Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(4): 784-799.e19, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32413299

ABSTRACT

Swelling of the brain or spinal cord (CNS edema) affects millions of people every year. All potential pharmacological interventions have failed in clinical trials, meaning that symptom management is the only treatment option. The water channel protein aquaporin-4 (AQP4) is expressed in astrocytes and mediates water flux across the blood-brain and blood-spinal cord barriers. Here we show that AQP4 cell-surface abundance increases in response to hypoxia-induced cell swelling in a calmodulin-dependent manner. Calmodulin directly binds the AQP4 carboxyl terminus, causing a specific conformational change and driving AQP4 cell-surface localization. Inhibition of calmodulin in a rat spinal cord injury model with the licensed drug trifluoperazine inhibited AQP4 localization to the blood-spinal cord barrier, ablated CNS edema, and led to accelerated functional recovery compared with untreated animals. We propose that targeting the mechanism of calmodulin-mediated cell-surface localization of AQP4 is a viable strategy for development of CNS edema therapies.


Subject(s)
Aquaporin 4/metabolism , Edema/metabolism , Edema/therapy , Animals , Aquaporin 4/physiology , Astrocytes/metabolism , Brain/metabolism , Brain Edema/metabolism , Calmodulin/metabolism , Central Nervous System/metabolism , Edema/physiopathology , Male , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Trifluoperazine/pharmacology
2.
Toxicol Appl Pharmacol ; 492: 117080, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39216834

ABSTRACT

The survival rates for patients with osteosarcoma (OS) have stagnated over the past few decades. It is essential to find new therapies and drugs. A licensed antipsychotic medication called trifluoperazine (TFP) significantly reduces the growth of several cancers. However, the exact molecular pathways of TFP in OS remain to be discovered. Our research revealed that TFP greatly reduced OS cell migration and growth and caused the arrest of G0/G1 cell cycle. Combined with RNA-Seq data and further research, we confirmed that TFP promoted reactive oxygen species (ROS) production by elevating thioredoxin binding protein (TXNIP) expression to induce mitochondria-dependent apoptosis. Interestingly, we first demonstrated that AKT was an upstream regulatory target of TXNIP in OS cells. Dephosphorylation of AKT led to an increase in TXNIP expression, further elucidating the anticancer mechanism of TFP. In vivo, TFP inhibited subcutaneous OS cell proliferation and induced OS cell apoptosis without noticeable side effects. In conclusion, our findings imply that TFP is a potential treatment for OS.

3.
Int J Med Sci ; 20(6): 797-809, 2023.
Article in English | MEDLINE | ID: mdl-37213674

ABSTRACT

Currently, no specific and standard treatment for traumatic brain injury (TBI) has been developed. Therefore, studies on new therapeutic drugs for TBI treatment are urgently needed. Trifluoperazine (TFP) is a therapeutic agent for the treatment of psychiatric disorders that reduces edema of the central nervous system. However, the specific working mechanism of TFP is not fully understood in TBI. In this study, the immunofluorescence co-localization analysis revealed that the area and intensity covered by Aquaporin4 (AQP4) on the surface of brain cells (astrocyte endfeet) increased significantly after TBI. In contrast, TFP treatment reversed these phenomena. This finding showed that TFP inhibited AQP4 accumulation on the surface of brain cells (astrocyte endfeet). The tunel fluorescence intensity and fluorescence area were lower in the TBI+TFP group compared to the TBI group. Additionally, the brain edema, brain defect area, and modified neurological severity score (mNSS) were lower in the TBI+TFP. The RNA-seq was performed on the cortical tissues of rats in the Sham, TBI, and TBI+TFP groups. A total of 3774 genes differently expressed between the TBI and the Sham group were identified. Of these, 2940 genes were up-regulated and 834 genes were down-regulated. A total of 1845 differently expressed genes between the TBI+TFP and TBI group were also identified, in which 621 genes were up-regulated and 1224 genes were down-regulated. Analysis of the common differential genes in the three groups showed that TFP could reverse the expression of apoptosis and inflammation genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that the differentially expressed genes (DEGs) were highly enriched in the signaling pathways regulating inflammation. In conclusion, TFP alleviates brain edema after TBI by preventing the accumulation of AQP4 on the surface of brain cells. Generally, TFP alleviates apoptosis and inflammatory response induced by TBI, and promotes the recovery of nerve function in rats after TBI. Thus, TFP is a potential therapeutic agent for TBI treatment.


Subject(s)
Brain Edema , Brain Injuries, Traumatic , Animals , Rats , Apoptosis/genetics , Aquaporin 4/antagonists & inhibitors , Aquaporin 4/genetics , Aquaporin 4/metabolism , Brain , Brain Edema/etiology , Brain Edema/genetics , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Trifluoperazine/pharmacology , Trifluoperazine/therapeutic use , Trifluoperazine/metabolism
4.
J Biol Chem ; 297(2): 101012, 2021 08.
Article in English | MEDLINE | ID: mdl-34324830

ABSTRACT

Repair of damaged plasma membrane in eukaryotic cells is largely dependent on the binding of annexin repair proteins to phospholipids. Changing the biophysical properties of the plasma membrane may provide means to compromise annexin-mediated repair and sensitize cells to injury. Since, cancer cells experience heightened membrane stress and are more dependent on efficient plasma membrane repair, inhibiting repair may provide approaches to sensitize cancer cells to plasma membrane damage and cell death. Here, we show that derivatives of phenothiazines, which have widespread use in the fields of psychiatry and allergy treatment, strongly sensitize cancer cells to mechanical-, chemical-, and heat-induced injury by inhibiting annexin-mediated plasma membrane repair. Using a combination of cell biology, biophysics, and computer simulations, we show that trifluoperazine acts by thinning the membrane bilayer, making it more fragile and prone to ruptures. Secondly, it decreases annexin binding by compromising the lateral diffusion of phosphatidylserine, inhibiting the ability of annexins to curve and shape membranes, which is essential for their function in plasma membrane repair. Our results reveal a novel avenue to target cancer cells by compromising plasma membrane repair in combination with noninvasive approaches that induce membrane injuries.


Subject(s)
Annexins/antagonists & inhibitors , Cell Membrane/drug effects , Molecular Dynamics Simulation , Neoplasms/drug therapy , Phenothiazines/pharmacology , Annexins/metabolism , Antipsychotic Agents/pharmacology , Calcium/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylserines/metabolism , Phospholipids/metabolism
5.
Biochem Biophys Res Commun ; 610: 182-187, 2022 06 25.
Article in English | MEDLINE | ID: mdl-35468422

ABSTRACT

Rv1211 is a conserved hypothetical protein in Mycobacterium tuberculosis and is required for the growth and pathogenesis of the bacteria. The protein has been suggested as a calmodulin-like calcium-binding protein with an EF-hand motif and as a target of trifluoperazine, a calmodulin antagonist in eukaryotes that inhibits mycobacterial growth. Here, we expressed the recombinant protein of Rv1211 and performed structural and biochemical studies of Rv1211 and its interaction with Ca2+ or trifluoperazine. Surprisingly, Rv1211 exhibited an elution property typical of a natively unfolded protein. Subsequent circular dichroism experiments with temperature elevation and trifluoroethanol treatment showed that Rv1211 has unfolded structure. Additional NMR experiment confirmed the unfolded state of the protein and further showed that it does not bind to Ca2+. Still, Rv1211 did bind to trifluoperazine, as evidenced by the two-dimensional NMR spectra of 15N-labeled Rv1211. However, there were no peak shifts upon binding, showing that Rv1211 retained its unfolded state even after the trifluoperazine binding. The residues involved in the binding were clustered in the C-terminal region, as identified by the sequence assignment. Isothermal titration calorimetry showed that the Kd of trifluoperazine-Rv1211 binding is 41 µM and that the stoichiometry is 1 : 2 (Rv1211: trifluoperazine). Our results argue against the suggestion of Rv1211 as a Ca2+-binding calmodulin-like protein, and show that Rv1211 is a natively unfolded protein that binds to trifluoperazine. In addition, our results suggest the evidence of the "Fuzziness" in the Rv1211-trifluoperazine interaction that differs from the conventional binding-induced folding of natively unfolded proteins.


Subject(s)
Intrinsically Disordered Proteins , Mycobacterium tuberculosis , Calcium/metabolism , Calmodulin/metabolism , EF Hand Motifs , Intrinsically Disordered Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Trifluoperazine/chemistry , Trifluoperazine/pharmacology
6.
Biomed Chromatogr ; 36(12): e5499, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36064842

ABSTRACT

A sensitive, selective and rapid bioanalytical method using liquid chromatography-tandem mass spectrometry has been developed for the quantification of trifluoperazine in human plasma. Trifluoperazine-D8 was used as the internal standard and the extraction from human plasma was performed by liquid-liquid extraction technique using tertiary butyl methyl ether as the solvent. Chromatographic separation was carried out on Zodiac C18 column (50 × 4.6 mm, 3 µm) employing a mixture of acetonitrile, methanol and 5 mm ammonium bicarbonate buffer in water (85:10:5, v/v/v) at a flow rate of 0.55 ml/min. The linearity was established within the concentration range of 5-1,250 pg/ml with r2 > 0.99. The results of all of the validation parameters as per the US Food and Drug Administration guidelines were within the acceptance limits. The pharmacokinetics of trifluoperazine after oral administration of a syrup of 1 mg dose under fasting conditions was determined by successful application of the present method.


Subject(s)
Tandem Mass Spectrometry , Trifluoperazine , Humans , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Kinetics , Reproducibility of Results , Chromatography, High Pressure Liquid/methods
7.
Tohoku J Exp Med ; 257(4): 315-326, 2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35644544

ABSTRACT

Multiple myeloma (MM) is a common hematological malignancy. Bortezomib (BTZ) is a traditional medicine for MM treatment, but there are limitations for current treatment methods. Trifluoperazine (TFP) is a clinical drug for acute and chronic psychosis therapy. Lately, researchers have found that TFP can suppress tumor growth in many cancers. We attempted to study the effects of BTZ and TFP on MM in vivo and in vitro. We concentrated on the individual and combined impact of BTZ and TFP on the proliferation and apoptosis of MM cells via Cell Counting kit-8 assay, EdU assay, western blot, and flow cytometry. We found that combination therapy has a strong synergistic impact on MM cells. Combination therapy could induce cell arrest during G2/M phase and induce apoptosis in MM cells. Meanwhile, BTZ combined with TFP could play a better role in the anti-MM effect in vivo through MM.1s xenograft tumor models. Furthermore, we explored the mechanism of TFP-induced apoptosis in MM, and we noticed that TFP might induce MM apoptosis by inhibiting p-P38 MAPK/NUPR1. In summary, our findings suggest that TFP could synergistically enhance the BTZ-induced anti-cancer effect in multiple myeloma, which might be a promising therapeutic strategy for MM treatment.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Tumor , Cell Proliferation , Humans , Multiple Myeloma/drug therapy , Neoplasm Proteins/metabolism , Trifluoperazine/pharmacology , Trifluoperazine/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Biochem Biophys Res Commun ; 570: 148-153, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34284140

ABSTRACT

P-glycoprotein, member of the B-subfamily of the ATP-binding cassette (ABC) superfamily (e.g., ABCB1), has been demonstrated to confer resistance to clinically relevant anticancer drugs. Paradoxically, ABCB1-expressing multidrug resistant (MDR) cells are hypersensitivity or collateral sensitivity to non-toxic drugs. In this report, we demonstrate the capacity of trifluoperazine (TFP), a calmodulin inhibitor, to confer a collateral sensitivity onto ABCB1-overexpressing MDR cells. We show TFP-induced collateral sensitivity to be linked to ABCB1 expression and ATPase activity, as such phenotype is abolished in ABCB1-knockout MDR cells (CHORC5ΔABCB1 clones A1-A3) or with inhibitors of ABCB1 ATPase. TFP-induced collateral sensitivity is mediated by apoptotic cell death, due to enhanced oxidative stress. The findings in this study show for first time the use TFP as a collateral sensitivity drug, at clinically relevant concentrations. Moreover, given the use of trifluoperazine in the treatment for symptoms of schizophrenia and the role of ABCB1 transporter in tissue blood barriers and other physiologic functions, the finding in this study may have implications beyond cancer chemotherapy.


Subject(s)
Drug Resistance, Multiple , Drug Resistance, Neoplasm , Phenothiazines/pharmacology , Trifluoperazine/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Apoptosis/drug effects , Cell Line , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Macrolides/pharmacology , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism
9.
Pharmacol Res ; 163: 105295, 2021 01.
Article in English | MEDLINE | ID: mdl-33176207

ABSTRACT

Targeted therapies and immunotherapy have brought substantial benefits to patients with melanoma. However, brain metastases remain the biggest threat to the survival and quality of life of melanoma patients. One of the major challenges to an effective therapy is the inability of drugs to penetrate the blood-brain barrier (BBB). Anti-schizophrenic drugs can cross the BBB, and many of them have demonstrated anti-cancer effects. Repurposing existing drugs for new clinical indications is an alluring strategy for anticancer drug discovery. Herein, we applied this strategy and screened a small collection of existing anti-schizophrenic drugs to use as anti-melanoma agents. Among them, trifluoperazine dihydrochloride (TFP) exhibited promising potencies for suppressing the growth and metastasis of melanoma, both in vitro and in vivo. TFP obviously suppressed the viability of melanoma cells within the micromolar range and inhibited the growth of melanoma in the subcutaneous mice models. Notably, intraperitoneal (i.p.) administration of TFP (40 mg/kg/day) obviously inhibited the growth of intra-carotid-injection established melanoma brain metastasis and extended the survival of brain metastasis-bearing mice. Moreover, TFP significantly suppressed lung metastasis and bone metastasis of melanoma in preclinical metastasis models. Mechanistically, TFP caused G0/G1 cell cycle arrest and mitochondrial-dependent intrinsic apoptosis of melanoma cells. In addition, TFP treatment increased the expression of microtubule associated protein 1 light chain 3 beta-II (LC3B-II) and p62 in vitro, suggesting an inhibition of autophagic flux. TFP decreased LysoTracker Red uptake after treatment, indicating impaired acidification of lysosomes. Moreover, the colocalization of LC3 with lysosomal-associated membrane protein 1 (LAMP1), a lysosome marker, was also suppressed after TFP treatment, suggesting that TFP might block the fusion of autophagosomes with lysosomes, which led to autophagosome accumulation. Taken together, our data highlight the potential of repurposing TFP as a new adjuvant drug for treating melanoma patients with brain, lung, and bone metastases.


Subject(s)
Antineoplastic Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Bone Neoplasms/drug therapy , Brain Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Trifluoperazine/therapeutic use , Animals , Autophagy/drug effects , Bone Neoplasms/secondary , Brain Neoplasms/secondary , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Repositioning , Female , Humans , Lung Neoplasms/secondary , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Skin Neoplasms/pathology
10.
J Cell Biochem ; 120(9): 15756-15765, 2019 09.
Article in English | MEDLINE | ID: mdl-31081173

ABSTRACT

The development of cancer in patients with schizophrenia is affected by genetic and environmental factors and antipsychotic medication. Several studies found that schizophrenia was associated with decreased risk of some cancers, and the neuroleptic medication might help to reduce the risk of colorectal cancer (CRC). Phenothiazine drugs including trifluoperazine (TFP) are widely used antipsychotic drugs and showed some antitumor effects, we here investigated the potential application of TFP in the treatment of colon cancer. A series doses of TFP were treated to the colon cancer cell line HCT116 and the inhibitory concentration (IC50 ) of TFP for HCT116 was determined by cell counting kit-8. The results indicated that the treatment of TFP impaired the cell vitality of HCT116 in a dose- and time-dependent manner. Meanwhile, the Edu assay demonstrated that the proliferation was also inhibited by TFP, which was accompanied with the induction of apoptosis and autophagy. The expression of CCNE1, CDK4, and antiapoptosis factor BCL-2 was downregulated but the proapoptosis factor BAX was upregulated. The autophagy inhibitor chloroquine could significantly reverse the TFP-induced apoptosis. Moreover, the ability of migration and invasion of HCT116 was found to be suppressed by TFP, which was associated with the inhibition of epithelial-mesenchymal transition (EMT). The function of TFP in vivo was further confirmed. The results showed that the administration of TFP remarkably abrogated the tumor growth with decreased tumor volume and proliferation index Ki-67 level in tumor tissues. The EMT phenotype was also confirmed to be inhibited by TFP in vivo, suggesting the promising antitumor effects of TFP in CRC.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Trifluoperazine/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Mice, Nude , Time Factors , Trifluoperazine/pharmacology , Xenograft Model Antitumor Assays
11.
Int J Mol Sci ; 20(13)2019 Jun 30.
Article in English | MEDLINE | ID: mdl-31262032

ABSTRACT

Cisplatin-based chemotherapy is the primary treatment for metastatic bladder urothelial carcinoma (UC). Most patients inevitably encounter drug resistance and resultant disease relapse. Reduced apoptosis plays a critical role in chemoresistance. Trifluoperazine (TFP), an antipsychotic agent, has demonstrated antitumor effects on various cancers. This study investigated the efficacy of TFP in inhibiting cisplatin-resistant bladder UC and explored the underlying mechanism. Our results revealed that cisplatin-resistant UC cells (T24/R) upregulated the antiapoptotic factor, B-cell lymphoma-extra large (Bcl-xL). Knockdown of Bcl-xL by siRNA resensitized cisplatin-resistant cells to the cisplatin cytotoxic effect. TFP (10-45 µM) alone elicited dose-dependent cytotoxicity, apoptosis, and G0/G1 arrest on T24/R cells. Co-treatment of TFP potentiated cisplatin-induced cytotoxicity in T24/R cells. The phenomenon that TFP alleviated cisplatin resistance to T24/R was accompanied with concurrent suppression of Bcl-xL. In vivo models confirmed that TFP alone effectively suppressed the T24/R xenograft in nude mice. TFP co-treatment enhanced the antitumor effect of cisplatin on the T24/R xenograft. Our results demonstrated that TFP effectively inhibited cisplatin-resistant UCs and circumvented cisplatin resistance with concurrent Bcl-xL downregulation. These findings provide a promising insight to develop a therapeutic strategy for chemoresistant UCs.


Subject(s)
Antipsychotic Agents/pharmacology , Carcinoma/drug therapy , Drug Resistance, Neoplasm , Trifluoperazine/pharmacology , Urinary Bladder Neoplasms/drug therapy , bcl-X Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Apoptosis , Carcinoma/metabolism , Cell Line , Cisplatin/pharmacology , Cisplatin/therapeutic use , Down-Regulation , Humans , Mice , Trifluoperazine/therapeutic use , Urinary Bladder Neoplasms/metabolism , Urothelium/drug effects , Urothelium/metabolism , Urothelium/pathology , bcl-X Protein/genetics
12.
Biochem Biophys Res Commun ; 505(1): 208-214, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30243724

ABSTRACT

Bacterial fatty acid synthesis (FAS) has been extensively studied as a potential target of antimicrobials. In FAS, FabD mediates transacylation of the malonyl group from malonyl-CoA to acyl-carrier protein (ACP). The mounting threat of nosocomial infection by multidrug-resistant Acinetobacter baumannii warrants a deeper understanding of its essential cellular mechanisms, which could lead to effective control of this highly competent pathogen. The molecular mechanisms involved in A. baumannii FAS are poorly understood, and recent research has suggested that Pseudomonas aeruginosa, a closely related nosocomial pathogen of A. baumannii, utilizes FAS to produce virulence factors. In this study, we solved the crystal structure of A. baumannii FabD (AbFabD) to provide a platform for the development of new antibacterial agents. Analysis of the structure of AbFabD confirmed the presence of highly conserved active site residues among bacterial homologs. Binding constants between AbFabD variants and A. baumannii ACP (AbACP) revealed critical conserved residues Lys195 and Lys200 involved in AbACP binding. Computational docking of a potential inhibitor, trifluoperazine, revealed a unique inhibitor-binding pocket near the substrate-binding site. The structural study presented herein will be useful for the structure-based design of potent AbFabD inhibitors.


Subject(s)
Acinetobacter baumannii/genetics , Acyl-Carrier Protein S-Malonyltransferase/genetics , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Fatty Acid Synthase, Type II/genetics , Acinetobacter baumannii/enzymology , Acyl-Carrier Protein S-Malonyltransferase/chemistry , Acyl-Carrier Protein S-Malonyltransferase/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites/genetics , Crystallography, X-Ray , Fatty Acid Synthase, Type II/chemistry , Fatty Acid Synthase, Type II/metabolism , Models, Molecular , Mutation , Protein Domains , Sequence Homology, Amino Acid
13.
Biochem Biophys Res Commun ; 499(2): 136-142, 2018 05 05.
Article in English | MEDLINE | ID: mdl-29580626

ABSTRACT

Glioblastoma (GBM) is regarded as the most common malignant brain tumor but treatment options are limited. Thus, there is an unmet clinical need for compounds and corresponding targets that could inhibit GBM growth. We screened a library of 80 dopaminergic ligands with the aim of identifying compounds capable of inhibiting GBM cell line proliferation and survival. Out of 45 active compounds, 8 were further validated. We found that the dopamine receptor D2 antagonist trifluoperazine 2HCl inhibits growth and proliferation of GBM cells in a dose dependent manner. Trifluoperazine's inhibition of GBM cells is cell line dependent and correlates with variations in dopamine receptor expression profile. We conclude that components of the dopamine receptor signaling pathways are potential targets for pharmacological interventions of GBM growth.


Subject(s)
Drug Evaluation, Preclinical , Glioblastoma/pathology , Trifluoperazine/pharmacology , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Dopamine/metabolism , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Humans , Ligands , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Signal Transduction/drug effects , Trifluoperazine/chemistry
14.
Traffic ; 16(6): 635-54, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25693808

ABSTRACT

Chlorpromazine is a phenothiazine-derived antipsychotic drug (APD) that inhibits clathrin-mediated endocytosis (CME) in cells by an unknown mechanism. We examined whether its action and that of other APDs might be mediated by the GTPase activity of dynamin. Eight of eight phenothiazine-derived APDs inhibited dynamin I (dynI) in the 2-12 µm range, the most potent being trifluoperazine (IC50 2.6 ± 0.7 µm). They also inhibited dynamin II (dynII) at similar concentrations. Typical and atypical APDs not based on the phenothiazine scaffold were 8- to 10-fold less potent (haloperidol and clozapine) or were inactive (droperidol, olanzapine and risperidone). Kinetic analysis showed that phenothiazine-derived APDs were lipid competitive, while haloperidol was uncompetitive with lipid. Accordingly, phenothiazine-derived APDs inhibited dynI GTPase activity stimulated by lipids but not by various SH3 domains. All dynamin-active APDs also inhibited transferrin (Tfn) CME in cells at related potencies. Structure-activity relationships (SAR) revealed dynamin inhibition to be conferred by a substituent group containing a terminal tertiary amino group at the N2 position. Chlorpromazine was previously proposed to target AP-2 recruitment in the formation of clathrin-coated vesicles (CCV). However, neither chlorpromazine nor thioridazine affected AP-2 interaction with amphiphysin or clathrin. Super-resolution microscopy revealed that chlorpromazine blocks neither clathrin recruitment by AP-2, nor AP-2 recruitment, showing that CME inhibition occurs downstream of CCV formation. Overall, potent dynamin inhibition is a shared characteristic of phenothiazine-derived APDs, but not other typical or atypical APDs, and the data indicate that dynamin is their likely in-cell target in endocytosis.


Subject(s)
Antipsychotic Agents/pharmacology , Clathrin/metabolism , Dynamins/metabolism , Endocytosis/drug effects , Phenothiazines/pharmacology , Cell Line, Tumor , Clathrin-Coated Vesicles/metabolism , Humans , Transferrin/metabolism
15.
Biochem Biophys Res Commun ; 494(3-4): 477-483, 2017 12 16.
Article in English | MEDLINE | ID: mdl-29066348

ABSTRACT

Glioblastoma (GBM) is regarded as the most common malignant brain tumor but treatment options are limited. Thus, there is an unmet clinical need for compounds and corresponding targets that could inhibit GBM growth. We screened a library of 80 dopaminergic ligands with the aim of identifying compounds capable of inhibiting GBM cell line proliferation and survival. Out of 45 active compounds, 8 were further validated. We found that the dopamine receptor D2 antagonist trifluoperazine 2HCl inhibits growth and proliferation of GBM cells in a dose dependent manner. Trifluoperazine's inhibition of GBM cells is cell line dependent and correlates with variations in dopamine receptor expression profile. We conclude that components of the dopamine receptor signaling pathways are potential targets for pharmacological interventions of GBM growth.


Subject(s)
Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor/methods , Glioblastoma/drug therapy , Glioblastoma/pathology , Trifluoperazine/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Dopamine Antagonists/administration & dosage , Dose-Response Relationship, Drug , Drug Discovery/methods , Glioblastoma/metabolism , Humans , Receptors, Dopamine/metabolism
16.
Biochem Biophys Res Commun ; 479(3): 584-589, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27666479

ABSTRACT

Trifluoperazine is a phenothiazine derivative which is mainly used in the management of schizophrenia and also acts as a calmodulin inhibitor. We used the whole-cell patch-clamp technique to study the effects of trifluoperazine on human Nav1.5 (hNav1.5) currents expressed in HEK293 cells. The 50% inhibitory concentration of trifluoperazine was 15.5 ± 0.3 µM and the Hill coefficient was 2.7 ± 0.1. The effects of trifluoperazine on hNav1.5 were completely and repeatedly reversible after washout. Trifluoperazine caused depolarizing shifts in the activation and hyperpolarizing shifts in the steady-state inactivation of hNav1.5. Trifluoperazine also showed strong use-dependent inhibition of hNav1.5. The blockade of hNav1.5 currents by trifluoperazine was not affected by the whole cell dialysis of the calmodulin inhibitory peptide. Our results indicated that trifluoperazine blocks hNav1.5 current in concentration-, state- and use-dependent manners rather than via calmodulin inhibition.


Subject(s)
NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium Channel Blockers/chemistry , Trifluoperazine/chemistry , Antipsychotic Agents/chemistry , Calmodulin/chemistry , HEK293 Cells , Humans , Inhibitory Concentration 50 , Membrane Potentials/drug effects , Patch-Clamp Techniques , Peptides/chemistry , Renal Dialysis
17.
Biochim Biophys Acta ; 1843(2): 398-435, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24188867

ABSTRACT

Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.


Subject(s)
Apoptosis , Autophagy , Calmodulin/metabolism , Neoplasms/pathology , Animals , Cell Proliferation , Humans , Models, Biological
18.
Purinergic Signal ; 11(4): 481-90, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26341077

ABSTRACT

P2X7 receptor (P2X7) activity may link inflammation to depressive disorders. Genetic variants of human P2X7 have been linked with major depression and bipolar disorders, and the P2X7 knockout mouse has been shown to exhibit anti-depressive-like behaviour. P2X7 is an ATP-gated ion channel and is a major regulator of the pro-inflammatory cytokine interleukin 1ß (IL-1ß) secretion from monocytes and microglia. We hypothesised that antidepressants may elicit their mood enhancing effects in part via modulating P2X7 activity and reducing inflammatory responses. In this study, we determined whether common psychoactive drugs could affect recombinant and native human P2X7 responses in vitro. Common antidepressants demonstrated opposing effects on human P2X7-mediated responses; paroxetine inhibited while fluoxetine and clomipramine mildly potentiated ATP-induced dye uptake in HEK-293 cells stably expressing recombinant human P2X7. Paroxetine inhibited dye uptake mediated by human P2X7 in a concentration-dependent manner with an IC(50) of 24 µM and significantly reduces ATP-induced inward currents. We confirmed that trifluoperazine hydrochloride suppressed human P2X7 responses (IC(50) of 6.4 µM). Both paroxetine and trifluoperazine did not inhibit rodent P2X7 responses, and mutation of a known residue (F 95L) did not alter the effect of either drug, suggesting neither drug binds at this site. Finally, we demonstrate that P2X7-induced IL-1ß secretion from lipopolysaccharide (LPS)-primed human CD14(+) monocytes was suppressed with trifluoperazine and paroxetine.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Paroxetine/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Animals , Antidepressive Agents, Tricyclic/pharmacology , Antipsychotic Agents/pharmacology , Clomipramine/pharmacology , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , Humans , Interleukin-1beta/metabolism , Lipopolysaccharide Receptors/metabolism , Mice , Monocytes/drug effects , Monocytes/metabolism , Purinergic P2X Receptor Agonists/pharmacology , Rats , Recombinant Proteins/drug effects , Trifluoperazine/pharmacology
19.
Gastroenterology ; 145(6): 1347-57, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973919

ABSTRACT

BACKGROUND & AIMS: Intestinal epithelial cells aid in mucosal defense by providing a physical barrier against entry of pathogenic bacteria and secreting antimicrobial peptides (AMPs). Autophagy is an important component of immune homeostasis. However, little is known about its role in specific cell types during bacterial infection in vivo. We investigated the role of autophagy in the response of intestinal epithelial and antigen-presenting cells to Salmonella infection in mice. METHODS: We generated mice deficient in Atg16l1 in epithelial cells (Atg16l1(f/f) × Villin-cre) or CD11c(+) cells (Atg16l1(f/f) × CD11c-cre); these mice were used to assess cell type-specific antibacterial autophagy. All responses were compared with Atg16l1(f/f) mice (controls). Mice were infected with Salmonella enterica serovar typhimurium; cecum and small-intestine tissues were collected for immunofluorescence, histology, and quantitative reverse-transcription polymerase chain reaction analyses of cytokines and AMPs. Modulators of autophagy were screened to evaluate their effects on antibacterial responses in human epithelial cells. RESULTS: Autophagy was induced in small intestine and cecum after infection with S typhimurium, and required Atg16l1. S typhimurium colocalized with microtubule-associated protein 1 light chain 3ß (Map1lc3b or LC3) in the intestinal epithelium of control mice but not in Atg16l1(f/f) × Villin-cre mice. Atg16l1(f/f) × Villin-cre mice also had fewer Paneth cells and abnormal granule morphology, leading to reduced expression of AMPs. Consistent with these defective immune responses, Atg16l1(f/f) × Villin-cre mice had increased inflammation and systemic translocation of bacteria compared with control mice. In contrast, we observed few differences between Atg16l1(f/f) × CD11c-cre and control mice. Trifluoperazine promoted autophagy and bacterial clearance in HeLa cells; these effects were reduced upon knockdown of ATG16L1. CONCLUSIONS: Atg16l1 regulates autophagy in intestinal epithelial cells and is required for bacterial clearance. It also is required to prevent systemic infection of mice with enteric bacteria.


Subject(s)
Autophagy/physiology , Carrier Proteins/physiology , Intestinal Mucosa/physiology , Salmonella Infections, Animal/prevention & control , Animals , Autophagy-Related Proteins , CD11c Antigen/physiology , Carrier Proteins/genetics , Disease Models, Animal , HeLa Cells , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Microfilament Proteins/physiology , Microtubule-Associated Proteins/physiology , Salmonella Infections, Animal/pathology , Salmonella Infections, Animal/physiopathology , Salmonella typhimurium/isolation & purification
20.
Biochem Biophys Res Commun ; 443(1): 321-5, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24316213

ABSTRACT

We investigated the effect of the calmodulin inhibitor and antipsychotic drug trifluoperazine on voltage-dependent K(+) (Kv) channels. Kv currents were recorded by whole-cell configuration of patch clamp in freshly isolated rabbit coronary arterial smooth muscle cells. The amplitudes of Kv currents were reduced by trifluoperazine in a concentration-dependent manner, with an apparent IC50 value of 1.58±0.48 µM. The rate constants of association and dissociation by trifluoperazine were 3.73±0.33 µM(-1) s(-1) and 5.84±1.41 s(-1), respectively. Application of trifluoperazine caused a positive shift in the activation curve but had no significant effect on the inactivation curve. Furthermore, trifluoperazine provoked use-dependent inhibition of the Kv current under train pulses (1 or 2 Hz). These findings suggest that trifluoperazine interacts with Kv current in a closed state and inhibits Kv current in the open state in a time- and use-dependent manner, regardless of its function as a calmodulin inhibitor and antipsychotic drug.


Subject(s)
Antipsychotic Agents/pharmacology , Calmodulin/antagonists & inhibitors , Coronary Vessels/drug effects , Muscle, Smooth, Vascular/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Trifluoperazine/pharmacology , Animals , Male , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL