinase 3/antagonists & inhibitors , Osmotic Pressure/drug effects , Oxidative Stress/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Sucrose/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
Bioorg Chem ; 99: 103806, 2020 06.
Article in English | MEDLINE | ID: mdl-32279038

ABSTRACT

The optimization and synthesis of new CK2 and CK1 inhibitors are the basis for the development of new therapeutic strategies for the treatment of cancer and neurodegenerative disorders associated with overexpression and abnormal functioning of these enzymes. Triazole derivatives appear to be especially interesting as potential kinase inhibitors. In this context we synthesized a series of 1,2,4-triazolin-5-thione derivatives as CK1γ kinase inhibitors. The antiproliferative activity of synthesized compounds was assessed against cancer cells: human lung adenocarcinoma (A549), human hepatoma (HepG2), and human breast adenocarcinoma (MCF-7). Compound 1 exhibited antiproliferative potency against A549 cancer cells and was characterized by a selective antiproliferative effect. Additionally, this compound has high apoptotic activity against A549, HepG2, MCF-7 cells and induced only slight amount of necrotic cells in these cell lines. In order to decipher the mechanism of anticancer activity of the studied compounds PASS software was used and these compounds were assayed for the inhibition of CK1γ and CK2α kinases. The reported series of 1,2,4-triazolin-5-thiones inhibits CK1γ and CK2α kinases in micromolar range. The most active compound shows activity against isoform γ3 which at concentration of 50 µM reduced the kinase activity by 69% while at 100 µM by 80%. CK2α was found to be less susceptible to the effects of the triazoles tested, as the reduction in kinase activity by 29% was observed for compound 15, and by 27% for compound 1 only at the concentration of 100 µM. The inhibition of CK1γ and CK2α kinases was rationalized using molecular docking.


Subject(s)
Antineoplastic Agents/pharmacology , Casein Kinase I/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Casein Kinase I/metabolism , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
11.
Nat Commun ; 11(1): 384, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959826

ABSTRACT

Emergence of an aggressive androgen receptor (AR)-independent neuroendocrine prostate cancer (NEPC) after androgen-deprivation therapy (ADT) is well-known. Nevertheless, the majority of advanced-stage prostate cancer patients, including those with SPINK1-positive subtype, are treated with AR-antagonists. Here, we show AR and its corepressor, REST, function as transcriptional-repressors of SPINK1, and AR-antagonists alleviate this repression leading to SPINK1 upregulation. Increased SOX2 expression during NE-transdifferentiation transactivates SPINK1, a critical-player for maintenance of NE-phenotype. SPINK1 elicits epithelial-mesenchymal-transition, stemness and cellular-plasticity. Conversely, pharmacological Casein Kinase-1 inhibition stabilizes REST, which in cooperation with AR causes SPINK1 transcriptional-repression and impedes SPINK1-mediated oncogenesis. Elevated levels of SPINK1 and NEPC markers are observed in the tumors of AR-antagonists treated mice, and in a subset of NEPC patients, implicating a plausible role of SPINK1 in treatment-related NEPC. Collectively, our findings provide an explanation for the paradoxical clinical-outcomes after ADT, possibly due to SPINK1 upregulation, and offers a strategy for adjuvant therapies.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Trypsin Inhibitor, Kazal Pancreatic/metabolism , Androgen Receptor Antagonists/therapeutic use , Animals , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/metabolism , Cell Line, Tumor , Co-Repressor Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Nerve Tissue Proteins/metabolism , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , SOXB1 Transcription Factors/metabolism , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
12.
J Am Chem Soc ; 141(40): 15784-15791, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31509406

ABSTRACT

Circadian clocks, biological timekeepers that are present in almost every cell of our body, are complex systems whose disruption is connected to various diseases. Controlling cellular clock function with high temporal resolution in an inducible manner would yield an innovative approach for the circadian rhythm regulation. In the present study, we present structure-guided incorporation of photoremovable protecting groups into a circadian clock modifier, longdaysin, which inhibits casein kinase I (CKI). Using photodeprotection by UV or visible light (400 nm) as the external stimulus, we have achieved quantitative and light-inducible control over the CKI activity accompanied by an accurate regulation of circadian period in cultured human cells and mouse tissues, as well as in living zebrafish. This research paves the way for the application of photodosing in achieving precise temporal control over the biological timing and opens the door for chronophotopharmacology to deeper understand the circadian clock system.


Subject(s)
Adenine/analogs & derivatives , Casein Kinase I/antagonists & inhibitors , Circadian Clocks/drug effects , Protein Kinase Inhibitors/pharmacology , Ultraviolet Rays , Zebrafish/metabolism , Adenine/chemistry , Adenine/pharmacology , Animals , Cell Line , Circadian Clocks/radiation effects , Circadian Rhythm/drug effects , Circadian Rhythm/radiation effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Humans , Larva/drug effects , Larva/enzymology , Larva/radiation effects , Light Signal Transduction , Mice , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Spleen/drug effects , Spleen/enzymology , Spleen/radiation effects , Time Factors
13.
Sci Rep ; 9(1): 12405, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455847

ABSTRACT

The circadian clock ensures that behavioral and physiological processes occur at appropriate times during the 24-hour day/night cycle, and is regulated at both the cellular and organismal levels. To identify pathways acting on intact animals, we performed a small molecule screen using a luminescent reporter of molecular circadian rhythms in zebrafish larvae. We identified both known and novel pathways that affect circadian period, amplitude and phase. Several drugs identified in the screen did not affect circadian rhythms in cultured cells derived from luminescent reporter embryos or in established zebrafish and mammalian cell lines, suggesting they act via mechanisms absent in cell culture. Strikingly, using drugs that promote or inhibit inflammation, as well as a mutant that lacks microglia, we found that inflammatory state affects circadian amplitude. These results demonstrate a benefit of performing drug screens using intact animals and provide novel targets for treating circadian rhythm disorders.


Subject(s)
Circadian Rhythm/drug effects , Small Molecule Libraries/pharmacology , Zebrafish/physiology , Animals , Animals, Genetically Modified/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/metabolism , Larva/drug effects , Larva/physiology , Protein Kinase Inhibitors/pharmacology , Receptors, Glycine/agonists , Receptors, Glycine/metabolism , Taurine/pharmacology , Zebrafish/growth & development , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
14.
Eur J Med Chem ; 181: 111581, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31400711

ABSTRACT

Casein kinase (CK) is a type of conserved serine/threonine protein kinase that phosphorylates many important proteins in body. Researchers found that CK is involved in a variety of signaling pathways, and also plays an important role in inflammation, cancer, and nervous system diseases. Thus, it is considered to be a promising target for the treatment of related diseases. Many CK small molecule inhibitors have been reported so far, and most are ATP competitive inhibitors. However, these CK inhibitors lack the basic properties required for in vivo use, such as selectivity, cell permeability, metabolic stability, correct pharmacokinetic characteristics, and cellular environment. But small molecule inhibitors still have an advantage in drug research due to their controllable pharmacological and pharmacokinetic properties. CX-4945 discovered by Cylene Pharmaceutical is the only one CK2 inhibitor entering into Phase II clinical trials till now. In recent years, significant advances have been made in the design of non-competitive inhibitors of CK and in the application of multi-target inhibition strategies. Here, we review the published CK inhibitors and analyze their structure-activity relationships (SAR). We also summarized the eutectic structure with identified hot spots to provide a reference for future drug discovery.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Casein Kinase I/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Casein Kinase I/chemistry , Casein Kinase I/metabolism , Casein Kinase II/chemistry , Casein Kinase II/metabolism , Drug Discovery , Humans , Molecular Docking Simulation
15.
Reprod Domest Anim ; 54(8): 1104-1112, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31155763

ABSTRACT

The objectives of present study were to evaluate the effect of casein kinase 1 (CK1) inhibition D4476 on in vitro maturation (IVM) and developmental competence of bovine oocytes. The cumulus oocyte complexes (COCs) were cultured in maturation medium with D4476 (0, 2, 5, 10, 20 µM) for 24 hr. After IVM and in vitro fertilization, through expansion average scores of cumulus cells (CCs), oocyte maturation efficiency, cleavage rate and blastocyst rate of zygote, we found 5 µM D4476 could increase the development potential of oocytes. After the COCs were treated with 5 µM D4476, the results of quantitative real-time PCR analysis, Lichen red staining and PI staining showed that under without affecting germinal vesicle breakdown and nuclear morphology, D4476 could significantly decrease CK1 and upregulate TCF-4 in oocytes. Furthermore, without influencing the level of Bad and CTSB, D4476 could significantly increase the expression of ß-catenin, TCF-4, Cx43, MAPK, PTGS-2, PTX-3, TGS-6, Bax and Bcl-2 in CCs. Western blot analysis revealed that the addition of 5 µM D4476 during the maturation of COCs resulted in a lower level of Cx43 protein at 12 hr and a higher expression of Cx43 protein at 24 hr compared to the group without D4476. These results indicate that adding optimum D4476 (5 µM) to maturation medium is beneficial to maturity efficiency and development competence of bovine oocytes.


Subject(s)
Benzamides/pharmacology , Casein Kinase I/antagonists & inhibitors , Cattle , Fertilization in Vitro/veterinary , Imidazoles/pharmacology , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/physiology , Animals , Casein Kinase I/metabolism , Embryo Culture Techniques , Embryo, Mammalian/physiology , Embryonic Development , Fertilization in Vitro/drug effects , In Vitro Oocyte Maturation Techniques/methods , Male , Meiosis
16.
Mol Neurobiol ; 56(8): 5273-5286, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30592011

ABSTRACT

Mutations and variations in the leucine-rich repeat kinase 2 (LRRK2) gene are strongly associated with an increased risk to develop Parkinson's disease (PD). Most pathogenic LRRK2 mutations display increased kinase activity, which is believed to underlie LRRK2-mediated toxicity. Therefore, major efforts have been invested in the development of potent and selective LRRK2 kinase inhibitors. Several of these compounds have proven beneficial in cells and in vivo, even in a LRRK2 wild-type background. Therefore, LRRK2 kinase inhibition holds great promise as disease-modifying PD therapy, and is currently tested in preclinical and early clinical studies. One of the safety concerns is the development of lung pathology in mice and non-human primates, which is most likely related to the strongly reduced LRRK2 protein levels after LRRK2 kinase inhibition. In this study, we aimed to better understand the molecular consequences of chronic LRRK2 kinase inhibition, which may be pivotal in the further development of a LRRK2 kinase inhibitor-based PD therapy. We found that LRRK2 protein levels are not restored during long-term LRRK2 kinase inhibition, but are recovered upon inhibitor withdrawal. Interestingly, LRRK2 kinase inhibitor-induced destabilization does not occur in all pathogenic LRRK2 variants and the N-terminal part of LRRK2 appears to play a crucial role in this process. In addition, we identified CK1, an upstream kinase of LRRK2, as a regulator of LRRK2 protein stability in cell culture and in vivo. We propose that pharmacological LRRK2 kinase inhibition triggers a cascade that results in reduced CK1-mediated phosphorylation of yet unidentified LRRK2 phosphorylation sites. This process involves the N-terminus of LRRK2 and ultimately leads to LRRK2 protein degradation.


Subject(s)
Casein Kinase I/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Animals , Casein Kinase I/metabolism , Cell Line, Tumor , Enzyme Stability/drug effects , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lung/metabolism , Mice, Inbred C57BL , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sulfonamides/pharmacology
17.
Sci Rep ; 8(1): 13621, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30206363

ABSTRACT

Glioblastoma is the most common malignant brain cancer with a dismal prognosis. The difficulty in treating glioblastoma is largely attributed to the lack of effective therapeutic targets. In our previous work, we identified casein kinase 1 ε (CK1ε, also known as CSNK1E) as a potential survival factor in glioblastoma. However, how CK1ε controls cell survival remains elusive and whether targeting CK1ε is a possible treatment for glioblastoma requires further investigation. Here we report that CK1ε was expressed at the highest level among six CK1 isoforms in glioblastoma and enriched in high-grade glioma, but not glia cells. Depletion of CK1ε remarkably inhibited the growth of glioblastoma cells and suppressed self-renewal of glioblastoma stem cells, while having limited effect on astrocytes. CK1ε deprivation activated ß-catenin and induced apoptosis, which was further counteracted by knockdown of ß-catenin. The CK1ε inhibitor IC261, but not PF-4800567, activated ß-catenin and blocked the growth of glioblastoma cells and glioblastoma stem cells. Congruently, IC261 elicited a robust growth inhibition of human glioblastoma xenografts in mice. Together, our results demonstrate that CK1ε regulates the survival of glioblastoma cells and glioblastoma stem cells through ß-catenin signaling, underscoring the importance of targeting CK1ε as an effective treatment for glioblastoma.


Subject(s)
Casein Kinase I/metabolism , Glioblastoma/enzymology , Neoplasm Proteins/metabolism , Signal Transduction , Animals , Casein Kinase I/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays , beta Catenin/metabolism
18.
Bioorg Med Chem Lett ; 28(13): 2234-2238, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29859907

ABSTRACT

The reference standards methyl 3-((2,2-difluoro-5H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]imidazol-6-yl)carbamoyl)benzoate (5a) and N-(2,2-difluoro-5H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]imidazol-6-yl)-3-methoxybenzamide (5c), and their corresponding desmethylated precursors 3-((2,2-difluoro-5H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]imidazol-6-yl)carbamoyl)benzoic acid (6a) and N-(2,2-difluoro-5H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]imidazol-6-yl)-3-hydroxybenzamide (6b), were synthesized from 5-amino-2,2-difluoro-1,3-benzodioxole and 3-substituted benzoic acids in 5 and 6 steps with 33% and 11%, 30% and 7% overall chemical yield, respectively. Carbon-11-labeled casein kinase 1 (CK1) inhibitors, [11C]methyl 3-((2,2-difluoro-5H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]imidazol-6-yl)carbamoyl)benzoate ([11C]5a) and N-(2,2-difluoro-5H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]imidazol-6-yl)-3-[11C]methoxybenzamide ([11C]5c), were prepared from their O-desmethylated precursor 6a or 6b with [11C]CH3OTf through O-[11C]methylation and isolated by HPLC combined with SPE in 40-45% radiochemical yield, based on [11C]CO2 and decay corrected to end of bombardment (EOB). The radiochemical purity was >99%, and the molar activity (MA) at EOB was 370-740 GBq/µmol with a total synthesis time of ∼40-min from EOB.


Subject(s)
Benzimidazoles/chemical synthesis , Benzoates/chemical synthesis , Casein Kinase I/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Alzheimer Disease/diagnostic imaging , Benzamides/chemical synthesis , Benzamides/chemistry , Benzimidazoles/chemistry , Benzoates/chemistry , Carbon Radioisotopes , Hydrophobic and Hydrophilic Interactions , Positron-Emission Tomography , Protein Kinase Inhibitors/chemistry , Radiopharmaceuticals/chemistry
19.
J Enzyme Inhib Med Chem ; 33(1): 665-670, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29536773

ABSTRACT

Several neurodegenerative disorders including Alzheimer's disease (AD) have been connected with deregulation of casein kinase 1 (CK1) activity. Inhibition of CK1 therefore presents a potential therapeutic strategy against such pathologies. Recently, novel class of CK1-specific inhibitors with N-(benzo[d]thiazol-2-yl)-2-phenylacetamide structural scaffold has been discovered. 1-(benzo[d]thiazol-2-yl)-3-phenylureas, on the other hand, are known inhibitors amyloid-beta binding alcohol dehydrogenase (ABAD), an enzyme also involved in pathophysiology of AD. Based on their tight structural similarity, we decided to evaluate series of previously published benzothiazolylphenylureas, originally designed as ABAD inhibitors, for their inhibitory activity towards CK1. Several compounds were found to be submicromolar CK1 inhibitors. Moreover, two compounds were found to inhibit both, ABAD and CK1. Such dual-activity could be of advantage for AD treatment, as it would simultaneously target two distinct pathological processes involved in disease's progression. Based on PAMPA testing both compounds were suggested to permeate the blood-brain barrier, which makes them, together with their unique dual activity, interesting lead compounds for further development.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/metabolism , Casein Kinase I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neurodegenerative Diseases/drug therapy , Phenylurea Compounds/pharmacology , Casein Kinase I/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Neurodegenerative Diseases/metabolism , Phenylurea Compounds/chemistry , Structure-Activity Relationship
20.
Nat Struct Mol Biol ; 25(3): 261-269, 2018 03.
Article in English | MEDLINE | ID: mdl-29483652

ABSTRACT

The survival rate of cancer patients is steadily increasing, owing to more efficient therapies. Understanding the molecular mechanisms of chemotherapy-induced premature ovarian insufficiency (POI) could identify targets for prevention of POI. Loss of the primordial follicle reserve is the most important cause of POI, with the p53 family member p63 being responsible for DNA-damage-induced apoptosis of resting oocytes. Here, we provide the first detailed mechanistic insight into the activation of p63, a process that requires phosphorylation by both the priming kinase CHK2 and the executioner kinase CK1 in mouse primordial follicles. We further describe the structural changes induced by phosphorylation that enable p63 to adopt its active tetrameric conformation and demonstrate that previously discussed phosphorylation by c-Abl is not involved in this process. Inhibition of CK1 rescues primary oocytes from doxorubicin and cisplatin-induced apoptosis, thus uncovering a new target for the development of fertoprotective therapies.


Subject(s)
Casein Kinase I/metabolism , Checkpoint Kinase 2/metabolism , DNA Damage , Oocytes/enzymology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Casein Kinase I/antagonists & inhibitors , Cell Line, Tumor , Cisplatin/toxicity , Doxorubicin/toxicity , Humans , Mice , Oocytes/drug effects , Oocytes/metabolism , Phosphorylation , Protein Multimerization
SELECTION OF CITATIONS
SEARCH DETAIL
...