Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.368
Filter
1.
Biomed Pharmacother ; 174: 116602, 2024 May.
Article in English | MEDLINE | ID: mdl-38636396

ABSTRACT

The development of new molecules for the treatment of calmodulin related cardiovascular or neurodegenerative diseases is an interesting goal. In this work, we introduce a novel strategy with four main steps: (1) chemical synthesis of target molecules, (2) Förster Resonance Energy Transfer (FRET) biosensor development and in vitro biological assay of new derivatives, (3) Cheminformatics models development and in vivo activity prediction, and (4) Docking studies. This strategy is illustrated with a case study. Firstly, a series of 4-substituted Riluzole derivatives 1-3 were synthetized through a strategy that involves the construction of the 4-bromoriluzole framework and its further functionalization via palladium catalysis or organolithium chemistry. Next, a FRET biosensor for monitoring Ca2+-dependent CaM-ligands interactions has been developed and used for the in vitro assay of Riluzole derivatives. In particular, the best inhibition (80%) was observed for 4-methoxyphenylriluzole 2b. Besides, we trained and validated a new Networks Invariant, Information Fusion, Perturbation Theory, and Machine Learning (NIFPTML) model for predicting probability profiles of in vivo biological activity parameters in different regions of the brain. Next, we used this model to predict the in vivo activity of the compounds experimentally studied in vitro. Last, docking study conducted on Riluzole and its derivatives has provided valuable insights into their binding conformations with the target protein, involving calmodulin and the SK4 channel. This new combined strategy may be useful to reduce assay costs (animals, materials, time, and human resources) in the drug discovery process of calmodulin inhibitors.


Subject(s)
Calmodulin , Cardiovascular Agents , Molecular Docking Simulation , Neuroprotective Agents , Riluzole , Riluzole/analogs & derivatives , Riluzole/chemical synthesis , Riluzole/chemistry , Riluzole/pharmacology , Calmodulin/antagonists & inhibitors , Calmodulin/chemistry , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Cardiovascular Agents/chemical synthesis , Cardiovascular Agents/chemistry , Cardiovascular Agents/pharmacology , Drug Development , Molecular Docking Simulation/methods , Biosensing Techniques , Machine Learning , Humans , Animals , Cell Line , Fluorescence Resonance Energy Transfer/methods , Brain/drug effects , Ligands , Protein Conformation
2.
PLoS One ; 16(8): e0255656, 2021.
Article in English | MEDLINE | ID: mdl-34343209

ABSTRACT

Although quiescent hepatic stellate cells (HSCs) have been suggested to regulate hepatic blood flow, there is no direct evidence that quiescent HSCs display contractile abilities. Here, we developed a new method to quantitatively measure the contraction of single isolated HSCs and evaluated whether endothelin-1 (ET-1) induced contraction of HSCs in a non-activated state. HSCs isolated from mice were seeded on collagen gel containing fluorescent beads. The beads around a single HSC were observed gravitating toward the cell upon contraction. By recording the movement of each bead by fluorescent microscopy, the real-time contraction of HSCs was quantitatively evaluated. ET-1 induced a slow contraction of non-activated HSCs, which was inhibited by the non-muscle myosin II inhibitor blebbistatin, the calmodulin inhibitor W-7, and the ETA receptor antagonist ambrisentan. ET-1-induced contraction was also largely reduced in Ca2+-free conditions, but sustained contraction still remained. The tonic contraction was further diminished by the Rho-kinase inhibitor H-1152. The mRNA expression of P/Q-type voltage-dependent Ca2+ channels (VDCC), as well as STIM and Orai, constituents of store-operated channels (SOCs), was observed in mouse non-activated HSCs. ET-1-induced contraction was not affected by amlodipine, a VDCC blocker, whereas it was partly reduced by Gd3+ and amiloride, non-selective cation channel blockers. However, neither YM-58483 nor SKF-96365, which inhibit SOCs, had any effects on the contraction. These results suggest that ET-1 leads to Ca2+-influx through cation channels other than SOCs and produces myosin II-mediated contraction of non-activated HSCs via ETA receptors, as well as via mechanisms involving Ca2+-calmodulin and Rho kinase.


Subject(s)
Cell Physiological Phenomena/drug effects , Endothelin-1/pharmacology , Hepatic Stellate Cells/metabolism , Signal Transduction/drug effects , Animals , Calcium/metabolism , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Cells, Cultured , Endothelin Receptor Antagonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Male , Mice , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Phenylpropionates/pharmacology , Pyridazines/pharmacology , RNA, Messenger/genetics , Receptor, Endothelin A/metabolism , Sulfonamides/pharmacology , rho-Associated Kinases/metabolism
3.
J Biol Chem ; 297(4): 101126, 2021 10.
Article in English | MEDLINE | ID: mdl-34461097

ABSTRACT

Transient receptor potential (TRP) cation channels, which are conserved across mammals, flies, fish, sea squirts, worms, and fungi, essentially contribute to cellular Ca2+ signaling. The activity of the unique TRP channel in yeast, TRP yeast channel 1 (TRPY1), relies on the vacuolar and cytoplasmic Ca2+ concentration. However, the mechanism(s) of Ca2+-dependent regulation of TRPY1 and possible contribution(s) of Ca2+-binding proteins are yet not well understood. Our results demonstrate a Ca2+-dependent binding of yeast calmodulin (CaM) to TRPY1. TRPY1 activity was increased in the cmd1-6 yeast strain, carrying a non-Ca2+-binding CaM mutant, compared with the parent strain expressing wt CaM (Cmd1). Expression of Cmd1 in cmd1-6 yeast rescued the wt phenotype. In addition, in human embryonic kidney 293 cells, hypertonic shock-induced TRPY1-dependent Ca2+ influx and Ca2+ release were increased by the CaM antagonist ophiobolin A. We found that coexpression of mammalian CaM impeded the activity of TRPY1 by reinforcing effects of endogenous CaM. Finally, inhibition of TRPY1 by Ca2+-CaM required the cytoplasmic amino acid stretch E33-Y92. In summary, our results show that TRPY1 is under inhibitory control of Ca2+-CaM and that mammalian CaM can replace yeast CaM for this inhibition. These findings add TRPY1 to the innumerable cellular proteins, which include a variety of ion channels, that use CaM as a constitutive or dissociable Ca2+-sensing subunit, and contribute to a better understanding of the modulatory mechanisms of Ca2+-CaM.


Subject(s)
Calcium Signaling , Calcium/metabolism , Calmodulin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , TRPC Cation Channels/metabolism , Vacuoles/metabolism , Calcium/chemistry , Calmodulin/antagonists & inhibitors , Calmodulin/chemistry , Calmodulin/genetics , HEK293 Cells , Humans , Protein Domains , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sesterterpenes/pharmacology , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics , Vacuoles/chemistry , Vacuoles/genetics
4.
Mol Pharmacol ; 100(3): 237-257, 2021 09.
Article in English | MEDLINE | ID: mdl-34127538

ABSTRACT

Ion channels are attractive drug targets for many therapeutic applications. However, high-throughput screening (HTS) of drug candidates is difficult and remains very expensive. We thus assessed the suitability of the bioluminescence resonance energy transfer (BRET) technique as a new HTS method for ion-channel studies by taking advantage of our recently characterized intra- and intermolecular BRET probes targeting the transient receptor potential vanilloid type 1 (TRPV1) ion channel. These BRET probes monitor conformational changes during TRPV1 gating and subsequent coupling with calmodulin, two molecular events that are intractable using reference techniques such as automated calcium assay (ACA) and automated patch-clamp (APC). We screened the small-sized Prestwick chemical library, encompassing 1200 compounds with high structural diversity, using either intra- and intermolecular BRET probes or ACA. Secondary screening of the detected hits was done using APC. Multiparametric analysis of our results shed light on the capability of calmodulin inhibitors included in the Prestwick library to inhibit TRPV1 activation by capsaicin. BRET was the lead technique for this identification process. Finally, we present data exemplifying the use of intramolecular BRET probes to study other transient receptor potential (TRP) channels and non-TRPs ion channels. Knowing the ease of use of BRET biosensors and the low cost of the BRET technique, these assays may advantageously be included for extending ion-channel drug screening. SIGNIFICANCE STATEMENT: This study screened a chemical library against TRPV1 ion channel using bioluminescence resonance energy transfer (BRET) molecular probes and compared the results with the ones obtained using reference techniques such as automated calcium assay and automated patch-clamp. Multiparametric analysis of our results shed light on the capability of calmodulin antagonists to inhibit chemical activation of TRPV1 and indicates that BRET probes may advantageously be included in ion channel drug screening campaigns.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , TRPV Cation Channels/metabolism , Biological Assay/methods , Calcium/chemistry , Calmodulin/antagonists & inhibitors , HEK293 Cells , Humans , Ligands , Membrane Potentials/drug effects , Patch-Clamp Techniques , Small Molecule Libraries , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors
5.
Anim Sci J ; 92(1): e13552, 2021.
Article in English | MEDLINE | ID: mdl-33890345

ABSTRACT

In mammals, hyperactivation is essential for sperm fertilization with oocytes in vivo. Two types of hyperactivation "full-type and nonfull-type patterns" can be observed in the spermatozoa from boars, bulls, and mice. We have a hypothesis that the full-type hyperactivation is a physiological (in vivo) pattern and are elucidating its molecular bases. The aims of this study were to detect calmodulin in boar sperm flagella by Western blotting and indirect immunofluorescence and to investigate effects of extracellular Ca2+ and calmodulin antagonists "W-7 and W-5 (W-5; a less potent antagonist)" on the occurrence of full-type hyperactivation in boar spermatozoa. Calmodulin was specifically detected as the 17-kDa antigen in the flagella and postacrosomal region of the heads. Full-type hyperactivation could be induced effectively in the samples incubated with 3.42 mM CaCl2 for 120-180 min, and it was significantly reduced in the concentration-dependent manners of W-7 and W-5. Suppressing effects of W-7 on the full-type hyperactivation were stronger than those of W-5. These observations indicate that flagellar calmodulin is involved in the occurrence of extracellular Ca2+ -dependent full-type hyperactivation in boar spermatozoa. This is the first indication of the intracellular Ca2+ -sensing molecule which can function in the full-type hyperactivation.


Subject(s)
Calcium/metabolism , Calmodulin/physiology , Cyclic AMP/pharmacology , Sperm Motility/drug effects , Spermatozoa/physiology , Animals , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Calmodulin/pharmacology , Cattle , Cells, Cultured , Male , Mice , Sulfonamides/pharmacology , Swine
6.
Anticancer Res ; 41(3): 1315-1325, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33788723

ABSTRACT

BACKGROUND/AIM: The aim of this study was to reveal the novel roles of calmodulin 2 (CALM2) in hepatocellular carcinoma (HCC) progression. MATERIALS AND METHODS: The effects of knockdown of CALM2 expression by siRNA were investigated using various experimental approaches in both cellular and molecular levels. RESULTS: Silencing of CALM2 inhibited HCC cell proliferation and colony formation through induction of apoptosis. At the molecular level, CALM2-specific knockdown led to the common dysregulation of 154 genes in HCC cells. Notably, E2F transcription factor 5 (E2F5), which is functionally associated with migration, invasion and proliferation, was generally down-regulated. These functional associations were confirmed in HCC clinical samples. Reflecting the molecular changes, CALM2 knockdown reduced the migration and invasion abilities of HCC cells and abrogated the potency of tumor formation in vivo. CONCLUSION: Targeting CALM2 may be a molecular strategy for both primary HCC treatment and prevention of metastasis or recurrence.


Subject(s)
Calmodulin/physiology , Carcinoma, Hepatocellular/pathology , E2F5 Transcription Factor/physiology , Liver Neoplasms/pathology , Apoptosis/drug effects , Calmodulin/antagonists & inhibitors , Cell Cycle , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Neoplasm Metastasis , Neoplastic Stem Cells/physiology
7.
Dev Comp Immunol ; 116: 103910, 2021 03.
Article in English | MEDLINE | ID: mdl-33129883

ABSTRACT

Unlike vertebrate species, invertebrates lack antigen-antibody mediated immune response and mainly rely on haemocyte phagocytosis to fight against pathogen infection. Recently, studies conducted in model vertebrates demonstrated that the multifunctional protein calmodulin (CaM) plays an important role in regulating immune responses. However, the intrinsic relation between CaM and phagocytosis process remains poorly understood in invertebrate species such as bivalve mollusks. Therefore, in the present study, the immunomodulatory function of CaM on haemocyte phagocytosis was verified in the blood clam, Tegillarca granosa, using the CaM-specific inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride (W-7). Results obtained show that CaM inhibition significantly suppressed the phagocytic activity of haemocytes. In addition, CaM inhibition constrained intracellular Ca2+ elevation, hampered actin cytoskeleton assembly, suppressed calcineurin (CaN) activity, and disrupted NF-κB activation in haemocytes upon LPS induction. Furthermore, expression of seven selected genes from the actin cytoskeleton regulation- and immune-related pathways were significantly downregulated whereas those of CaM and CaN from the Ca2+-signaling pathway were significantly upregulated by in vitro incubation of haemocytes with W-7. For the first time, the present study demonstrated that CaM play an important role in phagocytosis modulation in bivalve species. In addition, the intracellular Ca2+ and downstream Ca2+-signaling-, actin cytoskeleton regulation-, and immune-related pathways offer candidate routes through which CaM modulates phagocytosis.


Subject(s)
Arcidae/drug effects , Calmodulin/antagonists & inhibitors , Gene Expression Regulation/drug effects , Phagocytosis/drug effects , Sulfonamides/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Arcidae/genetics , Arcidae/metabolism , Calcineurin/genetics , Calcineurin/metabolism , Calcium/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Hemocytes/cytology , Hemocytes/drug effects , Hemocytes/metabolism , Immunity/drug effects , Immunity/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Phagocytosis/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
8.
FEBS Open Bio ; 11(1): 75-84, 2021 01.
Article in English | MEDLINE | ID: mdl-33125833

ABSTRACT

The aquaporins (AQPs) are a family of integral membrane proteins which play critical roles in controlling transcellular water movement in various tissues throughout the body. AQP1 helps mediate the cellular response to osmotic stress and tissue water permeability. However, the mechanism by which AQP1 mediates changes in cell volume is not completely clear. Here, we investigated how AQP1 responds to and controls cell volume upon osmotic stimuli during the early phase after the immediate response. Cells overexpressing AQP1 were exposed to hypotonic or hypertonic medium in the presence or absence of staurosporine or W-7 hydrochloride, and fluorescence imaging was performed at 0, 5, 10, and 15 min later. Osmotic stimuli induced redistribution of AQP1 into the cell membrane, hypotonic stimuli caused cell enlargement, and hypertonic stimuli induced a reduction in cell size, which was blocked by T157A/T239A mutations. Changes in cell size induced by osmotic stimuli were blocked by an antagonist of calmodulin kinase, W-7 hydrochloride, but not by the PKC inhibitor staurosporine. These results suggest that calmodulin kinase regulates AQP1 activity during the early response to osmotic stimuli.


Subject(s)
Aquaporin 1/metabolism , Calmodulin/metabolism , Aquaporin 1/genetics , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/antagonists & inhibitors , Cell Membrane/metabolism , Cell Size/drug effects , Culture Media/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mutation , Osmosis/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Staurosporine/pharmacology , Sulfonamides/pharmacology
9.
Drug Des Devel Ther ; 14: 2715-2723, 2020.
Article in English | MEDLINE | ID: mdl-32764873

ABSTRACT

OBJECTIVE: To investigate the neuroprotective effect of chrysophanol (CHR) on PC12 treated with Aß25-35, and the involved mechanism. METHODS: After the establishment of an AD cell model induced by Aß25-35, the cell survival rate was detected by MTT, cell apoptosis was assayed by Hoechst 33342 staining, mRNA expressions of calmodulin (CaM), calcium/calmodulin-dependent protein kinase kinase (CaMKK), calcium/calmodulin-dependent protein kinase IV (CaMKIV) and tau (MAPT; commonly known as tau) were determined by qRT-PCR, and protein levels of CaM, CaMKK, CaMKIV, phospho-CaMKIV (p-CaMKIV), tau and phospho-tau (p-tau) were detected by Western blot analysis. RESULTS: When pretreated with CHR before exposure to Aß25-35, PC12 cells showed that increased cell viability and reduced apoptosis. The qRT-PCR results indicated that the deposition of Aß25-35 triggers a decrease in levels of CaM, CaMKK, CaMKIV, and tau in PC12 cells. In addition, Western blot results also suggested that Aß25-35 decreases the protein expression of CaM, CaMKK, CaMKIV, p-CaMKIV, and the ratio of p-tau to tau in PC12 cells. However, the above effects were significantly alleviated after the treatment of CHR. CONCLUSION: CHR plays a neuroprotective role in AD though decreasing the protein level of CaM-CaMKK-CaMKIV and the expression of p-tau downstream.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Anthraquinones/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Calmodulin/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Anthraquinones/metabolism , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Cell Survival/drug effects , Neuroprotective Agents/metabolism , PC12 Cells , Peptide Fragments/metabolism , Rats , Tumor Cells, Cultured
10.
Anticancer Agents Med Chem ; 20(8): 1017-1027, 2020.
Article in English | MEDLINE | ID: mdl-32271699

ABSTRACT

BACKGROUND: Targeting evolutionarily conserved proteins in malignant cells and the adapter proteins involved in signalling that generates from such proteins may play a cardinal role in the selection of anti-cancer drugs. Drugs targeting these proteins could be of importance in developing anti-cancer drugs. OBJECTIVES: We inferred that drugs like loperamide and promethazine that act as antagonists of proteins conserved in cancer cells like voltage-gated Calcium channels (Cav), Calmodulin (CaM) and drug efflux (ABCB1) pump may have the potential to be re-purposed as an anti-cancer agent in Prostate Cancer (PCa). METHODS: Growth and cytotoxic assays were performed by selecting loperamide and promethazine to target Cav, CaM and drug efflux (ABCB1) pumps to elucidate their effects on androgen-independent PC3 and DU145 PCa cell lines. RESULT: We show that loperamide and promethazine in doses of 80-100µg/ml exert oncocidal effects when tested in DU145 and PC3 cell lines. Diphenhydramine, which shares its targets with promethazine, except the CaM, failed to exhibit oncocidal effects. CONCLUSION: Anti-cancer effects can be of significance if structural analogues of loperamide and promethazine that specifically target Cav, CaM and ABCB1 drug efflux pumps can be synthesized, or these two drugs could be re-purposed after human trials in PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium Channels, L-Type/metabolism , Calmodulin/antagonists & inhibitors , Diphenhydramine/pharmacology , Loperamide/pharmacology , Promethazine/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Androgens/metabolism , Antineoplastic Agents/chemistry , Calmodulin/metabolism , Cell Proliferation/drug effects , Diphenhydramine/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Loperamide/chemistry , Molecular Structure , Promethazine/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Cell Calcium ; 86: 102161, 2020 03.
Article in English | MEDLINE | ID: mdl-31981914

ABSTRACT

SmTAL1 is a calcium binding protein from the parasitic worm, Schistosoma mansoni. Structurally it is comprised of two domains - an N-terminal EF-hand domain and a C-terminal dynein light chain (DLC)-like domain. The protein has previously been shown to interact with the anti-schistosomal drug, praziquantel (PZQ). Here, we demonstrated that both EF-hands in the N-terminal domain are functional calcium ion binding sites. The second EF-hand appears to be more important in dictating affinity and mediating the conformational changes which occur on calcium ion binding. There is positive cooperativity between the four calcium ion binding sites in the dimeric form of SmTAL1. Both the EF-hand domain and the DLC-domain dimerise independently suggesting that both play a role in forming the SmTAL1 dimer. SmTAL1 binds non-cooperatively to PZQ and cooperatively to an IQ-motif from SmCav1B, a voltage-gated calcium channel. PZQ tends to strengthen this interaction, although the relationship is complex. These data suggest the hypothesis that SmTAL1 regulates at least one voltage-gated calcium channel and PZQ interferes with this process. This may be important in the molecular mechanism of this drug. It also suggests that compounds which bind SmTAL1, such as six from the Medicines for Malaria Box identified in this work, may represent possible leads for the discovery of novel antagonists.


Subject(s)
Allergens/metabolism , Ion Channels/chemistry , Praziquantel/pharmacology , Protozoan Proteins/metabolism , Schistosoma mansoni/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Calcium/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , EF Hand Motifs , Ion Channels/metabolism , Ions , Peptides/chemistry , Protein Binding/drug effects , Protein Domains , Protein Multimerization/drug effects , Proteolysis/drug effects , Protozoan Proteins/chemistry
12.
Mol Oncol ; 14(3): 625-644, 2020 03.
Article in English | MEDLINE | ID: mdl-31944520

ABSTRACT

The MYC protein is a transcription factor with oncogenic potential controlling fundamental cellular processes such as cell proliferation, metabolism, differentiation, and apoptosis. The MYC gene is a major cancer driver, and elevated MYC protein levels are a hallmark of most human cancers. We have previously shown that the brain acid-soluble protein 1 gene (BASP1) is specifically downregulated by the v-myc oncogene and that ectopic BASP1 expression inhibits v-myc-induced cell transformation. The 11-amino acid effector domain of the BASP1 protein interacts with the calcium sensor calmodulin (CaM) and is mainly responsible for this inhibitory function. We also reported recently that CaM interacts with all MYC variant proteins and that ectopic CaM increases the transactivation and transformation potential of the v-Myc protein. Here, we show that the presence of excess BASP1 or of a synthetic BASP1 effector domain peptide leads to displacement of v-Myc from CaM. The protein stability of v-Myc is decreased in cells co-expressing v-Myc and BASP1, which may account for the inhibition of v-Myc. Furthermore, suppression of v-Myc-triggered transcriptional activation and cell transformation is compensated by ectopic CaM, suggesting that BASP1-mediated withdrawal of CaM from v-Myc is a crucial event in the inhibition. In view of the tumor-suppressive role of BASP1 which was recently also reported for human cancer, small compounds or peptides based on the BASP1 effector domain could be used in drug development strategies aimed at tumors with high MYC expression.


Subject(s)
Calmodulin/metabolism , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , Gene Expression , Humans , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Binding , Protein Domains , Protein Stability , Proto-Oncogene Proteins c-myc/genetics , Quail , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
13.
Cell Calcium ; 83: 102058, 2019 11.
Article in English | MEDLINE | ID: mdl-31425929

ABSTRACT

Bone morphogenetic protein-2 (BMP-2) promotes the differentiation of non-osteogenic mesenchymal cells to osteogenic cells. In this study, we isolated human adipose-derived stem cells (hASCs) and investigated the effects of recombinant human BMP-2 (rhBMP-2) and extracellular Ca2+ concentration ([Ca2+]out) on the osteogenic differentiation of hASCs. rhBMP-2 promoted calcium deposition in hASCs and stimulated the mRNA expressions of six proteins known to be involved in the osteogenic differentiation of hASCs: Runx2, osterix, alkaline phosphatase, osteonectin, bone sialoprotein and osteocalcin. Elevation of [Ca2+]out enhanced the level of alkaline phosphatase enzyme, increased the mRNA expressions of Runx2 and osteocalcin and induced the expressions of BMP-2 mRNA and protein in hASCs. Elevation of [Ca2+]out transiently increased the intracellular Ca2+ concentration ([Ca2+]in) due to activation of the calcium-sensing receptor (CaSR). The Ca2+-induced expressions of BMP-2 mRNA and protein were inhibited by the calmodulin antagonist, W-7. Furthermore, elevation of [Ca2+]out decreased the cytoplasmic level of phosphorylated nuclear factor of activated T-cell-2 (NFAT-2) and increased the nuclear level of dephosphorylated NFAT2. Taken together, these results suggest that rhBMP-2 promotes the osteogenic differentiation of hASCs. Furthermore, an increase in [Ca2+]out enhances the expression of BMP-2 via activation of the CaSR, elevation of [Ca2+]in and stimulation of Ca2+/calmodulin-dependent NFAT-signaling pathways.


Subject(s)
Adipose Tissue/cytology , Bone Morphogenetic Protein 2/metabolism , Calcium/metabolism , Calmodulin/metabolism , Extracellular Space/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Messenger/genetics , Transforming Growth Factor beta/metabolism , Calcium Signaling , Calmodulin/antagonists & inhibitors , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis/genetics , Receptors, Calcium-Sensing/metabolism , Recombinant Proteins/metabolism , Sulfonamides/pharmacology
14.
Vet Microbiol ; 232: 156-161, 2019 May.
Article in English | MEDLINE | ID: mdl-30967327

ABSTRACT

Calcium (Ca2+) is a pivotal intracellular second messenger and calmodulin (CaM) acts as a multifunctional Ca2+-binding protein that regulates downstream Ca2+ dependent signaling. Together they play an important role in regulating various cellular functions, including gene expression, maturation of phagolysosome, apoptosis, and immune response. Intracellular Ca2+ has been shown to play a critical role in Toll-like receptor-mediated immune response to microbial agonists in the HD11 chicken macrophage cell line. The role of that the Ca2+/CaM pathway plays in the intracellular survival of Salmonella in chicken macrophages has not been reported. In this study, kinome peptide array analysis indicated that the Ca2+/CaM pathway was significantly activated when chicken macrophage HD11 cells were infected with S. Enteritidis or S. Heidelberg. Further study demonstrated that treating cells with a pharmaceutical CaM inhibitor W-7, which disrupts the formation of Ca2+/CaM, significantly inhibited macrophages to produce nitric oxide and weaken the control of intracellular Salmonella replication. These results strongly indicate that CaM plays an important role in the innate immune response of chicken macrophages and that the Ca2+/CaM mediated signaling pathway is critically involved in the host cell response to Salmonella infection.


Subject(s)
Calmodulin/antagonists & inhibitors , Macrophages/microbiology , Nitric Oxide/metabolism , Salmonella enteritidis/growth & development , Animals , Calcium/metabolism , Cell Line , Chickens , Enzyme Inhibitors/pharmacology , Immunity, Innate , Macrophages/drug effects , Microbial Viability/drug effects , Protein Array Analysis , Signal Transduction/drug effects , Sulfonamides/pharmacology
15.
Anal Biochem ; 572: 25-32, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30825429

ABSTRACT

The KRAS gene is highly mutated in human cancers and the focus of current Ras drug development efforts. Recently the interface between the C-terminus of K-Ras and calmodulin (CaM) was proposed as a target site to block K-Ras driven cancer cell stemness. We therefore aimed at developing a high-throughput amenable screening assay to identify novel CaM-inhibitors as potential K-Ras stemness-signaling disruptors. A modulated time-resolved Förster resonance energy transfer (mTR-FRET)-assay was developed and benchmarked against an identically designed fluorescence anisotropy (FA)-assay. In both assays, two CaM-binding peptides were labeled with Eu(III)-chelate or fluorescein and used as single-label reporter probes that were displaced from CaM upon competitor binding. Thus, peptidic and small molecule competitors with nanomolar to micromolar affinities to CaM could be detected, including a peptide that was derived from the C-terminus of K-Ras. In order to detect CaM-residue specific covalent inhibitors, a cell lysate-based Förster resonance energy transfer (FRET)-assay was furthermore established. This assay enabled us to measure the slow, residue-specific, covalent inhibition by ophiobolin A in the presence of other endogenous proteins. In conclusion, we have developed a panel of fluorescence-assays that allows identification of conventional and covalent CaM-inhibitors as potential disruptors of K-Ras driven cancer cell stemness.


Subject(s)
Calmodulin/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays/methods , Calmodulin/genetics , Calmodulin/metabolism , Enzyme Inhibitors/metabolism , Europium/chemistry , Fluorescein/chemistry , Humans , Inhibitory Concentration 50 , Kinetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sesterterpenes/chemistry , Sesterterpenes/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
16.
Protoplasma ; 256(4): 1165-1169, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30675652

ABSTRACT

Glutamate (Glu), a neurotransmitter in animal, is a novel signaling molecule in plants, which takes part in cellular metabolism, seed germination, plant growth, development, and long-distance information transfer. However, whether Glu can enhance the heat tolerance in maize seedlings and its relation to calcium signaling is still elusive. In this study, maize seedlings were pretreated with Glu and then exposed to heat stress. The results showed that Glu pretreatment enhanced the survival percentage of maize seedlings under heat tolerance, indicating that Glu could increase the heat tolerance of maize seedlings. The Glu-induced heat tolerance was weakened by exogenous calcium chloride, plasma membrane Ca2+ channel blocker (LaCl3), Ca2+ chelator (ethylene glycol-bis(b-aminoethylether)-N,N, N΄,N΄-tetraacetic acid), calmodulin antagonists (trifluoperazine and chlopromazine), and plant glutamate receptor-like antagonists (MgCl2 and 6,7-dinitroquinoxaline- 2,3-(1H,4H)- dione). These findings for the first time reported that Glu could increase the heat tolerance of maize seedlings by plant glutamate receptor-like channels-mediated calcium signaling.


Subject(s)
Glutamates/metabolism , Heat-Shock Response/physiology , Plant Proteins/metabolism , Seedlings/physiology , Zea mays/physiology , Calcium Signaling , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Edetic Acid/pharmacology , Glutamates/pharmacology , Heat-Shock Response/drug effects , Magnesium Chloride , Plant Proteins/antagonists & inhibitors , Receptors, Glutamate , Seedlings/drug effects , Seedlings/metabolism , Trifluoperazine/pharmacology , Zea mays/drug effects , Zea mays/metabolism
17.
Arthritis Rheumatol ; 71(1): 63-72, 2019 01.
Article in English | MEDLINE | ID: mdl-30003689

ABSTRACT

OBJECTIVE: To investigate mechanisms underlying the capability of ADAM15 to transform FasL-mediated death-inducing signals into prosurvival activation of Src and focal adhesion kinase (FAK) in rheumatoid arthritis synovial fibroblasts (RASFs). METHODS: Caspase 3/7 activity and apoptosis rate were determined in RASFs and ADAM15-transfected T/C28a4 cells upon Fas/CD95 triggering using enzyme assays and annexin V staining. Phosphorylated Src and FAK were analyzed by immunoblotting. Interactions of ADAM15 and CD95 with calmodulin (CaM), Src, or FAK were analyzed by pull-downs using CaM-Sepharose and coimmunoprecipitations with specific antibodies. Protein binding assays were performed using recombinant CaM and ADAM15. Immunofluorescence was performed to investigate subcellular colocalization of ADAM15, Fas/CD95, and CaM. RESULTS: The antiapoptotic effect of ADAM15 in FasL-stimulated cells was demonstrated either by increased apoptosis of cells transfected with an ADAM15 construct lacking the cytoplasmic domain compared to cells transfected with full-length ADAM15 or by reduced apoptosis resistance of RASFs upon RNA interference silencing of ADAM15. Fas ligation triggered a Ca2+  release-activated Ca2+ /calcium release-activated calcium channel protein 1 (CRAC/Orai1) channel-dependent CaM recruitment to Fas/CD95 and ADAM15 in the cell membrane. Simultaneously, Src associated with CaM was shown to become engaged in the ADAM15 complex also containing cytoplasmic-bound FAK. Accordingly, Fas ligation in RASFs led to ADAM15-dependent phosphorylation of Src and FAK, which was associated with increased survival. Pharmacologic interference with either the CaM inhibitor trifluoperazine or the CRAC/Orai inhibitor BTP-2 simultaneously applied with FasL synergistically enhanced Fas-mediated apoptosis in RASFs. CONCLUSION: ADAM15 provides a scaffold for formation of CaM-dependent prosurvival signaling complexes upon CRAC/Orai coactivation by FasL-induced death signals and a potential therapeutic target to break apoptosis resistance in RASFs.


Subject(s)
ADAM Proteins/genetics , Apoptosis/genetics , Arthritis, Rheumatoid/metabolism , Calmodulin/metabolism , Chondrocytes/metabolism , Fas Ligand Protein/metabolism , Fibroblasts/metabolism , Membrane Proteins/genetics , ADAM Proteins/metabolism , Anilides/pharmacology , Apoptosis/drug effects , Calmodulin/antagonists & inhibitors , Cell Line , Focal Adhesion Kinase 1/metabolism , Humans , Membrane Proteins/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/metabolism , Phosphorylation , RNA Interference , Synovial Membrane/cytology , Thiadiazoles/pharmacology , Trifluoperazine/pharmacology , fas Receptor/metabolism , src-Family Kinases/metabolism
18.
J Reprod Dev ; 64(5): 445-449, 2018 Oct 12.
Article in English | MEDLINE | ID: mdl-29887540

ABSTRACT

We aimed to test whether the calmodulin (CaM) inhibitors, calmidazolium (CZ) and N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide (W-7), can be used to assess lipid disorder by flow cytometry using Merocyanine 540 (M540). Boar spermatozoa were incubated in non-capacitating conditions for 10 min at room temperature with 1 µM CZ, 200 µM W-7, or 1 mM 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP). Then, sperm were 1) directly evaluated, 2) centrifuged and washed prior to evaluation, or 3) diluted with PBS prior to evaluation. Direct evaluation showed an increase in high M540 fluorescence in spermatozoa treated with the inhibitors (4.7 ± 1.8 [control] vs. 70.4 ± 4.0 [CZ] and 71.4 ± 4.2 [W-7], mean % ± SD, P < 0.001); washing decreased the percentage of sperm showing high M540 fluorescence for W-7 (4.8 ± 2.2, mean % ± SD) but not for CZ (69.4 ± 3.9, mean % ± SD, P < 0.001), and dilution showed an increase in high M540 fluorescence for both CZ and W-7; 8-Br-cAMP did not induce a rise in sperm showing high M540 fluorescence. Therefore, special care must be taken when M540 is used in spermatozoa with CaM inhibitors.


Subject(s)
Calmodulin/antagonists & inhibitors , Pyrimidinones/chemistry , Sperm Capacitation , Spermatozoa/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/chemistry , Animals , Cell Membrane/drug effects , Flow Cytometry , Lipids/chemistry , Male , Swine
19.
Biomol Concepts ; 9(1): 53-63, 2018 May 19.
Article in English | MEDLINE | ID: mdl-29779013

ABSTRACT

Treatment with analogues of the SERCA-inhibitor Thapsigargin is a promising new approach for a wide variety of cancer entities. However, our previous studies on various tumor cells suggested resistance of SEC62 over-expressing tumors to this treatment. Therefore, we proposed the novel concept that e.g. lung-, prostate-, and thyroid-cancer patients should be tested for SEC62 over-expression, and developed a novel therapeutic strategy for a combinatorial treatment of SEC62 over-expressing tumors. The latter was based on the observations that treatment of SEC62 over-expressing tumor cells with SEC62-targeting siRNAs showed less resistance to Thapsigargin as well as a reduction in migratory potential and that the siRNA effects can be mimicked by the Calmodulin antagonist Trifluoperazine. Therefore, the combinatorial treatment of SEC62 over-expressing tumors was proposed to involve Thapsigargin and Trifluoperazine. Here, we addressed the impact of Thapsigargin and Trifluoperazine in separate and combined treatments of heterotopic tumors, induced by inoculation of human hypopharyngeal squamous cell carcinoma (FaDu)-cells into the mouse flank. Seeding of the tumor cells and/or their growth rate were significantly reduced by all three treatments, suggesting Trifluoperazine is a small molecule to be considered for future therapeutic strategies for patients, suffering from Sec62-overproducing tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Enzyme Inhibitors/therapeutic use , Head and Neck Neoplasms/drug therapy , Hypopharyngeal Neoplasms/drug therapy , Membrane Transport Proteins/metabolism , Thapsigargin/therapeutic use , Trifluoperazine/therapeutic use , Animals , Calmodulin/antagonists & inhibitors , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation , Enzyme Inhibitors/blood , Head and Neck Neoplasms/genetics , Humans , Hypopharyngeal Neoplasms/genetics , Mice , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck , Thapsigargin/blood , Trifluoperazine/blood
20.
J Cell Biochem ; 119(7): 6216-6230, 2018 07.
Article in English | MEDLINE | ID: mdl-29663486

ABSTRACT

Patients with triple negative breast cancer (TNBC) have no successful "targeted" treatment modality, which represents a priority for novel therapy strategies. Upregulated death receptor 5 (DR5) expression levels in breast cancer cells compared to normal cells enable TRA-8, a DR5 specific agonistic antibody, to specifically target malignant cells for apoptosis without inducing normal hepatocyte apoptosis. Drug resistance is a common obstacle in TRAIL-based therapy for TNBC. Calmodulin (CaM) is overexpressed in breast cancer. In this study, we characterized the novel function of CaM antagonist in enhancing TRA-8 induced cytotoxicity in TRA-8 resistant TNBC cells and its underlying molecular mechanisms. Results demonstrated that CaM antagonist(s) enhanced TRA-8 induced cytotoxicity in a concentration and time-dependent manner for TRA-8 resistant TNBC cells. CaM directly bound to DR5 in a Ca2+ dependent manner, and CaM siRNA promoted DR5 recruitment of FADD and caspase-8 for DISC formation and TRA-8 activated caspase cleavage for apoptosis in TRA-8 resistant TNBC cells. CaM antagonist, trifluoperazine, enhanced TRA-8 activated DR5 oligomerization, DR5-mediated DISC formation, and TRA-8 activated caspase cleavage for apoptosis, and decreased anti-apoptotic pERK, pAKT, XIAP, and cIAP-1 expression in TRA-8 resistant TNBC cells. These results suggest that CaM could be a key regulator to mediate DR5-mediated apoptotic signaling, and suggests a potential strategy for using CaM antagonists to overcome drug resistance of TRAIL-based therapy for TRA-8 resistant TNBC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Calmodulin/antagonists & inhibitors , Drug Resistance, Neoplasm , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Triple Negative Breast Neoplasms/pathology , Female , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...