Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 620
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pharmacol Res ; 160: 105148, 2020 10.
Article in English | MEDLINE | ID: mdl-32858121

ABSTRACT

Neuropathic pain, a specific type of chronic pain resulting from persistent nervous tissue lesions, is a debilitating condition that affects about 7% of the population. This condition remains particularly difficult to treat because of the poor understanding of its underlying mechanisms. Drugs currently used to alleviate this chronic pain syndrome are of limited benefit due to their lack of efficacy and the elevated risk of side effects, especially after a prolonged period of treatment. Although drugs targeting G protein-coupled receptors (GPCR) also have several limitations, such as progressive loss of efficacy due to receptor desensitization or unavoidable side effects due to wide receptor distribution, the identification of several molecular partners that contribute to the fine-tuning of receptor activity has raised new opportunities for the development of alternative therapeutic approaches. Regulators of G protein signalling (RGS) act intracellularly by influencing the coupling process and activity of G proteins, and are amongst the best-characterized physiological modulators of GPCR. Changes in RGS expression have been documented in a range of models of neuropathic pain, or after prolonged treatment with diverse analgesics, and could participate in altered pain processing as well as impaired physiological or pharmacological control of nociceptive signals. The present review summarizes the experimental data that implicates RGS in the development of pain with focus on the pathological mechanisms of neuropathic pain, including the impact of neuropathic lesions on RGS expression and, reciprocally, the influence of modifying RGS on GPCRs involved in the modulation of nociception as well as on the outcome of pain. In this context, we address the question of the relevance of RGS as promising targets in the treatment of neuropathic pain.


Subject(s)
GTP-Binding Proteins/drug effects , Neuralgia/drug therapy , Signal Transduction/drug effects , Animals , Chronic Pain , GTP-Binding Proteins/agonists , Humans , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/drug effects
2.
Arch Toxicol ; 94(11): 3861-3875, 2020 11.
Article in English | MEDLINE | ID: mdl-32749514

ABSTRACT

Organophosphate compounds (OPs) induce both acute and delayed neurotoxic effects, the latter of which is believed to involve their interaction with proteins other than acetylcholinesterase. However, few OP-binding proteins have been identified that may have a direct role in OP-induced delayed neurotoxicity. Given their ability to disrupt Ca2+ homeostasis, a key aim of the current work was to investigate the effects of sub-lethal neurite outgrowth inhibitory levels of OPs on the Ca2+-dependent enzyme tissue transglutaminase (TG2). At 1-10 µM, the OPs phenyl saligenin phosphate (PSP) and chlorpyrifos oxon (CPO) had no effect cell viability but induced concentration-dependent decreases in neurite outgrowth in differentiating N2a neuroblastoma cells. The activity of TG2 increased in cell lysates of differentiating cells exposed for 24 h to PSP and chlorpyrifos oxon CPO (10 µM), as determined by biotin-cadaverine incorporation assays. Exposure to both OPs (3 and/or 10 µM) also enhanced in situ incorporation of the membrane permeable substrate biotin-X-cadaverine, as indicated by Western blot analysis of treated cell lysates probed with ExtrAvidin peroxidase and fluorescence microscopy of cell monolayers incubated with FITC-streptavidin. Both OPs (10 µM) stimulated the activity of human and mouse recombinant TG2 and covalent labelling of TG2 with dansylamine-labelled PSP was demonstrated by fluorescence imaging following SDS-PAGE. A number of TG2 substrates were tentatively identified by mass spectrometry, including cytoskeletal proteins, chaperones and proteins involved protein synthesis and gene regulation. We propose that the elevated TG2 activity observed is due to the formation of a novel covalent adduct between TG2 and OPs.


Subject(s)
Cell Differentiation/drug effects , GTP-Binding Proteins/drug effects , Neuroblastoma/metabolism , Neuronal Outgrowth/drug effects , Organophosphates/toxicity , Transglutaminases/drug effects , Amines/metabolism , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Cell Line, Tumor , Cell Survival , Chlorpyrifos/analogs & derivatives , Chlorpyrifos/toxicity , Humans , Mice , Organophosphorus Compounds/toxicity , Protein Glutamine gamma Glutamyltransferase 2 , Proteomics , Rats , Reactive Oxygen Species
3.
Int J Mol Sci ; 21(7)2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32260198

ABSTRACT

In a recent report, no significance of transglutaminase 2 (TGase 2) was noted in the analyses of expression differences between normal and clear cell renal cell carcinoma (ccRCC), although we found that knock down of TGase 2 induced significant p53-mediated cell death in ccRCC. Generally, to find effective therapeutic targets, we need to identify targets that belong specifically to a cancer phenotype that can be differentiated from a normal phenotype. Here, we offer precise reasons why TGase 2 may be the first therapeutic target for ccRCC, according to several lines of evidence. TGase 2 is negatively regulated by von Hippel-Lindau tumor suppressor protein (pVHL) and positively regulated by hypoxia-inducible factor 1-α (HIF-1α in renal cell carcinoma (RCC). Therefore, most of ccRCC presents high level expression of TGase 2 because over 90% of ccRCC showed VHL inactivity through mutation and methylation. Cell death, angiogenesis and drug resistance were specifically regulated by TGase 2 through p53 depletion in ccRCC because over 90% of ccRCC express wild type p53, which is a cell death inducer as well as a HIF-1α suppressor. Although there have been no detailed studies of the physiological role of TGase 2 in multi-omics analyses of ccRCC, a life-long study of the physiological roles of TGase 2 led to the discovery of the first target as well as the first therapeutic treatment for ccRCC in the clinical field.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/genetics , GTP-Binding Proteins/genetics , Kidney Neoplasms/genetics , Transglutaminases/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Drug Resistance, Neoplasm , GTP-Binding Proteins/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Molecular Targeted Therapy , Precision Medicine , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/drug effects , Tumor Suppressor Protein p53/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
4.
Am J Physiol Endocrinol Metab ; 304(7): E703-10, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23360825

ABSTRACT

The developmentally regulated GTP-binding protein-2 (DRG2) is a novel subclass of GTP-binding proteins. Many functional characteristics of osteoclasts (OC) are associated with small GTPases. We hypothesized that DRG2 affects bone mass via modulating OC activity. Using DRG2 transgenic mice, we investigated the role of DRG2 in bone remodeling. DRG2 overexpression caused a decrease in bone mass and an increase in the number and activity of OC in vivo. DRG2 overexpression increased fusion, spreading, survival, and resorption activity of OC in vitro. Downregulation of DRG2 by siRNA decreased fusion, spreading, and survival of OC, supporting the observations found in DRG2 transgenic OC. Transgenic mature OCs were larger, with actin rings and higher ERK, Akt, Rac1 and Rho activities than wild-type OCs. Inhibition of these proteins abolished the effects of DRG2 on formation of large OCs with actin rings, implying that DRG2 affects cytoskeleton reorganization in a Rac1/Rho/ERK/Akt-dependent manner. In summary, DRG2 is associated with survival and cytoskeleton organization of OC under influence of macrophage colony-stimulating factor, and its overexpression leads to elevated bone resorptive activity of OC, resulting in bone loss.


Subject(s)
Bone Remodeling/physiology , Bone Resorption/etiology , GTP-Binding Proteins/metabolism , Osteoclasts/metabolism , Signal Transduction/physiology , Animals , Bone Remodeling/drug effects , Bone Remodeling/genetics , Cell Fusion , Cell Movement/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Cell Survival/physiology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/genetics , Macrophage Colony-Stimulating Factor/drug effects , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Transgenic , Osteoclasts/drug effects , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects
5.
Brain Behav Immun ; 27(1): 71-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23036922

ABSTRACT

A range of adverse, early life environmental influences such as viral infection and social deprivation are thought to increase risk of psychiatric illness later in life. Here, we used peripheral administration of the viral infection mimic polyriboinosinic-polyribocytidylic acid (polyI:C) to compare the consequences of peripubertal infection and isolation rearing. Isolation rearing induced deficits in sensorimotor gating and recognition memory while no changes in social interaction or spatial learning were observed. PolyI:C injection during the peripubertal period markedly increased expression of interferon-stimulated genes (Ifit2, Prkr, Mx2 and Irf7) in the hippocampal dentate gyrus demonstrating that peripheral administration of the viral mimic in the adolescent animal does have direct effects in the brain. Peripubertal infection mimicry induced a similar but later emerging behavioural deficit in prepulse inhibition implying the existence of a peripubertal window of opportunity for viral-mediated cytokine increases to impact brain development and function. PolyI:C treatment also impaired novel object recognition but did not alter spatial reference memory or social interaction. Combining the polyI:C challenge with social isolation did not exacerbate the behavioural deficits seen with isolation rearing alone. Using Irf7 as a marker, peripubertal viral infection mimicry, isolation rearing and a combination of both were all seen to produce a long-lasting molecular imprint on the interferon-associated signalling pathway in the principal neuron population of the hippocampal dentate gyrus. The data suggest that the sensitivity of brain structure and function to disruption by viral infection extends into the peripubertal period. Moreover, augmented interferon signalling in hippocampus may represent a common molecular imprint of environmental insults associated with neuropsychiatric illnesses like schizophrenia.


Subject(s)
Behavior, Animal , Dentate Gyrus , Interferon Inducers/pharmacology , Interferon Regulatory Factor-7 , Interferons/metabolism , Poly I-C/pharmacology , Virus Diseases/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Interferon Regulatory Factor-7/drug effects , Interferon Regulatory Factor-7/metabolism , Male , Myxovirus Resistance Proteins , Rats , Rats, Wistar , Sensory Gating/drug effects , Sensory Gating/physiology , Sexual Maturation/physiology , Social Isolation
6.
Diabetes Obes Metab ; 15(3): 284-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23061407

ABSTRACT

No clinical studies on the lipolytic effect of guanine nucleotide-binding protein ß3 subunit gene (GNB3) 825T polymorphism have been performed. This study was a subinvestigation of a 12-week randomized controlled trial (NCT01184560) for the additive effect of orlistat on sibutramine treatment. The analysis involved 101 obese females aged 18-49 years, genotyped at the GNB3 825 locus. To exclude any influence from potential confounders, we used an analysis of covariance model. After the intervention, fat mass proportion in total weight loss was significantly lower in subjects with a T allele than in those without a T allele (p = 0.034). GNB3 825T allele was associated with blunted fat mass reduction in obese females.


Subject(s)
Adiposity/genetics , GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Obesity/genetics , Weight Loss/genetics , Adiposity/drug effects , Adult , Alleles , Anti-Obesity Agents/therapeutic use , Appetite Depressants/therapeutic use , Cyclobutanes/therapeutic use , Drug Synergism , Female , GTP-Binding Proteins/drug effects , Genotype , Humans , Lactones/therapeutic use , Middle Aged , Obesity/drug therapy , Obesity/metabolism , Orlistat , Polymorphism, Genetic , Weight Loss/drug effects
7.
Purinergic Signal ; 9(1): 51-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22806273

ABSTRACT

Inosine is the first metabolite of adenosine. It exerts an antinociceptive effect by activating the adenosine A(1) and A(2A) receptors. We have previously demonstrated that inosine exhibits antinociceptive properties in acute and chronic mice models of nociception. The aim of this study was to investigate the involvement of pertussis toxin-sensitive G-protein-coupled receptors, as well as K(+) and Ca(2+) channels, in the antinociception promoted by inosine in the formalin test. Mice were pretreated with pertussis toxin (2.5 µg/site, i.t., an inactivator of G(i/0) protein); after 7 days, they received inosine (10 mg/kg, i.p.) or morphine (2.5 mg/kg, s.c., used as positive control) immediately before the formalin test. Another group of animals received tetraethylammonium (TEA) or 4-aminopyridine (4-AP) (1 µg/site, i.t., a non-specific voltage-gated K(+) channel blockers), apamin (50 ng/site, i.t., a small conductance Ca(2+)-activated K(+) channel blocker), charybdotoxin (250 pg/site, i.t., a large-conductance Ca(2+)-activated K(+) channel blocker), glibenclamide (100 µg/site, i.t., an ATP-sensitive K(+) channel blocker) or CaCl(2) (200 nmol/site, i.t.). Afterwards, the mice received inosine (10 mg/kg, i.p.), diclofenac (10 mg/kg, i.p., a positive control), or morphine (2.5 mg/kg, s.c., a positive control) immediately before the formalin test. The antinociceptive effect of inosine was reversed by the pre-administration of pertussis toxin (2.5 µg/site, i.t.), TEA, 4-aminopyridine, charybdotoxin, glibenclamide, and CaCl(2), but not apamin. Further, all K(+) channel blockers and CaCl(2) reversed the antinociception induced by diclofenac and morphine, respectively. Taken together, these data suggest that the antinociceptive effect of inosine is mediated, in part, by pertussis toxin-sensitive G-protein coupled receptors and the subsequent activation of voltage gated K(+) channel, large conductance Ca(2+)-activated and ATP-sensitive K(+) channels or inactivation of voltage-gated Ca(2+) channels. Finally, small conductance Ca(2+)-activated K(+) channels are not involved in the antinociceptive effect of inosine.


Subject(s)
Analgesics , Calcium Channels/physiology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/physiology , Inosine/pharmacology , Pertussis Toxin/pharmacology , Potassium Channels/physiology , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diclofenac/pharmacology , Injections, Spinal , Male , Mice , Morphine/pharmacology , Pain Measurement
8.
Addict Biol ; 18(4): 614-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-21812868

ABSTRACT

In the present study, we investigated the possible development of tolerance to the antihyperalgesic effect of µ-opioid receptor (MOR) agonists under a neuropathic pain-like state. Repeated treatment with fentanyl, but not morphine or oxycodone, produced a rapid development of tolerance to its antihyperalgesic effect in mice with sciatic nerve ligation. Like the behavioral study, G-protein activation induced by fentanyl was significantly reduced in membranes obtained from the spinal cord of nerve-ligated mice with in vivo repeated injection of fentanyl. In ß-endorphin-knockout mice with nerve ligation, developed tolerance to the antihyperalgesic effect of fentanyl was abolished, and reduced G-protein activation by fentanyl after nerve ligation with fentanyl was reversed to the normal level. The present findings indicate that released ß-endorphin within the spinal cord may be implicated in the rapid development of tolerance to fentanyl under a neuropathic pain-like state.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance/physiology , Fentanyl/pharmacology , Neuralgia/drug therapy , Receptors, Opioid, mu/agonists , Spinal Cord/metabolism , beta-Endorphin/physiology , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Female , Fentanyl/administration & dosage , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hot Temperature , Hyperalgesia/drug therapy , Injections, Subcutaneous , Ligation , Male , Mice , Mice, Knockout , Morphine/administration & dosage , Morphine/pharmacology , Neuralgia/metabolism , Oxycodone/administration & dosage , Oxycodone/pharmacology , Pain Measurement/methods , Pain Threshold/drug effects , Radioligand Assay , Receptors, Opioid, mu/physiology , Sciatic Nerve/surgery , Sodium Chloride/administration & dosage , beta-Endorphin/genetics
9.
Endocr J ; 60(3): 337-46, 2013.
Article in English | MEDLINE | ID: mdl-23171706

ABSTRACT

The aim of this study was to determine whether dexmedetomidine (DEX) and medetomidine (MED), α2-adrenergic agonists clinically used as sedatives, influence insulin secretion from rat pancreatic islets. Islets were isolated from adult male Wistar rats after collagenase digestion. Static incubation was used to determine effects of DEX or MED on insulin secretion and ionic-channel currents of ß-cells. Results indicate that both drugs dose-dependently inhibit insulin secretion, DEX more potently than MED. The inhibitory effects were attenuated by addition of yohimbine or by pretreatment of rats with pertussis toxin (PTX). 10 nM DEX decreased the current amplitude of voltage-dependent Ca2+ channels, but this did not occur when the N-type Ca2+ channel blocker ω-conotoxin was added. In the presence of tetraethylammonium, a classical voltage-gated K+ channel (Kv channel) blocker, the magnitude of inhibition of insulin secretion by MED was reduced. However, when tolbutamide, a specific blocker of the ATP-sensitive K+ channel (KATP channel), was present, the magnitude of MED inhibition of insulin secretion was not influenced, suggesting that Kv-channel activity alteration, but not that of KATP channels, is involved in MED-associated insulin secretory inhibition. The Kv-channel currents were increased during 1 nM MED exposure at membrane potentials ranging from -30 mV to -10 mV, where action potentials were generated in response to glucose stimulation. These results indicate that DEX and MED inhibit insulin secretion through an α2-adrenoceptor and PTX-sensitive GTP-binding protein pathway that eventually involves Kv channel activation and Ca2+ channel inhibition.


Subject(s)
Dexmedetomidine/adverse effects , Hypnotics and Sedatives/adverse effects , Insulin Antagonists , Insulin/metabolism , Islets of Langerhans/drug effects , Medetomidine/adverse effects , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Dexmedetomidine/pharmacology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/physiology , Hypnotics and Sedatives/pharmacology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Medetomidine/pharmacology , Pertussis Toxin/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/physiology , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/physiology , Tetraethylammonium/pharmacology , Yohimbine/pharmacology , omega-Conotoxins/pharmacology
10.
Biol Reprod ; 86(4): 106, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22262694

ABSTRACT

The testis is an immunoprivileged site where local cell-initiated innate immunity plays a crucial role in antimicrobial responses. Toll-like receptors (TLRs) mediate innate immune responses in testicular somatic cells. Although several TLRs are expressed in some stages of male germ cells, the potential role of TLRs in triggering antimicrobial responses in the germ cells has yet to be exclusively studied. The current study demonstrates that TLR3 is constitutively expressed in spermatogonia and spermatocytes and can be activated by a synthetic double-strained RNA analog, polyinosinic-polycytidylic acid. TLR3 activation in these male germ cells up-regulates the expression of proinflammatory cytokines, such as interleukin IL1B, IL6, and tumor necrosis factor alpha, through activation of nuclear factor kappa B; it also induces production of type 1 interferons (IFNA and IFNB) through the activation of IFN regulatory factor 3. In addition, TLR3 activation increases the production of two major antiviral proteins, namely, double-stranded RNA-activated protein kinase and MX1 protein, by germ cells. Data in this article describe an antiviral response of male germ cells through the activation of TLR3 in vitro.


Subject(s)
Cytokines/metabolism , Spermatocytes/metabolism , Spermatogonia/metabolism , Toll-Like Receptor 3/immunology , Toll-Like Receptor 3/metabolism , Animals , Cytokines/drug effects , Cytokines/immunology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , Immunity, Innate/immunology , Interferon Inducers/pharmacology , Interferon Type I/drug effects , Interferon Type I/immunology , Interferon Type I/metabolism , Male , Mice , Mice, Inbred C57BL , Myxovirus Resistance Proteins , Poly I-C/pharmacology , Spermatocytes/immunology , Spermatogonia/immunology , Toll-Like Receptor 3/drug effects , Up-Regulation/drug effects , Up-Regulation/immunology , eIF-2 Kinase/drug effects , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism
11.
Sci Rep ; 11(1): 3501, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568753

ABSTRACT

The development of functionally selective or biased ligands is a promising approach towards drugs with less side effects. Biased ligands for G protein-coupled receptors can selectively induce G protein activation or ß-arrestin recruitment. The consequences of this selective action on cellular functions, however, are not fully understood. Here, we investigated the impact of five biased and balanced dopamine D2 receptor agonists and antagonists on the global protein expression in HEK293T cells by untargeted nanoscale liquid chromatography-tandem mass spectrometry. The proteome analysis detected 5290 protein groups. Hierarchical clustering and principal component analysis based on the expression levels of 1462 differential proteins led to a separation of antagonists and balanced agonist from the control treatment, while the biased ligands demonstrated larger similarities to the control. Functional analysis of affected proteins revealed that the antagonists haloperidol and sulpiride regulated exocytosis and peroxisome function. The balanced agonist quinpirole, but not the functionally selective agonists induced a downregulation of proteins involved in synaptic signaling. The ß-arrestin-preferring agonist BM138, however, regulated several proteins related to neuron function and the dopamine receptor-mediated signaling pathway itself. The G protein-selective partial agonist MS308 influenced rather broad functional terms such as DNA processing and mitochondrial translation.


Subject(s)
Dopamine Agonists/pharmacology , Mitochondria/drug effects , Receptors, Dopamine D2/drug effects , beta-Arrestins/metabolism , Arrestins/metabolism , Dopamine/metabolism , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Mitochondria/metabolism , Quinpirole/pharmacology , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects
12.
Eur J Pharmacol ; 890: 173640, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33045198

ABSTRACT

Opioids are the most powerful analgesics used clinically; however, severe side effects limit their long-term use. Various concepts involving biased intracellular signaling, partial agonism or multi-receptor targeting have been proposed to identify novel opioids with increased analgesic efficacy but reduced side effects. The search for such 'better opioids' implies screening of huge compound libraries and requires highly reliable, easy to perform and high throughput screening (HTS) assays. Here, we utilize an established membrane potential assay to monitor activation of G protein-coupled inwardly rectifying potassium (GIRK) channels, one of the main effectors of opioid receptor signaling, as readout to determine pharmacological profiles of opioids in a non-invasive manner. Specifically, in this study, we optimize assay conditions and extend the application of this assay to screen all four members of the opioid receptor family, stably expressed in AtT-20 and HEK293 cells. This ultra-sensitive system yielded EC50 values in the nano-molar range. We further validate this system for screening cells stably co-expressing two opioid receptors, which could be a valuable tool for investigating bi-functional ligands and studying interactions between receptors. Additionally, we demonstrate the utility of this assay to study antagonists as well as ligands with varying efficacies. Our results suggest that this assay could easily be up-scaled to HTS assay in order to efficiently study receptor activation and screen for novel opioids.


Subject(s)
GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , High-Throughput Screening Assays/methods , Membrane Potentials/drug effects , Receptors, Opioid/metabolism , Signal Transduction/drug effects , Analgesics, Opioid/pharmacology , Animals , Cell Line, Tumor , Cell Separation , Flow Cytometry , Fluorescence , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Proteins/agonists , GTP-Binding Proteins/antagonists & inhibitors , HEK293 Cells , Humans , Ligands , Mice
13.
Neurobiol Dis ; 38(1): 27-35, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20053373

ABSTRACT

We investigated the functional characteristics of pre- and postsynaptic cholinergic transmission in APPswe/PS1dE9 double transgenic mice at a young age (7-10 weeks) before the onset of amyloid plaque formation and at adult age (5-6 months) at its onset. We compared brain slices from cerebral cortex and hippocampus with amyloid deposits to slices from striatum with no amyloid plaques by 6 months of age. In young transgenic mice we found no impairments of preformed and newly synthesized [(3)H]-ACh release, indicating intact releasing machinery and release turnover, respectively. Adult transgenic mice displayed a significant increase in preformed [(3)H]-ACh release in cortex but a decrease in hippocampus and striatum. The extent of presynaptic muscarinic autoregulation was unchanged. Evoked release of newly synthesized [(3)H]-ACh was significantly reduced in the cortex and hippocampus but unchanged in the striatum. Carbachol-induced G-protein activation in cortical membranes displayed decreased potency but normal efficacy in adult animals and no changes in young animals. These results indicate that functional pre- and postsynaptic cholinergic deficits are not present in APPswe/PS1dE9 transgenic mice before 10 weeks of age, but develop along with beta-amyloid accumulation in the brain.


Subject(s)
Acetylcholine/deficiency , Alzheimer Disease/metabolism , Amyloid/metabolism , Brain/metabolism , Cholinergic Fibers/metabolism , Nerve Degeneration/metabolism , Age Factors , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/growth & development , Brain/physiopathology , Brain Chemistry/genetics , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cholinergic Agonists/pharmacology , Cholinergic Fibers/pathology , Disease Models, Animal , Down-Regulation/genetics , Female , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Transgenic , Nerve Degeneration/pathology , Organ Culture Techniques , Presenilin-1/genetics , Receptors, Muscarinic/metabolism
14.
J Exp Med ; 190(1): 101-11, 1999 Jul 05.
Article in English | MEDLINE | ID: mdl-10429674

ABSTRACT

Trichosanthin (TCS), an active protein component isolated from a traditional Chinese medicinal herb Trichosanthes kirilowii, has been shown to inhibit HIV infection and has been applied in clinical treatment of AIDS. The recent development that chemokines and chemokine receptors play important roles in HIV infection led us to investigate the possible functional interaction of TCS with chemokines and their receptors. This study demonstrated that TCS greatly enhanced both RANTES (regulated upon activation, normal T cell expressed and secreted)- and stromal cell-derived factor (SDF)-1 alpha-stimulated chemotaxis (EC50 approximately equal to 1 nM) in leukocytes (THP-1, Jurkat, and peripheral blood lymphocyte cells) and activation of pertussis toxin-sensitive G proteins (EC50 approximately equal to 20 nM). TCS also significantly augmented chemokine-stimulated activation of chemokine receptors CCR5 and CXCR4 as well as CCR1, CCR2B, CCR3, and CCR4 transiently expressed in HEK293 cells. A mutant TCS with 4,000-fold lower ribosome-inactivating activity showed similar augmentation activity as wild-type TCS. Moreover, flow cytometry demonstrated that the specific association of TCS to the cell membranes required the presence of chemokine receptors, and laser confocal microscopy reveals that TCS was colocalized with chemokine receptors on the membranes. The results from TCS-Sepharose pull-down and TCS and chemokine receptor coimmunoprecipitation and cross-linking experiments demonstrated association of TCS with CCR5. Thus, our data clearly demonstrated that TCS synergizes activities of chemokines to stimulate chemotaxis and G protein activation, and the effects of TCS are likely to be mediated through its interaction with chemokine receptors.


Subject(s)
Anti-HIV Agents/pharmacology , Chemokines/pharmacology , Chemotaxis/drug effects , GTP-Binding Proteins/metabolism , Receptors, Chemokine/metabolism , Trichosanthin/pharmacology , Animals , Anti-HIV Agents/metabolism , Cell Line , Chemokine CCL5/pharmacology , Cloning, Molecular , Fluorescent Antibody Technique , GTP-Binding Proteins/drug effects , Humans , Mice , Rabbits , Receptors, CCR5/metabolism , Trichosanthin/metabolism
15.
J Exp Med ; 188(5): 941-52, 1998 Sep 07.
Article in English | MEDLINE | ID: mdl-9730895

ABSTRACT

The neisserial porin P.I is a GTP binding protein that forms a voltage-gated channel that translocates into mammalian cell membranes and modulates host cell signaling events. Here, we report that P.I confers invasion of the bacterial pathogen Neisseria gonorrhoeae into Chang epithelial cells and that this event is controlled by GTP, as well as other phosphorus-containing compounds. Bacterial invasion was observed only for strains carrying the P.IA subtype of porin, which is typically associated with the development of disseminated neisserial disease, and did not require opacity outer membrane proteins, previously recognized as gonococcal invasins. Allelic replacement studies showed that bacterial invasiveness cotransferred with the P.IA (por1A) gene. Mutation of the P.I-associated protein Rmp did not alter the invasive properties. Cross-linking of labeled GTP to the porin revealed more efficient GTP binding to the P.IA than P.IB porin subtype. GTP binding was inhibited by an excess of unlabeled GTP, ATP, and GDP, as well as inorganic phosphate, but not by UTP or beta-glycerophosphate, fully in line with the respective invasion-inhibitory activities observed for these compounds. The P.IA-mediated cellular invasion may explain the more invasive behavior of P.IA strains in the natural infection and may broaden the basis for the development of a P.I-based gonococcal vaccine.


Subject(s)
Antigens, Bacterial/physiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , GTP-Binding Proteins/immunology , Ion Channels/immunology , Neisseria gonorrhoeae/pathogenicity , Porins/metabolism , Alleles , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Cell Line , Epithelial Cells/drug effects , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Humans , Ion Channels/drug effects , Ion Channels/metabolism , Mutagenesis, Insertional , Neisseria gonorrhoeae/drug effects , Phosphates/pharmacology , Porins/drug effects , Porins/genetics
16.
J Pharmacol Exp Ther ; 333(2): 547-54, 2010 May.
Article in English | MEDLINE | ID: mdl-20160007

ABSTRACT

The effect of the plant-derived nonpsychotropic cannabinoid, cannabidiol (CBD), on the function of hydroxytryptamine (5-HT)3A receptors expressed in Xenopus laevis oocytes was investigated using two-electrode voltage-clamp techniques. CBD reversibly inhibited 5-HT (1 microM)-evoked currents in a concentration-dependent manner (IC50 = 0.6 microM). CBD (1 microM) did not alter specific binding of the 5-HT3A antagonist [3H]3-(5-methyl-1H-imidazol-4-yl)-1-(1-methylindol-3-yl)propan-1-one (GR65630), in oocytes expressing 5-HT3A receptors. In the presence of 1 microM CBD, the maximal 5-HT-induced currents were also inhibited. The EC50 values were 1.2 and 1.4 microM, in the absence and presence of CBD, indicating that CBD acts as a noncompetitive antagonist of 5-HT3 receptors. Neither intracellular BAPTA injection nor pertussis toxin pretreatment (5 microg/ml) altered the CBD-evoked inhibition of 5-HT-induced currents. CBD inhibition was inversely correlated with 5-HT3A expression levels and mean 5-HT3 receptor current density. Pretreatment with actinomycin D, which inhibits protein transcription, decreased the mean 5-HT3 receptor current density and increased the magnitude of CBD inhibition. These data demonstrate that CBD is an allosteric inhibitor of 5-HT3 receptors expressed in X. laevis oocytes. They further suggest that allosteric inhibition of 5-HT3 receptors by CBD may contribute to its physiological roles in the modulation of nociception and emesis.


Subject(s)
Cannabidiol/pharmacology , Membrane Potentials/drug effects , Oocytes/drug effects , Serotonin 5-HT3 Receptor Antagonists , Action Potentials/drug effects , Animals , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/physiology , Imidazoles/pharmacology , Indoles/pharmacology , Membrane Potentials/physiology , Oocytes/physiology , Pertussis Toxin/pharmacology , Receptors, Serotonin, 5-HT3/biosynthesis , Receptors, Serotonin, 5-HT3/drug effects , Serotonin/pharmacology , Signal Transduction/drug effects , Xenopus laevis
17.
Postepy Hig Med Dosw (Online) ; 64: 188-94, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20400782

ABSTRACT

BACKGROUND: Calcium ions regulate the function of cells in many ways, acting as first messengers of intercellular information and second messengers of intracellular information. Changes in cytoplasmic calcium levels depend on calcium influx from the extracellular space or calcium release from cellular stores. Increase in calcium ion concentration takes place in pathological situations, such as ischemia. In the present study the roles of calcium and G protein in contraction induced by angiotensin II (agonist of the metabotropic receptor AT1), phenylephrine (agonist of alpha1-adrenergic metabotropic receptor), and Bay K8644 (a calcium channel agonist) after ischemia/reperfusion were investigated. MATERIAL/METHODS: Experiments were performed on perfused male Wistar rats' tail arteries. Contraction induced by angiotensin II, phenylephrine, and Bay K8644 mediated by intracellular or extracellular calcium after ischemia/reperfusion and in the presence of the blocker of G protein Bordetella pertussis toxin (P 7208) was analyzed. RESULTS: Ischemia reduced while reperfusion augmented the response of vascular smooth muscle cells to angiotensin II and phenylephrine, but they did not change the effects of Bay K8644. P 7208 decreased the effects of phenylephrine mediated by intracellular and extracellular calcium and reduced the reactions of angiotensin II mediated only by intracellular calcium, but did not change the effects of Bay K8644. CONCLUSIONS: Ischemia/reperfusion modulates vascular contraction induced by angiotensin II and phenylephrine. Both intracellular and extracellular calcium ions mediate the contraction induced by angiotensin II and phenylephrine. The results suggests that G protein modulates the effects of angiotensin II mediated by intracellular calcium ions while it plays a role in the reactions of phenylephrine mediated by calcium coming from both sources, intracellular and extracellular.


Subject(s)
Calcium/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Reperfusion Injury/drug therapy , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Angiotensin II/pharmacology , Animals , GTP-Binding Proteins/drug effects , Male , Phenylephrine/pharmacology , Rats , Rats, Wistar
18.
Heart Fail Clin ; 6(1): 45-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19945060

ABSTRACT

Hypertension is a common condition that is a well-described risk factor for the development of cerebrovascular disease, myocardial infarction and heart failure. Medical therapy for hypertension may not only prevent development of these potentially devastating illnesses but may be used to treat high blood pressure, which coexists with these conditions. Therapeutic response to medical therapy for hypertension is variable and may be related to the individual genetic profile of patients. We have described here only several of the HTN-related polymorphisms that may impact clinical response in patients who have high blood pressure and coexisting conditions. Future studies are needed to identify other potential genotypes that may influence therapeutic response in addition to clinical trials of specific medical therapy in individual patients based on specific genotypes.


Subject(s)
Antihypertensive Agents/therapeutic use , Hypertension/drug therapy , AMP Deaminase/genetics , Chromogranins , Endothelins/genetics , Endothelins/physiology , Endothelium, Vascular/drug effects , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Hypertension/genetics , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Synthase/genetics , Polymorphism, Single Nucleotide , Receptors, Adrenergic, alpha/genetics , Receptors, Adrenergic, alpha/physiology , Regional Blood Flow/drug effects , Risk Factors , Signal Transduction/genetics , Vasodilation/drug effects
19.
Trends Biochem Sci ; 30(6): 318-24, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950876

ABSTRACT

Modulators of G-protein signaling have a central role in controlling cell physiology and represent over half of all marketed prescription drugs. G-protein pathways have traditionally been targeted by developing ligands to the extracellular surface of a small subset of the estimated approximately 1000 G-protein-coupled receptors in humans. The intracellular machinery, consisting of the cytosolic receptor surfaces and heterotrimeric G proteins, provides an equivalent diversity of targets that has remained relatively unexplored until now. This review summarizes recent efforts using combinatorial peptide libraries to develop new G-protein signaling modulators targeting intracellular components.


Subject(s)
Combinatorial Chemistry Techniques/methods , GTP-Binding Proteins/drug effects , Peptide Library , Peptides/pharmacology , Receptors, G-Protein-Coupled/drug effects , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Humans , Ligands , Peptides/chemistry , RNA, Messenger , Receptor Cross-Talk , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
20.
Neuropharmacology ; 181: 108356, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33069757

ABSTRACT

Endothelin-1 (ET-1), an endogenous vasoconstrictor, has been known as a pro-nociceptive agent involved in multitude of pain. ET-1 acts on endothelin receptors on vascular endothelial cells, sensitizes release of ATP, which then acts on P2X3 receptors on nociceptors and results in mechanical hyperalgesia. Both endothelin receptors and P2X3 receptors are present in primary sensory neuron, where it remains unclear whether there is an interaction between them. Herein, we reported that ET-1 potentiated the electrophysiological activity of P2X3 receptors in rat dorsal root ganglia (DRG) neurons. ET-1 concentration-dependently increased α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents, which were mediated by P2X3 receptors. ET-1 shifted the α,ß-meATP concentration-response curve upwards, with an increase of 34.38 ± 4.72% in the maximal current response to α,ß-meATP in the presence of ET-1. ET-1 potentiation of α,ß-meATP-evoked currents was voltage-independent. ET-1 potentiated P2X3 receptor-mediated currents through endothelin-A receptors (ETAR), but not endothelin-B receptors (ETBR). ET-1 potentiation was supressed by blockade of intracellular G-protein or protein kinase C (PKC) signaling. Moreover, there is a synergistic effect on mechanical allodynia induced by intraplantar injection of ET-1 and α,ß-meATP in rats. Pharmacological blockade of P2X3 receptors also alleviated ET-1-induced mechanical allodynia. These results suggested that ET-1 sensitized P2X3 receptors in primary sensory neurons via an ETAR and PKC signaling pathway. Our data provide evidence that cutaneous ET-1 induced mechanical allodynia not only by increasing the release of ATP from vascular endothelial cells, but also by sensitizing P2X3 receptors on nociceptive DRG neurons.


Subject(s)
Endothelin-1/pharmacology , Ganglia, Spinal/drug effects , Neurons/drug effects , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Purinergic P2X3/drug effects , Adenosine Triphosphate/analogs & derivatives , Animals , Behavior, Animal , Electrophysiological Phenomena , GTP-Binding Proteins/drug effects , Ganglia, Spinal/cytology , Hyperalgesia/chemically induced , Male , Protein Kinase C/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A/drug effects , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL