Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Theranostics ; 11(17): 8500-8516, 2021.
Article in English | MEDLINE | ID: mdl-34373755

ABSTRACT

Rationale: Bak is a major proapoptotic Bcl2 family member and a required molecule for apoptotic cell death. High levels of endogenous Bak were observed in both small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) cell lines. Increased Bak expression was correlated with poor prognosis of NSCLC patients, suggesting that Bak protein is an attractive target for lung cancer therapy. The BH3 domain functions as death domain and is required for Bak to initiate apoptotic cell death. Thus, the BH3 domain is attractive target for discovery of Bak agonist. Methods: The BH3 death domain binding pocket (aa75-88) of Bak was chosen as a docking site for screening of small molecule Bak activators using the UCSF DOCK 6.1 program suite and the NCI chemical library (300,000 small molecules) database. The top 500 compounds determined to have the highest affinity for the BH3 domain were obtained from the NCI and tested for cytotoxicity for further screening. We identified a small molecule Bak activator BKA-073 as the lead compound. The binding affinity of BKA-073 with Bak protein was analyzed by isothermal titration calorimetry (ITC) assay. BKA-073-mediated Bak activation via oligomerization was analyzed by a cross-linking with Bis (maleimido) hexane (BMH). Sensitivity of BKA-073 to lung cancer cells in vitro was evaluated by dynamic BH3 profiling (DBP) and apoptotic cell death assay. The potency of BKA-073 alone or in combination with radiotherapy or Bcl2 inhibitor was evaluated in animal models. Results: We found that BKA-073 binds Bak at BH3 domain with high affinity and selectivity. BKA-073/Bak binding promotes Bak oligomerization and mitochondrial priming that activates its proapoptotic function. BKA-073 potently suppresses tumor growth without significant normal tissue toxicity in small cell lung cancer (SCLC) and NSCLC xenografts, patient-derived xenografts, and genetically engineered mouse models of mutant KRAS-driven cancer. Bak accumulates in radioresistant lung cancer cells and BKA-073 reverses radioresistance. Combination of BKA-073 with Bcl-2 inhibitor venetoclax exhibits strong synergy against lung cancer in vivo. Conclusions: Development of small molecule Bak activator may provide a new class of anticancer agents to treat lung cancer.


Subject(s)
Lung Neoplasms/therapy , Small Molecule Libraries/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Peptide Fragments/metabolism , Protein Binding , Protein Domains/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays/methods
3.
Mol Cancer Ther ; 16(4): 729-738, 2017 04.
Article in English | MEDLINE | ID: mdl-28119490

ABSTRACT

We previously reported that the EGFR-targeted inhibitor erlotinib induces G1 arrest of squamous cell carcinoma of the head and neck (SCCHN) cell lines without inducing significant apoptosis. Large-scale genomic studies suggest that >50% of SCCHN cases have activation of PI3K pathways. This study investigated whether cotargeting of EGFR and PI3K has synergistic antitumor effects and apoptosis induction. We examined growth suppression, apoptosis, and signaling pathway modulation resulting from single and combined targeting of EGFR and PI3K with erlotinib and BKM120, respectively, in a panel of SCCHN cell lines and a xenograft model of SCCHN. In a panel of 12 cell lines, single targeting of EGFR with erlotinib or PI3K with BKM120 suppressed cellular growth without inducing significant apoptosis. Cotargeting of EGFR and PI3K synergistically inhibited SCCHN cell line and xenograft tumor growth, but induced variable apoptosis; some lines were highly sensitive, others were resistant. Mechanistic studies revealed that the combination inhibited both axes of the mTORC1 (S6 and 4EBP1) pathway in apoptosis-sensitive cell lines along with translational inhibition of Bcl-2, Bcl-xL, and Mcl-1, but failed to inhibit p-4EBP1, Bcl-2, Bcl-xL, and Mcl-1 in an apoptosis-resistant cell line. siRNA-mediated knockdown of eIF4E inhibited Bcl-2 and Mcl-1 and sensitized this cell line to apoptosis. Our results strongly suggest that cotargeting of EGFR and PI3K is synergistic and induces apoptosis of SCCHN cell lines by inhibiting both axes of the AKT-mTOR pathway and translational regulation of antiapoptotic Bcl-2 proteins. These findings may guide the development of clinical trials using this combination of agents. Mol Cancer Ther; 16(4); 729-38. ©2017 AACR.


Subject(s)
Aminopyridines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm/drug effects , Erlotinib Hydrochloride/administration & dosage , Head and Neck Neoplasms/drug therapy , Morpholines/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , Aminopyridines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/drug effects , Drug Synergism , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Humans , Mice , Morpholines/pharmacology , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
4.
Cancer Prev Res (Phila) ; 9(1): 63-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26511491

ABSTRACT

Despite its high promise for cancer prevention and therapy, the potential utility of curcumin in cancer is compromised by its low bioavailability and weak potency. The purpose of the current study was to assess the in vitro and in vivo efficacy and pharmacokinetic parameters of the potent curcumin analogue FLLL12 in SCCHN and identify the mechanisms of its antitumor effect. IC50 values against a panel of one premalignant and eight malignant head and neck cancer cell lines as well as apoptosis assay results suggested that FLLL12 is 10- to 24-fold more potent than natural curcumin depending on the cell line and induces mitochondria-mediated apoptosis. In vivo efficacy (xenograft) and pharmacokinetic studies also suggested that FLLL12 is significantly more potent and has more favorable pharmacokinetic properties than curcumin. FLLL12 strongly inhibited the expression of p-EGFR, EGFR, p-AKT, AKT, Bcl-2, and Bid and increased the expression of Bim. Overexpression of constitutively active AKT or Bcl-2 or ablation of Bim or Bid significantly inhibited FLLL12-induced apoptosis. Further mechanistic studies revealed that FLLL12 regulated EGFR and AKT at transcriptional levels, whereas Bcl-2 was regulated at the translational level. Finally, FLLL12 strongly inhibited the AKT downstream targets mTOR and FOXO1a and 3a. Taken together, our results strongly suggest that FLLL12 is a potent curcumin analogue with more favorable pharmacokinetic properties that induces apoptosis of head and neck cancer cell lines by inhibition of survival proteins including EGFR, AKT, and Bcl-2 and increasing of the proapoptotic protein Bim.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Curcumin/analogs & derivatives , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/prevention & control , Animals , Apoptosis , Biological Availability , Cell Line, Tumor , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Mitochondria , Neoplasm Transplantation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , Reproducibility of Results
5.
FEBS Lett ; 589(23): 3640-7, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26519558

ABSTRACT

Impaired intestinal barrier function is one of the critical issues in inflammatory bowel diseases. The aim of this study is to investigate muscarinic cholinoceptor (mAChR)-mediated signaling for the amelioration of cytokine-induced barrier dysfunction in intestinal epithelium. Rat colon challenged with TNF-α and interferon γ reduced transepithelial electrical resistance (TER). This barrier injury was attenuated by muscarinic stimulation. In HT-29/B6 intestinal epithelial cells, muscarinic stimulation suppressed TNF-α-induced activation of NF-κB signaling and barrier disruption. Finally, muscarinic stimulation promoted the shedding of TNFR1, which would be a mechanism for the attenuation of TNF-α/NF-κB signaling and barrier disruption via mAChR.


Subject(s)
Intestinal Mucosa/cytology , Receptors, Muscarinic/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Colon/cytology , HT29 Cells , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , NF-kappa B/metabolism , Rats , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects
6.
J Pharmacol Sci ; 127(1): 150-3, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25704031

ABSTRACT

Regulation of intestinal secretion is important for body fluid homeostasis. We investigated the role of three MAP kinases (MAPKs) as negative regulators in muscarinic cholinoceptor (mAChR)-mediated intestinal secretion in mice. Electrophysiological analyses revealed that mAChR stimulation enhanced intestinal chloride secretion, which was further augmented by the inhibition of JNK but not by that of ERK or p38 with specific inhibitors SP600125, U0126 or SB203580, respectively. Immunoblot analyses in colonic mucosa showed that mAChR stimulation increased MAPKs phosphorylation that was suppressed by the specific inhibitor for each MAPK. This suggests that JNK is a major negative regulator in mAChR-induced intestinal secretion.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Intestinal Secretions/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscarinic Agonists/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anthracenes/pharmacology , Butadienes/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Male , Membrane Potentials/drug effects , Mice , Nitriles/pharmacology , Phosphorylation/drug effects , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Br J Pharmacol ; 171(21): 4890-901, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24923551

ABSTRACT

BACKGROUND AND PURPOSE: The pharmacological properties of particular receptors have recently been suggested to vary under different conditions. We compared the pharmacological properties of the α1B -adrenoceptor subtype in various tissue preparations and under various conditions. EXPERIMENTAL APPROACH: [(3) H]-prazosin binding to α1B -adrenoceptors in rat liver (segments, dispersed hepatocytes and homogenates) was assessed and the pharmacological profiles were compared with the functional and binding profiles in rat carotid artery and recombinant α1B -adrenoceptors. KEY RESULTS: In association and saturation-binding experiments with rat liver, binding affinity for [(3) H]-prazosin varied significantly between preparations (KD value approximately ten times higher in segments than in homogenates). The binding profile for various drugs in liver segments also deviated from the representative α1B -adrenoceptor profile observed in liver homogenates and recombinant receptors. L-765,314 and ALS-77, selective antagonists of α1B -adrenoceptors, showed high binding and antagonist affinities in liver homogenates and recombinant α1B -adrenoceptors. However, binding affinities for both ligands in the segments of rat liver and carotid artery were 10 times lower, and the antagonist potencies in α1B -adrenoceptor-mediated contractions of carotid artery were more than 100 times lower than the representative α1B -adrenoceptor profile. CONCLUSIONS AND IMPLICATIONS: In contrast to the consistent profile of recombinant α1B -adrenoceptors, the pharmacological profile of native α1B -adrenoceptors of rat liver and carotid artery varied markedly under various receptor environments, showing significantly different binding properties between intact tissues and homogenates, and dissociation between functional and binding affinities. In addition to conventional 'subtype' characterization, 'phenotype' pharmacology must be considered in native receptor evaluations in vivo and in future pharmacotherapy.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Indoles/pharmacology , Piperidines/pharmacology , Prazosin/analogs & derivatives , Prazosin/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Animals , CHO Cells , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/physiology , Cells, Cultured , Cricetulus , Hepatocytes/drug effects , Hepatocytes/metabolism , In Vitro Techniques , Liver/drug effects , Liver/metabolism , Male , Phenotype , Rats, Wistar
8.
Biochim Biophys Acta ; 1842(4): 635-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24365239

ABSTRACT

Impairment of epithelial barrier is observed in various intestinal disorders including inflammatory bowel diseases (IBD). Numerous factors may cause temporary damage of the intestinal epithelium. A complex network of highly divergent factors regulates healing of the epithelium to prevent inflammatory response. However, the exact repair mechanisms involved in maintaining homeostatic intestinal barrier integrity remain to be clarified. In this study, we demonstrate that activation of M1 muscarinic acetylcholine receptor (mAChR) augments the restitution of epithelial barrier function in T84 cell monolayers after ethanol-induced epithelial injury, via ERK-dependent phosphorylation of focal adhesion kinase (FAK). We have shown that ethanol injury decreased the transepithelial electrical resistance (TER) along with the reduction of ERK and FAK phosphorylation. Carbachol (CCh) increased ERK and FAK phosphorylation with enhanced TER recovery, which was completely blocked by either MT-7 (M1 antagonist) or atropine. The CCh-induced enhancement of TER recovery was also blocked by either U0126 (ERK pathway inhibitor) or PF-228 (FAK inhibitor). Treatment of T84 cell monolayers with interferon-γ (IFN-γ) impaired the barrier function with the reduction of FAK phosphorylation. The CCh-induced ERK and FAK phosphorylation were also attenuated by the IFN-γ treatment. Immunological and binding experiments exhibited a significant reduction of M1 mAChR after IFN-γ treatment. The reduction of M1 mAChR in inflammatory area was also observed in surgical specimens from IBD patients, using immunohistochemical analysis. These findings provide important clues regarding mechanisms by which M1 mAChR participates in the maintenance of intestinal barrier function under not only physiological but also pathological conditions.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/physiology , Intestinal Mucosa/metabolism , Receptor, Muscarinic M1/physiology , Cell Line, Tumor , Electric Impedance , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/analysis , Extracellular Signal-Regulated MAP Kinases/physiology , Focal Adhesion Protein-Tyrosine Kinases/analysis , Humans , Immunohistochemistry , Interferon-gamma/pharmacology , Phosphorylation , Receptor, Muscarinic M1/analysis
9.
Life Sci ; 93(1): 17-23, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23727356

ABSTRACT

AIMS: Recombinant systems have been used for evaluating the properties of G-protein-coupled receptors (GPCRs) on the assumption of cell surface expression. However, many GPCRs, including muscarinic acetylcholine receptors (mAChRs), have also been reported to be distributed in intracellular organelles in native tissues and cell lines. In this study, we compared the pharmacological profiles of exogenously and endogenously expressed M1-mAChRs, and evaluated the functional properties of these receptors. MAIN METHODS: Recombinant M1-mAChRs were expressed exogenously in Chinese hamster ovary cells (CHO-M1 cells) and compared with endogenously expressed M1-mAChRs in N1E-115 neuroblastoma cells. The pharmacological and functional profiles were evaluated using cell-permeable antagonists (1-quinuclidinyl-benzilate (QNB), pirenzepine and atropine) and cell-impermeable antagonists (N-methylscopolamine (NMS) or MT-7). KEY FINDINGS: M1-mAChRs were seen at the cell surface and intracellular sites in both cell lines. Under whole cell conditions, intracellular M1-mAChRs were mainly recognized by cell-permeable ligands, but scarcely by cell-impermeable ligands (at less than 100nM). In CHO-M1 cells, M1-mAChR activation by carbachol resulted in Ca(2+) mobilization, ERK1/2 phosphorylation and a reduction in thymidine incorporation, all of which were completely inhibited by MT-7, indicating the involvement of surface M1-mAChRs. In N1E-115 cells, Ca(2+) mobilization occurred through surface M1-mAChRs, whereas ERK1/2 phosphorylation and acceleration of thymidine incorporation were mediated through intracellular M1-mAChRs. SIGNIFICANCE: Exogenous and endogenous M1-mAChRs are present at both the cell surface and the intracellular organelles, and the pharmacological properties of geographically distinct M1-mAChRs are different, and may depend on cell background and/or exogenous or endogenous origin.


Subject(s)
Organelles/metabolism , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M1/metabolism , Recombinant Proteins/metabolism , Analysis of Variance , Animals , Atropine , Blotting, Western , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Mice , Microscopy, Confocal , N-Methylscopolamine , Pirenzepine , Quinuclidinyl Benzilate , Radioligand Assay , Tritium
10.
J Neurochem ; 126(3): 360-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23678982

ABSTRACT

Muscarinic acetylcholine receptors (mAChRs) are well known to transmit extracellular cholinergic signals into the cytoplasm from their position on the cell surface. However, we show here that M1-mAChRs are also highly expressed on intracellular membranes in neurons of the telencephalon and activate signaling cascades distinct from those of cell surface receptors, contributing uniquely to synaptic plasticity. Radioligand-binding experiments with cell-permeable and -impermeable ligands and immunohistochemical observations revealed intracellular and surface distributions of M1-mAChRs in the hippocampus and cortex of rats, mice, and humans, in contrast to the selective occurrence on the cell surface in other tissues. All intracellular muscarinic-binding sites were abolished in M1-mAChR-gene-knockout mice. Activation of cell surface M1-mAChRs in rat hippocampal neurons evoked phosphatidylinositol hydrolysis and network oscillations at theta rhythm, and transiently enhanced long-term potentiation. On the other hand, activation of intracellular M1-mAChRs phosphorylated extracellular-regulated kinase 1/2 and gradually enhanced long-term potentiation. Our data thus demonstrate that M1-mAChRs function at both surface and intracellular sites in telencephalon neurons including the hippocampus, suggesting a new mode of cholinergic transmission in the central nervous system.


Subject(s)
Hippocampus/metabolism , Neuronal Plasticity/physiology , Receptor, Muscarinic M1/metabolism , Aged , Aged, 80 and over , Animals , Blotting, Western , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytoplasm/chemistry , Cytoplasm/metabolism , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Synapses/metabolism , Synaptic Transmission/physiology
11.
J Gastroenterol ; 48(8): 885-96, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23242454

ABSTRACT

BACKGROUND: Muscarinic acetylcholine receptors (mAChRs) are major regulators of gut epithelial functions. However, the precise subtype composition has not been clarified. METHODS: We characterized the pharmacological profile of mAChRs on mouse colonic crypts, employing [(3)H]-N-methyl scopolamine chloride as a radioligand and several subtype-selective chemicals, and the functional aspect by measuring short-circuit current (I sc) in Ussing chambers and by evaluating MAP kinase phosphorylation in mouse colonic mucosal sheets. RESULTS: The mAChRs were detected on the crypts (K d = 163.2 ± 32.3 pM, B max = 47.3 ± 2.6 fmol/mg of total cell protein). Muscarinic toxin 7 (MT-7, M1 subtype selective) gave a displacement curve with high affinity, but there was a part insensitive to MT-7 (18.8 ± 0.4 % of the total specific binding). The MT-7-insensitive component was displaced completely by darifenacin (M3 selective) with high affinity. ACh induced an increase in I sc, which was significantly enhanced by MT-7 but was completely inhibited by darifenacin or atropine. Colitis induction resulted in a significant decrease in the density of mAChRs, which occurred mainly in the MT-7-sensitive component (M1 subtype). Immunological experiments exhibited a reduction of M1 but not of M3 signal after colitis induction. Muscarinic stimulation induced an increase in MAP kinase phosphorylation, which was completely suppressed by MT-7 and was attenuated by inflammation, in mouse colonic epithelium. CONCLUSIONS: These results suggest that mAChRs in mouse colonic epithelial cells consist of two subtypes, M1 (80 %) and M3 (20 %). The major M1 subtype was likely to regulate epithelial chloride secretion negatively and was susceptible to inflammation and may be relevant to inflammatory gut dysfunction.


Subject(s)
Colon/metabolism , Intestinal Mucosa/metabolism , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M3/metabolism , Animals , Atropine/pharmacology , Benzofurans/metabolism , Colitis/physiopathology , Colon/cytology , Colon/physiopathology , Elapid Venoms/metabolism , Epithelial Cells/metabolism , Inflammation/physiopathology , Intestinal Mucosa/cytology , Male , Mice , Mice, Inbred BALB C , N-Methylscopolamine/metabolism , Parasympatholytics/metabolism , Pyrrolidines/metabolism , Radioligand Assay
12.
Front Pharmacol ; 2: 65, 2011.
Article in English | MEDLINE | ID: mdl-22025914

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) of the cerebral cortex and cerebellum of rats were evaluated by a radioligand binding assay, employing tissue segments, or homogenates as materials. [(3)H]-epibatidine specifically bound to nAChRs in rat cortex or cerebellum, but the dissociation constants for [(3)H]-epibatidine differed between segments and homogenates (187 pM for segments and 42 pM for homogenates in the cortex and 160 pM for segments and 84 pM for homogenates in the cerebellum). The abundance of total nAChRs was approximately 310 fmol/mg protein in the segments of cortex and 170 fmol/mg protein in the segments of cerebellum, which were significantly higher than those estimated in the homogenates (115 fmol/mg protein in the homogenates of the cortex and 76 fmol/mg protein in the homogenates of the cerebellum). Most of the [(3)H]-epibatidine binding sites in the cortex segments (approximately 70% of the population) showed high affinity for nicotine (pK(i) = 7.9), dihydro-ß-erythroidine, and cytisine, but the binding sites in the cerebellum segments had slightly lower affinity for nicotine (pK(i) = 7.1). An upregulation of nAChRs by chronic administration of nicotine was observed in the cortex segments but not in the cerebellum segments with [(3)H]-epibatidine as a ligand. The upregulation in the cortex was caused by a specific increase in the high-affinity sites for nicotine (probably α4ß2). The present study shows that the native environment of nAChRs is important for a precise quantitative as well as qualitative estimation of nAChRs in rat brain.

13.
J Neurochem ; 118(6): 958-67, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21740440

ABSTRACT

Signaling by muscarinic agonists is thought to result from the activation of cell surface acetylcholine receptors (mAChRs) that transmit extracellular signals to intracellular systems. In N1E-115 neuroblastoma cells, we detected both plasma membrane and intracellular M(1) -mAChRs using both biochemical and pharmacological methods. In intact cells, both plasma membrane and intracellular M(1) -mAChRs were detected by the hydrophobic ligand probe, 1-quinuclidinyl-[phenyl-4-(3) H]-benzilate ([(3) H]-QNB) whereas the hydrophilic probe, 1-[N-methyl-(3) H] scopolamine ([(3) H]-NMS), detected only cell surface receptors. These probes detected comparable numbers of receptors in isolated membrane preparations. Immunohistochemical studies with M(1) -mAChR antibody also detected both cell-surface and intracellular M(1) -mAChRs. Carbachol-stimulated phosphatidylinositol hydrolysis and Ca(2+) mobilization were completely inhibited by a cell-impermeable M(1) antagonist, muscarinic toxin -7 and the G(q/11) inhibitor YM-254890. However, carbachol-stimulated extracellular-regulated kinase 1/2 activation was unaffected by muscarinic toxin-7, but was blocked by the cell-permeable antagonist, pirenzepine. extracellular regulated kinase 1/2 phosphorylation was resistant to blockade of G(q/11) (YM-254890) and protein kinase C (bisindolylmaleimide I). Our data suggest that the geographically distinct M(1) -mAChRs (cell surface versus intracellular) can signal via unique signaling pathways that are differentially sensitive to cell-impermeable versus cell-permeable antagonists. Our data are of potential physiological relevance to signaling that affects both cognitive and neurodegenerative processes.


Subject(s)
Neuroblastoma/metabolism , Receptor, Muscarinic M1/metabolism , Animals , Atropine/pharmacology , Blotting, Western , Calcium/metabolism , Carbachol/metabolism , Carbachol/pharmacology , Cell Line, Tumor , Elapid Venoms/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunohistochemistry , Inositol Phosphates/metabolism , Kinetics , Mice , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , N-Methylscopolamine/pharmacology , Peptides, Cyclic/pharmacology , Pirenzepine/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinuclidinyl Benzilate/pharmacology , Receptors, Cell Surface/drug effects
14.
J Pharmacol Sci ; 112(4): 444-51, 2010.
Article in English | MEDLINE | ID: mdl-20424384

ABSTRACT

Muscarinic acetylcholine receptors (mAChRs) of rat cerebral cortex were evaluated using a tissue segment radioligand binding assay. [(3)H]-Quinuclidinyl benzilate (QNB, a hydrophobic ligand) specifically bound to mAChRs in the cortex segments. The total mAChRs level was approximately 2,000 fmol/mg protein, which was estimated after incubation for 120 min at 37 degrees C or for 8 h at 4 degrees C. These mAChRs were a mixture of high- and low-affinity sites for N-methylscopolamine (NMS) in a 70:30 ratio. In contrast, only a single high-affinity site for NMS was detected following incubation for 30 min at 37 degrees C, whose abundance was about 70% of that of the total mAChRs. Atropine showed a single affinity for mAChRs under all conditions. These indicate that mAChRs are constitutively expressed not only on plasma membrane sites but also at intracellular sites in rat cerebral cortex and that the receptors at both sites have different affinities for NMS. Acetylcholine completely inhibited [(3)H]-QNB binding to both mAChRs without any change in the subcellular distribution, suggesting the possibility that acetylcholine can access, and bind to, both mAChRs in intact tissue. Two different affinity states for acetylcholine were detected only in plasma membrane mAChRs at 37 degrees C. The present study demonstrates a unique subcellular distribution, and distinct pharmacological profiles, of mAChRs in rat cerebral cortex.


Subject(s)
Cerebral Cortex/metabolism , Receptors, Muscarinic/metabolism , Acetylcholine/pharmacology , Animals , Atropine/pharmacology , Cerebral Cortex/drug effects , In Vitro Techniques , Male , N-Methylscopolamine/pharmacology , Quinuclidinyl Benzilate/metabolism , Radioligand Assay , Rats , Rats, Wistar
15.
Eur J Pharmacol ; 599(1-3): 146-51, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-18938154

ABSTRACT

Distinct muscarinic acetylcholine receptor subtypes widely distribute in stomach tissues and are involved in many physiological functions. Although mRNA of M(1) subtype was found in gastric mucosa, the M(1) subtype has not been detected by conventional membrane binding assays. In the present study, muscarinic receptor subtypes in the rat stomach were reevaluated by using the tissue segment binding technique recently developed to recognize the inherent/native profiles of receptors without receptor environment perturbation. [(3)H]-N-methylscopolamine (NMS) bound to muscarinic receptors in the intact segments of rat gastric mucosa and muscle layers. The muscarinic receptors in the mucosal segments were composed of M(1), M(2) and M(3) subtypes, among which the M(1) subtype selectively showed high affinity for pirenzepine. However, in the membrane preparations, binding sites with high affinity for pirenzepine could not be detected. In the muscle layer, M(2) and M(3) subtypes, but not M(1), were identified in tissue segment and conventional membrane binding assays. Western blotting analysis recognized the M(1) subtype in the membrane preparations of mucosal but not muscle layers. Chronic immobilization stress increased the M(3) subtype in mucosal and muscle layers and decreased the M(2) subtype in the muscle layer, whereas M(1) and M(2) subtypes in mucosal layer did not change after the stress. The current study shows that M(1) subtype occurs as a pirenzepine-high affinity entity in intact segments of rat gastric mucosa, but that it loses the affinity for pirenzepine upon homogenization. Careful identification of native in vivo muscarinic receptors may further elucidate their functions in stomach.


Subject(s)
Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M3/metabolism , Stress, Psychological/metabolism , Animals , Binding Sites , Blotting, Western , Gastric Mucosa/metabolism , Male , Pirenzepine/metabolism , Protein Binding , Radioligand Assay/methods , Rats , Rats, Wistar , Restraint, Physical , Stomach/physiology
16.
Eur J Pharmacol ; 584(2-3): 222-8, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18336813

ABSTRACT

The pharmacological profiles of alpha1-adrenoceptors for ketanserin, prazosin, silodosin, and BMY 7378 (8-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-8-azaspiro[4,5]decane-7,9-dione dihydrochloride) were examined under different assay conditions. Among the tested antagonists and alpha1-adrenoceptors subtypes, ketanserin showed significantly lower affinity for the alpha1B-adrenoceptor subtype in intact tissue sampled from the rat tail artery, thoracic aorta, and cerebral cortex (functional pKB and binding pKi were approximately 6), than in cerebral cortex membrane preparations or whole cell and membrane preparations of alpha1B-adrenoceptor transfected human embryonic kidney 293T (HEK 293T) cells (pKi was approximately 8). In these tissues and cells, however, ketanserin showed a similar affinity (pKi = approximately 8) for alpha1A- and alpha1D-adrenoceptors even though the assays were conducted under different conditions. In contrast, the affinities of alpha1A-, alpha1B-, and alpha1D-adrenoceptors for prazosin, silodosin, and BMY 7378 did not significantly change under different assay conditions and in different tissues. The present study reveals that the pharmacological profiles of native alpha 1B-adrenoceptors for ketanserin is strongly influenced by the assay conditions and suggest that antagonist affinity is not necessarily constant.


Subject(s)
Adrenergic alpha-Antagonists/metabolism , Aorta, Thoracic/metabolism , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Ketanserin/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Tail/blood supply , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Animals , Aorta, Thoracic/drug effects , Arteries/metabolism , Binding, Competitive , Cell Line , Dose-Response Relationship, Drug , Humans , Indoles/metabolism , Ketanserin/pharmacology , Male , Piperazines/metabolism , Prazosin/metabolism , Protein Binding , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/genetics , Recombinant Proteins/metabolism , Subcellular Fractions , Transfection , Vasoconstriction
17.
J Pharmacol Sci ; 106(2): 271-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18270476

ABSTRACT

Muscarinic receptors in the human and rat lower urinary tract (urinary bladder detrusor muscle and mucosa, and prostate) were identified by intact tissue segment binding assays with two radioligands, and the effects of prolonged receptor activation in vitro on muscarinic receptors were examined. Hydrophilic [(3)H]-NMS and hydrophobic [(3)H]-QNB bound to the detrusor muscle segments with the same density, suggesting that the muscarinic receptors were localized at the plasma membrane. While the density of muscarinic receptor was higher in detrusor muscle than in the bladder mucosa and prostate, there was no species-specific difference either in density or in subtype distribution (M(1), M(2), and M(3) subtypes in detrusor; M(2) and M(3) subtypes in bladder mucosa; and M(1) and M(2) subtypes in prostate). Incubation of detrusor strips with carbachol decreased [(3)H]-NMS binding sites within 20 min, followed by a reduction of [(3)H]-QNB binding sites after a 60-min lag phase. The loss of the binding sites over 3 h after carbachol treatment was the same (approximately 40%) for both radioligands. The present intact tissue segment binding assay reveals tissue-specific and plasma membrane distribution of distinct muscarinic receptor subtypes and their dynamic changes (internalization and down-regulation) in lower urinary tract of humans and rats.


Subject(s)
Cell Membrane/metabolism , Mucous Membrane/metabolism , Muscle, Smooth/metabolism , Prostate/metabolism , Receptors, Muscarinic/metabolism , Urinary Bladder/metabolism , Aged , Animals , Carbachol/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Muscarinic Agonists/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , N-Methylscopolamine/metabolism , Quinuclidinyl Benzilate/metabolism , Rats , Rats, Wistar , Receptors, Muscarinic/physiology , Urinary Bladder/drug effects , Urinary Bladder/physiology
18.
J Pharmacol Sci ; 98(4): 331-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16082176

ABSTRACT

The radioligand binding assay technique is an extremely powerful tool for studying receptors. It allows an analysis of the interactions of hormones, neurotransmitters, and related drugs with their receptors. Most of the binding assays have widely been applied to crude membrane fractions prepared from many tissues, but in the conventional method, there are some limitations such as a yield loss of receptor-bearing membranes and a change in receptor environment upon homogenization and fractionation. Recently, in order to overcome these problems, a binding assay has been developed using intact tissue segments. This article presents a brief overview of the tissue segment binding assay that has been developed mainly in our department. Practical guidelines for setting up this new assay are presented, including segment preparation, choice of appropriate radioligand, optimizing assay conditions, and appropriate methods for data analysis. The unique advantages and disadvantages of the tissue segment binding method are discussed in comparison with those of conventional membrane binding methods. We suggest that the tissue segment binding method is a powerful tool for detecting the native properties of receptors occurring in tissues and cells without altering their environment.


Subject(s)
Radioligand Assay/methods , Receptors, Cell Surface/analysis , Animals , Binding, Competitive , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL