Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732124

ABSTRACT

Oxytocin, a significant pleiotropic neuropeptide, regulates psychological stress adaptation and social communication, as well as peripheral actions, such as uterine contraction and milk ejection. Recently, a Japanese Kampo medicine called Kamikihito (KKT) has been reported to stimulate oxytocin neurons to induce oxytocin secretion. Two-pore-domain potassium channels (K2P) regulate the resting potential of excitable cells, and their inhibition results in accelerated depolarization that elicits neuronal and endocrine cell activation. We assessed the effects of KKT and 14 of its components on a specific K2P, the potassium channel subfamily K member 2 (TREK-1), which is predominantly expressed in oxytocin neurons in the central nervous system (CNS). KKT inhibited the activity of TREK-1 induced via the channel activator ML335. Six of the 14 components of KKT inhibited TREK-1 activity. Additionally, we identified that 22 of the 41 compounds in the six components exhibited TREK-1 inhibitory effects. In summary, several compounds included in KKT partially activated oxytocin neurons by inhibiting TREK-1. The pharmacological effects of KKT, including antistress effects, may be partially mediated through the oxytocin pathway.


Subject(s)
Neurons , Oxytocin , Potassium Channels, Tandem Pore Domain , Animals , Humans , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Medicine, Kampo , Neurons/metabolism , Neurons/drug effects , Oxytocin/pharmacology , Oxytocin/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors
2.
Molecules ; 28(19)2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37836768

ABSTRACT

In medicinal chemistry, the copper-catalyzed click reaction is used to prepare ligand candidates. This reaction is so clean that the bioactivities of the products can be determined without purification. Despite the advantages of this in situ screening protocol, the applicability of this method for transmembrane proteins has not been validated due to the incompatibility with copper catalysts. To address this point, we performed ligand screening for the µ, δ, and κ opioid receptors using this protocol. As we had previously reported the 7-azanorbornane skeleton as a privileged scaffold for the G protein-coupled receptors, we performed the click reactions between various 7-substituted 2-ethynyl-7-azanorbornanes and azides. Screening assays were performed without purification using the CellKeyTM system, and the putative hit compounds were re-synthesized and re-evaluated. Although the "hit" compounds for the µ and the δ receptors were totally inactive after purifications, three of the four "hits" for the κ receptor were true agonists for this receptor and also showed activities for the δ receptor. Although false positive/negative results exist as in other screening projects for soluble proteins, this in situ method is effective in identifying novel ligands for transmembrane proteins.


Subject(s)
Copper , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Ligands , Membrane Proteins , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/chemistry
3.
Int J Mol Sci ; 24(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37373107

ABSTRACT

Rubiscolins are naturally occurring opioid peptides derived from the enzymatic digestion of the ribulose bisphosphate carboxylase/oxygenase protein in spinach leaves. They are classified into two subtypes based on amino acid sequence, namely rubiscolin-5 and rubiscolin-6. In vitro studies have determined rubiscolins as G protein-biased delta-opioid receptor agonists, and in vivo studies have demonstrated that they exert several beneficial effects via the central nervous system. The most unique and attractive advantage of rubiscolin-6 over other oligopeptides is its oral availability. Therefore, it can be considered a promising candidate for the development of a novel and safe drug. In this review, we show the therapeutic potential of rubiscolin-6, mainly focusing on its effects when orally administered based on available evidence. Additionally, we present a hypothesis for the pharmacokinetics of rubiscolin-6, focusing on its absorption in the intestinal tract and ability to cross the blood-brain barrier.


Subject(s)
Receptors, Opioid, delta , Ribulose-Bisphosphate Carboxylase , Ribulose-Bisphosphate Carboxylase/metabolism , Receptors, Opioid, delta/metabolism , Oligopeptides , Opioid Peptides
4.
Int J Mol Sci ; 24(9)2023 May 06.
Article in English | MEDLINE | ID: mdl-37176075

ABSTRACT

Remifentanil (REM) and fentanyl (FEN) are commonly used analgesics that act by activating a µ-opioid receptor (MOR). Although optimal concentrations of REM can be easily maintained during surgery, it is sometimes switched to FEN for optimal pain regulation. However, standards for this switching protocol remain unclear. Opioid anesthetic efficacy is decided in part by MOR desensitization; thus, in this study, we investigated the desensitization profiles of REM and FEN to MOR. The efficacy and potency during the 1st administration of REM or FEN in activating the MOR were almost equal. Similarly, in ß arrestin recruitment, which determines desensitization processes, they showed no significant differences. In contrast, the 2nd administration of FEN resulted in a stronger MOR desensitization potency than that of REM, whereas REM showed a higher internalization potency than FEN. These results suggest that different ß arrestin-mediated signaling caused by FEN or REM led to their distinct desensitization and internalization processes. Our three-dimensional analysis, with in silico binding of REM and FEN to MOR models, highlighted that REM and FEN bound to similar but distinct sites of MOR and led to distinct ß arrestin-mediated profiles, suggesting that distinct binding profiles to MOR may alter ß arrestin activity, which accounts for MOR desensitization and internalization.


Subject(s)
Fentanyl , Receptors, Opioid , Receptors, Opioid/metabolism , Fentanyl/pharmacology , Remifentanil/pharmacology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , beta-Arrestins/metabolism , Morphine
5.
Peptides ; 159: 170901, 2023 01.
Article in English | MEDLINE | ID: mdl-36347314

ABSTRACT

Positive allosteric modulators (PAMs) of G protein-coupled receptors (GPCRs) have drawn attention as novel drug candidates. PAMs can enhance the activities of endogenous agonists which are not only secreted at appropriate times and in parts of the body, but also are immediately metabolized. Therefore, they are expected to show fewer side effects than exogeneous orthosteric ligands. Recently, we have reported that oxytocin (OT) functioned as a PAM of the µ opioid receptor (MOR) which was one of the most potent targets for analgesics. OT is thus thought to be a useful compound for the development of novel analgesics. In this study, several OT analogs were synthesized and evaluated with an intact cell-based assay to investigate the crucial structures of OT for exerting the PAM activity. The assay results indicated that the cyclic structure formed by an intramolecular disulfide bond and the three C-terminal residues containing a small Gly residue of OT were essential for their function as a MOR-PAM. Intriguingly, two analogs having an amide or an ethylene tether instead of the intramolecular disulfide bridge did not have any PAM effects. The results suggested that the disulfide linkage of OT would be a key structure for exerting the PAM activity at the MOR.


Subject(s)
Oxytocin , Receptors, Opioid , Allosteric Regulation , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship , Analgesics
6.
Molecules ; 27(20)2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36296658

ABSTRACT

Opioid receptors (ORs) are classified into three types (µ, δ, and κ), and opioid analgesics are mainly mediated by µOR activation; however, their use is sometimes restricted by unfavorable effects. The selective κOR agonist nalfurafine was initially developed as an analgesic, but its indication was changed because of the narrow safety margin. The activation of ORs mainly induces two intracellular signaling pathways: a G-protein-mediated pathway and a ß-arrestin-mediated pathway. Recently, the expectations for κOR analgesics that selectively activate these pathways have increased; however, the structural properties required for the selectivity of nalfurafine are still unknown. Therefore, we evaluated the partial structures of nalfurafine that are necessary for the selectivity of these two pathways. We assayed the properties of nalfurafine and six nalfurafine analogs (SYKs) using cells stably expressing κORs. The SYKs activated κORs in a concentration-dependent manner with higher EC50 values than nalfurafine. Upon bias factor assessment, only SYK-309 (possessing the 3S-hydroxy group) showed higher selectivity of G-protein-mediated signaling activities than nalfurafine, suggesting the direction of the 3S-hydroxy group may affect the ß-arrestin-mediated pathway. In conclusion, nalfurafine analogs having a 3S-hydroxy group, such as SYK-309, could be considered G-protein-biased κOR agonists.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, kappa , Analgesics , Analgesics, Opioid/pharmacology , beta-Arrestins/metabolism , GTP-Binding Proteins/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/metabolism
7.
Anesth Analg ; 134(5): 1082-1093, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35427270

ABSTRACT

BACKGROUND: Transdermal fentanyl is widely used in the treatment of severe pain because of convenience, safety, and stable blood concentrations. Nevertheless, patients often develop tolerance to fentanyl, necessitating the use of other opioids; transdermal buprenorphine patch is widely used as an analgesic agent, though available formulation does not provide comparable analgesic effect as transdermal fentanyl patch. Opioids bind to the opioid receptor (OR) to activate both G protein-mediated and ß-arrestin-mediated pathways. We synthesized morphine-related compounds with high transdermal absorbability (N1 and N2) and evaluated their OR activities pharmacologically in comparison with fentanyl and morphine. METHODS: In cells stably expressing µ-opioid receptor (MOR), δ-opioid receptor (DOR), and κ-opioid receptor (KOR), G protein-mediated pathways were assessed using the CellKey and an intracellular cyclic adenosine monophosphate (cAMP) assay, while ß-arrestin-mediated pathways were analyzed with ß-arrestin recruitment and receptor internalization assays. Furthermore, analgesic effects were evaluated using a tail-flick test in mice, and the analgesic effect on fentanyl-tolerant mice was evaluated. RESULTS: In the CellKey and cAMP assays, both N1 and N2 showed the highest affinity for MOR and acted as full agonists as well as partial agonists for DOR and KOR. In the ß-arrestin and internalization assays, only fentanyl acted as a full agonist; N1 and N2 acted as partial agonists of MOR. In the mouse tail-flick test, N1 and N2 showed analgesic effects equivalent to those of fentanyl and morphine. In fentanyl-tolerant mice, fentanyl showed a diminished analgesic effect, whereas N1 and N2 as well as morphine retained their analgesic effects. CONCLUSIONS: While N1 and N2 have higher transdermal absorbability than fentanyl, they also have analgesic effects comparable to those of morphine, suggesting that they may be attractive compounds for the development of novel opioid patches for transitioning from fentanyl patches.


Subject(s)
Fentanyl , Morphine , Analgesics, Opioid , Animals , GTP-Binding Proteins/metabolism , Humans , Mice , Receptors, Opioid/metabolism , Receptors, Opioid, mu/agonists , beta-Arrestins/metabolism
8.
Nutrients ; 14(5)2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35267998

ABSTRACT

The steroidal alkaloid tomatidine is an aglycone of α-tomatine, which is abundant in tomato leaves and has several biological activities. Tomatidine has been reported to inhibit the growth of cultured cancer cells in vitro, but its anti-cancer activity in vivo and inhibitory effect against gastric cancer cells remain unknown. We investigated the efficacy of tomatidine using human gastric cancer-derived 85As2 cells and its tumor-bearing mouse model and evaluated the effect of tomatidine-rich tomato leaf extract (TRTLE) obtained from tomato leaves. In the tumor-bearing mouse model, tumor growth was significantly inhibited by feeding a diet containing tomatidine and TRTLE for 3 weeks. Tomatidine and TRTLE also inhibited the proliferation of cultured 85As2 cells. Microarray data of gene expression analysis in mouse tumors revealed that the expression levels of mRNAs belonging to the type I interferon signaling pathway were altered in the mice fed the diet containing tomatidine and TRTLE. Moreover, the knockdown of one of the type I interferon-stimulated genes (ISGs), interferon α-inducible protein 27 (IFI27), inhibited the proliferation of cultured 85As2 cells. This study demonstrates that tomatidine and TRTLE inhibit the tumor growth in vivo and the proliferation of human gastric cancer-derived 85As2 cells in vitro, which could be due to the downregulation of ISG expression.


Subject(s)
Alkaloids , Solanum lycopersicum , Stomach Neoplasms , Alkaloids/metabolism , Alkaloids/pharmacology , Animals , Humans , Interferons , Mice , Plant Extracts/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Tomatine/analogs & derivatives
9.
Biomolecules ; 12(3)2022 03 10.
Article in English | MEDLINE | ID: mdl-35327617

ABSTRACT

The issue of tolerance to continuous or repeated administration of opioids should be addressed. The ability of ketamine to improve opioid tolerance has been reported in clinical studies, and its mechanism of tolerance may involve improved desensitization of µ-opioid receptors (MORs). We measured changes in MOR activity and intracellular signaling induced by repeated fentanyl and morphine administration and investigated the effects of ketamine on these changes with human embryonic kidney 293 cells expressing MOR using the CellKey™, cADDis cyclic adenosine monophosphate, and PathHunter® ß-arrestin recruitment assays. Repeated administration of fentanyl or morphine suppressed the second MOR responses. Administration of ketamine before a second application of opioids within clinical concentrations improved acute desensitization and enhanced ß-arrestin recruitment elicited by fentanyl but not by morphine. The effects of ketamine on fentanyl were suppressed by co-treatment with an inhibitor of G-protein-coupled receptor kinase (GRK). Ketamine may potentially reduce fentanyl tolerance but not that of morphine through modulation of GRK-mediated pathways, possibly changing the conformational changes of ß-arrestin to MOR.


Subject(s)
Ketamine , Morphine , Analgesics, Opioid/pharmacology , Drug Tolerance , Fentanyl/pharmacology , Humans , Ketamine/pharmacology , Morphine/pharmacology , Receptors, Opioid/metabolism , beta-Arrestins/metabolism
10.
PLoS One ; 17(2): e0262892, 2022.
Article in English | MEDLINE | ID: mdl-35157707

ABSTRACT

Mesenchymal stem cells (MSCs), which are isolated from adipose tissue (AD-MSCs), umbilical cord (UC-MSCs), or bone marrow, have therapeutic potential including anti-inflammatory and immunomodulatory activities. It was recently reported that MSCs are also effective as a therapeutic treatment for neuropathic pain, although the underlying mechanisms have yet to be resolved. Therefore, in this study, we investigated the effects of human AD- and UC-MSCs on neuropathic pain and its mechanisms using rat models of partial sciatic nerve ligation (PSNL). AD- or UC-MSCs were intravenously administered 4 days after PSNL. Antinociceptive effects were then evaluated using the von Frey and weight-bearing tests. We found that, 3-9 days after the administration of AD- or UC-MSCs to PSNL-exposed rats, both the mechanical threshold and differences in weight-bearing of the right and left hind paws were significantly improved. To reveal the potential underlying antinociceptive mechanisms of MSCs, the levels of activation transcription factor 3- and ionized calcium-binding adapter molecule 1-positive cells were measured by immunohistochemical analysis. AD- and UC-MSCs significantly decreased the levels of these proteins that were induced by PSNL in the dorsal root ganglia. Additionally, UC-MSC significantly improved the PSNL-induced decrease in the myelin basic protein level in the sciatic nerve, indicating that UC-MSC reversed demyelination of the sciatic nerve produced by PSNL. These data suggest that AD- and UC-MSCs may help in the recovery of neuropathic pain via the different regulation; AD-MSCs exhibited their effects via suppressed neuronal damage and anti-inflammatory actions, while UC-MSCs exhibited their effects via suppressed neuronal damage, anti-inflammatory actions and remyelination.


Subject(s)
Mesenchymal Stem Cell Transplantation , Neuralgia/therapy , Neurons/metabolism , Activating Transcription Factor 3/metabolism , Adipose Tissue/cytology , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microfilament Proteins/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sciatic Nerve/surgery , Umbilical Cord/cytology
11.
Circ Res ; 130(2): 234-248, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34875852

ABSTRACT

BACKGROUND: During the development of heart failure, a fetal cardiac gene program is reactivated and accelerates pathological cardiac remodeling. We previously reported that a transcriptional repressor, NRSF (neuron restrictive silencer factor), suppresses the fetal cardiac gene program, thereby maintaining cardiac integrity. The underlying molecular mechanisms remain to be determined, however. METHODS: We aim to elucidate molecular mechanisms by which NRSF maintains normal cardiac function. We generated cardiac-specific NRSF knockout mice and analyzed cardiac gene expression profiles in those mice and mice cardiac-specifically expressing a dominant-negative NRSF mutant. RESULTS: We found that cardiac expression of Gαo, an inhibitory G protein encoded in humans by GNAO1, is transcriptionally regulated by NRSF and is increased in the ventricles of several mouse models of heart failure. Genetic knockdown of Gnao1 ameliorated the cardiac dysfunction and prolonged survival rates in these mouse heart failure models. Conversely, cardiac-specific overexpression of GNAO1 in mice was sufficient to induce cardiac dysfunction. Mechanistically, we observed that increasing Gαo expression increased surface sarcolemmal L-type Ca2+ channel activity, activated CaMKII (calcium/calmodulin-dependent kinase-II) signaling, and impaired Ca2+ handling in ventricular myocytes, which led to cardiac dysfunction. CONCLUSIONS: These findings shed light on a novel function of Gαo in the regulation of cardiac Ca2+ homeostasis and systolic function and suggest Gαo may be an effective therapeutic target for the treatment of heart failure.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Repressor Proteins/metabolism , Animals , Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Heart Ventricles/cytology , Heart Ventricles/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Repressor Proteins/genetics
12.
Cells ; 10(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34685631

ABSTRACT

Oxytocin (OT) influences various physiological functions such as uterine contractions, maternal/social behavior, and analgesia. Opioid signaling pathways are involved in one of the analgesic mechanisms of OT. We previously showed that OT acts as a positive allosteric modulator (PAM) and enhances µ-opioid receptor (MOR) activity. In this study, which focused on other opioid receptor (OR) subtypes, we investigated whether OT influences opioid signaling pathways as a PAM for δ-OR (DOR) or κ-OR (KOR) using human embryonic kidney-293 cells expressing human DOR or KOR, respectively. The CellKeyTM results showed that OT enhanced impedance induced by endogenous/exogenous KOR agonists on KOR-expressing cells. OT did not affect DOR activity induced by endogenous/exogenous DOR agonists. OT potentiated the KOR agonist-induced Gi/o protein-mediated decrease in intracellular cAMP, but did not affect the increase in KOR internalization caused by the KOR agonists dynorphin A and (-)-U-50488 hydrochloride (U50488). OT did not bind to KOR orthosteric binding sites and did not affect the binding affinities of dynorphin A and U50488 for KOR. These results suggest that OT is a PAM of KOR and MOR and enhances G protein signaling without affecting ß-arrestin signaling. Thus, OT has potential as a specific signaling-biased PAM of KOR.


Subject(s)
GTP-Binding Proteins/metabolism , Oxytocin/pharmacology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Signal Transduction , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Allosteric Regulation/drug effects , Animals , Binding Sites , CHO Cells , Cricetulus , Cyclic AMP/metabolism , Diprenorphine/pharmacology , Dynorphins/pharmacology , Electric Impedance , Endocytosis/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Signal Transduction/drug effects
13.
Molecules ; 26(19)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34641621

ABSTRACT

Activated opioid receptors transmit internal signals through two major pathways: the G-protein-mediated pathway, which exerts analgesia, and the ß-arrestin-mediated pathway, which leads to unfavorable side effects. Hence, G-protein-biased opioid agonists are preferable as opioid analgesics. Rubiscolins, the spinach-derived naturally occurring opioid peptides, are selective δ opioid receptor agonists, and their p.o. administration exhibits antinociceptive effects. Although the potency and effect of rubiscolins as G-protein-biased molecules are partially confirmed, their in vitro profiles remain unclear. We, therefore, evaluated the properties of rubiscolins, in detail, through several analyses, including the CellKeyTM assay, cADDis® cAMP assay, and PathHunter® ß-arrestin recruitment assay, using cells stably expressing µ, δ, κ, or µ/δ heteromer opioid receptors. In the CellKeyTM assay, rubiscolins showed selective agonistic effects for δ opioid receptor and little agonistic or antagonistic effects for µ and κ opioid receptors. Furthermore, rubiscolins were found to be G-protein-biased δ opioid receptor agonists based on the results obtained in cADDis® cAMP and PathHunter® ß-arrestin recruitment assays. Finally, we found, for the first time, that they are also partially agonistic for the µ/δ dimers. In conclusion, rubiscolins could serve as attractive seeds, as δ opioid receptor-specific agonists, for the development of novel opioid analgesics with reduced side effects.


Subject(s)
Opioid Peptides/pharmacology , Receptors, Opioid, delta/agonists , Signal Transduction/drug effects , Spinacia oleracea/chemistry , GTP-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Molecular Structure , Opioid Peptides/chemistry , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Receptors, Opioid, mu/metabolism , Ribulose-Bisphosphate Carboxylase/chemistry , Ribulose-Bisphosphate Carboxylase/pharmacology , beta-Arrestins/metabolism
14.
Biomed Pharmacother ; 141: 111800, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34175819

ABSTRACT

BACKGROUND: The misuse of opioids has led to an epidemic in recent times. The endothelin A receptor (ETAR) has recently attracted attention as a novel therapeutic target to enhance opioid analgesia. We hypothesized that endothelin A receptors may affect pain mechanisms by heterodimerization with µ opioid receptors. We examined the mechanisms of ETAR-mediated pain and the potential therapeutic effects of an ETAR antagonist, Compound-E, as an agent for analgesia. METHODS: Real-time in vitro effect of Compound-E on morphine response was assessed in HEK293 cells expressing both endothelin A and µ opioid receptors through CellKey™ and cADDis cAMP assays. Endothelin A/µ opioid receptor dimerization was assessed by immunoprecipitation and live cell imaging. The in vivo effect of Compound-E was evaluated using a morphine analgesia mouse model that observed escape response behavior, body temperature, and locomotor activity. RESULTS: In CellKey™ and cAMP assays, pretreatment of cells with endothelin-1 attenuated morphine-induced responses. These responses were improved by Compound-E, but not by BQ-123 nor by bosentan, an ETAR and endothelin B receptor antagonist. Dimerization of ETARs and µ opioid receptors was confirmed by Western blot and total internal reflection fluorescence microscopy in live cells. In vivo, Compound-E potentiated and prolonged the analgesic effects of morphine, enhanced hypothermia, and increased locomotor activity compared to morphine alone. CONCLUSION: The results suggest that attenuation by endothelin-1 of morphine analgesia may be caused by dimerization of Endothelin A/µ opioid receptors. The novel ETAR antagonist Compound-E could be an effective adjunct to reduce opioid use.


Subject(s)
Analgesics, Opioid/administration & dosage , Endothelin A Receptor Antagonists/administration & dosage , Morphine/administration & dosage , Protein Multimerization/physiology , Receptor, Endothelin A/metabolism , Receptors, Opioid, mu/metabolism , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Pain Measurement/drug effects , Pain Measurement/methods , Peptides, Cyclic/administration & dosage , Protein Multimerization/drug effects
15.
Biochem Pharmacol ; 190: 114599, 2021 08.
Article in English | MEDLINE | ID: mdl-33989656

ABSTRACT

With the development of new drugs, such as molecular-targeted drugs, and multidisciplinary therapies, cancer treatment outcomes have improved, and the number of cancer survivors is increasing every year. However, some chemotherapeutic agents cause cardiovascular complications (cancer treatment-related cardiovascular disease, CTRCVD), which affect the life prognosis and quality of life (QOL) of cancer patients. Therefore, it is necessary to select treatment methods that take into account the prognosis and QOL of cancer patients, and to take measures against CTRCVD. The mechanism of cardiotoxicity of high-risk drugs, such as doxorubicin and HER2 inhibitors, are still unclear; genetic factors, and cardiovascular disease risk factors (e.g., hypertension, dyslipidemia, and diabetes) are associated with CTRCVD progression. The establishment of methods for prevention, early diagnosis, and treatment of CTRCVD and the generation of evidence for these methods are needed. It is also necessary to develop screening methods for chemotherapy cardiotoxicity. In this review, we discuss the current status of CTRCVD, its complications, and expected countermeasures.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiovascular Diseases/chemically induced , Drug Screening Assays, Antitumor/trends , Neoplasms/drug therapy , Research/trends , Animals , Antineoplastic Agents/therapeutic use , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Drug Screening Assays, Antitumor/methods , Forecasting , Humans , Neoplasms/epidemiology
16.
Front Pharmacol ; 12: 695039, 2021.
Article in English | MEDLINE | ID: mdl-35145397

ABSTRACT

Several clinical studies have reported that Japanese herbal medicine Hangeshashinto (HST) has beneficial effects on chemotherapy-induced oral ulcerative mucositis (OUM). Our previous research demonstrated that HST improves chemotherapy-induced OUM through human oral keratinocyte (HOK) migration, which was suppressed by mitogen-activated protein kinase (MAPK) and C-X-C chemokine receptor 4 (CXCR4) inhibitors. However, the association between these molecules and HOK migration was unclear. Here, we examined the effects of HST on the expression of CXCR4/CXCR7 and C-X-C motif chemokine ligands 11 and 12 (CXCL11/CXCL12) in HOKs. Our results indicated that HST upregulated CXCL12, but not CXCR4, CXCR7, nor CXCL11 in HOKs. HST-induced expression of CXCL12 was significantly suppressed by an inhibitor of extracellular signal-regulated kinase (ERK), but not of p38 and c-Jun N-terminal kinase (JNK). In addition, HST induced phosphorylation of ERK in HOKs. These findings suggest that HST enhances HOK migration by upregulating CXCL12 via ERK.

18.
Curr Top Med Chem ; 20(31): 2822-2829, 2020.
Article in English | MEDLINE | ID: mdl-33115393

ABSTRACT

Opioid agonists elicit their analgesic action mainly via µ opioid receptors; however, their use is limited because of adverse events including constipation and respiratory depression. It has been shown that analgesic action is transduced by the G protein-mediated pathway whereas adverse events are by the ß-arrestin-mediated pathway through µ opioid receptor signaling. The first new-generation opioid TRV130, which preferentially activates G protein- but not ß-arrestin-mediated signal, was constructed and developed to reduce adverse events. TRV130 and other G protein-biased compounds tend to elicit desirable analgesic action with less adverse effects. In clinical trials, the intravenous TRV130 (oliceridine) was evaluated in Phase I, II and III clinical studies. Here we review the discovery and synthesis of TRV130, its main action as a novel analgesic having less adverse events, its up-to-date status in clinical trials, and additional concerns about TRV130 as demonstrated in the literature.


Subject(s)
Analgesics, Opioid/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Spiro Compounds/pharmacology , Thiophenes/pharmacology , Analgesics, Opioid/adverse effects , Analgesics, Opioid/chemistry , GTP-Binding Proteins/metabolism , Humans , Signal Transduction/drug effects , Spiro Compounds/adverse effects , Spiro Compounds/chemistry , Thiophenes/adverse effects , Thiophenes/chemistry
19.
J Pharmacol Sci ; 143(4): 320-324, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32505645

ABSTRACT

Cellular dielectric spectroscopy (CDS) is a novel technology enabling pharmacological evaluation of multiple receptor types with a label-free cell-based assay. We evaluated activities of a family of ligand-gated channels, transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1) channels by an electrical impedance-based biosensor (CellKey™ system) using CDS. Measures of both potency (EC50) and efficacy (Emax) of these agonists with CellKey™ were almost identical to those made using the traditional Ca2+ influx assay in TRPV1- or TRPA1-expressing cells, suggesting that CellKey™ is a simpler and easier means of evaluating TRP activities.


Subject(s)
Dielectric Spectroscopy/methods , Transient Receptor Potential Channels/metabolism , HEK293 Cells , Humans , TRPA1 Cation Channel , TRPV Cation Channels
20.
Nihon Yakurigaku Zasshi ; 155(3): 165-170, 2020.
Article in Japanese | MEDLINE | ID: mdl-32378637

ABSTRACT

Cardiovascular disorders in cancer patients with cachexia have recently become a great concern. However, the relationship between cancer cachexia and cardiac dysfunction remains unclear, due to lack of suitable models. We established a novel murine model of cancer cachexia by implantation of 85As2 cells, a cell line derived from human gastric cancer cells, presenting anorexia, weight loss and low fat-free mass similar to those observed in patients. Moreover, cardiac dysfunction is expected in this model, which has not been yet examined. In the present study, we firstly evaluated cardiac functions with the model. Secondly, we investigated effects of voluntary wheel running (VWR) on cachexia-induced cardiac dysfunction using this model, as the exercise is considered to be one of therapies for chronic heart failure. 85As2 cells were transplanted subcutaneously into mice, which observed a symptomatic cachexia; decrease in body, skeletal muscle weight, and food intake. In addition, this cachexia mouse developed severe cardiac atrophy and left ventricular ejection fraction (LVEF) also markedly reduced with cachexia progression. Moreover, VWR suppressed the decrease in food intake and skeletal muscle weight loss in this model, and improved LVEF with suppression of heart weight loss. These results imply that our 85As2-cachexia mice models show cardiac dysfunction and VWR may improve not only cachexia symptoms but also cardiac dysfunction. As exercise therapy is generally introduced for the purpose of improving heart failure symptoms, this study suggests a possible therapeutic effect of exercise on cardiac dysfunction induced by cancer cachexia.


Subject(s)
Cachexia/complications , Heart Diseases/etiology , Neoplasms/complications , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Motor Activity , Muscle, Skeletal , Myocardium/pathology , Stroke Volume , Ventricular Function, Left
SELECTION OF CITATIONS
SEARCH DETAIL
...