Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 866
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 294(41): 14896-14910, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31413114

ABSTRACT

The metabolic consequences and sequelae of obesity promote life-threatening morbidities. PKCƎĀ“I is an important elicitor of inflammation and apoptosis in adipocytes. Here we report increased PKCƎĀ“I activation via release of its catalytic domain concurrent with increased expression of proinflammatory cytokines in adipocytes from obese individuals. Using a screening strategy of dual recognition of PKCƎĀ“I isozymes and a caspase-3 binding site on the PKCƎĀ“I hinge domain with Schrƶdinger software and molecular dynamics simulations, we identified NP627, an organic small-molecule inhibitor of PKCƎĀ“I. Characterization of NP627 by surface plasmon resonance (SPR) revealed that PKCƎĀ“I and NP627 interact with each other with high affinity and specificity, SPR kinetics revealed that NP627 disrupts caspase-3 binding to PKCƎĀ“I, and in vitro kinase assays demonstrated that NP627 specifically inhibits PKCƎĀ“I activity. The SPR results also indicated that NP627 affects macromolecular interactions between protein surfaces. Of note, release of the PKCƎĀ“I catalytic fragment was sufficient to induce apoptosis and inflammation in adipocytes. NP627 treatment of adipocytes from obese individuals significantly inhibited PKCƎĀ“I catalytic fragment release, decreased inflammation and apoptosis, and significantly improved mitochondrial metabolism. These results indicate that PKCƎĀ“I is a robust candidate for targeted interventions to manage obesity-associated chronic inflammatory diseases. We propose that NP627 may also be used in other biological systems to better understand the impact of caspase-3-mediated activation of kinase activity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Obesity/pathology , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Adipocytes/pathology , Adipose Tissue/pathology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Respiration/drug effects , Humans , Obesity/metabolism , Protein Kinase C-delta/metabolism , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacology
2.
J Stroke Cerebrovasc Dis ; 28(4): 988-993, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30660483

ABSTRACT

OBJECTIVE: The efficacy of thyrotropin-releasing hormone tartrate (TRH-T) for treating prolonged disturbance of consciousness due to aneurysmal subarachnoid hemorrhage (SAH) remains unclear. The purpose of the present study was to determine whether TRH-T was really effective, and what was the recovery factor when it was valid. This was a retrospective study of a single facility. METHODS: We treated 208 patients with aneurysmal SAH at our hospital between 2011 and 2017. Among them, we investigated 97 cases in which TRH-T was administered to prolonged disturbance of consciousness. Thirty one patients with Hasegawa dementia rating scale-revised (HDS-R) score less than 20 were included. Patients' HDS-R scores were evaluated 7 days after clipping the aneurysm and 2 days after completing a course of TRH-T treatment. HDS-R score increases of greater than or over equal to 8 and less than 8 were defined as good and poor outcomes, respectively. Outcomes were compared to 11 patients who did not receive TRH-T treatment. RESULTS: Average initial and post-treatment HDS-R scores were 9 Ā± 6.6 and 19 Ā± 9.5, respectively. The good outcome group included 19 patients. Statistically significant differences in HDS-R score changes were observed between the group with initial HDS-R scores of 0-4 and the other groups. Poor outcomes were significantly correlated with age of greater than 60 years and initial HDS-R scores less than oroverequal to 4 points. The improvement in HDS-R score was significantly greater in the TRH-T administration group than the control group. CONCLUSIONS: TRH-T was effective for treating prolonged disturbance of consciousness due to aneurysmal SAH, especially in young patients with HDS-R scores between 5 and 20.


Subject(s)
Consciousness Disorders/drug therapy , Consciousness/drug effects , Subarachnoid Hemorrhage/drug therapy , Thyrotropin-Releasing Hormone/therapeutic use , Adult , Aged , Consciousness Disorders/diagnosis , Consciousness Disorders/etiology , Consciousness Disorders/physiopathology , Female , Humans , Male , Middle Aged , Recovery of Function , Retrospective Studies , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/diagnosis , Subarachnoid Hemorrhage/physiopathology , Thyrotropin-Releasing Hormone/analogs & derivatives , Time Factors , Treatment Outcome
3.
J Pharmacol Exp Ther ; 366(1): 105-112, 2018 07.
Article in English | MEDLINE | ID: mdl-29674333

ABSTRACT

Thyrotropin releasing hormone (TRH) is a tripeptide hormone and a neurotransmitter widely expressed in the central nervous system that regulates thyroid function and maintains physiologic homeostasis. Following injection in rodents, TRH has multiple effects including increased blood pressure and breathing. We tested the hypothesis that TRH and its long-acting analog, taltirelin, will reverse morphine-induced respiratory depression in anesthetized rats following intravenous or intratracheal (IT) administration. TRH (1 mg/kg plus 5 mg/kg/h, i.v.) and talitrelin (1 mg/kg, i.v.), when administered to rats pretreated with morphine (5 mg/kg, i.v.), increased ventilation from 50% Ā± 6% to 131% Ā± 7% and 45% Ā± 6% to 168% Ā± 13%, respectively (percent baseline; n = 4 Ā± S.E.M.), primarily through increased breathing rates (from 76% Ā± 9% to 260% Ā± 14% and 66% Ā± 8% to 318% Ā± 37%, respectively). By arterial blood gas analysis, morphine caused a hypoxemic respiratory acidosis with decreased oxygen and increased carbon dioxide pressures. TRH decreased morphine effects on arterial carbon dioxide pressure, but failed to impact oxygenation; taltirelin reversed morphine effects on both arterial carbon dioxide and oxygen. Both TRH and talirelin increased mean arterial blood pressure in morphine-treated rats (from 68% Ā± 5% to 126% Ā± 12% and 64% Ā± 7% to 116% Ā± 8%, respectively; n = 3 to 4). TRH, when initiated prior to morphine (15 mg/kg, i.v.), prevented morphine-induced changes in ventilation; and TRH (2 mg/kg, i.v.) rescued all four rats treated with a lethal dose of morphine (5 mg/kg/min, until apnea). Similar to intravenous administration, both TRH (5 mg/kg, IT) and taltirelin (2 mg/kg, IT) reversed morphine effects on ventilation. TRH or taltirelin may have clinical utility as an intravenous or inhaled agent to antagonize opioid-induced cardiorespiratory depression.


Subject(s)
Analgesics, Opioid/adverse effects , Isoflurane/pharmacology , Respiratory Insufficiency/drug therapy , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/pharmacology , Administration, Intravenous , Anesthesia , Animals , Blood Pressure/drug effects , Male , Morphine/adverse effects , Rats , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/physiopathology , Thyrotropin-Releasing Hormone/chemistry
4.
Am J Physiol Gastrointest Liver Physiol ; 313(4): G320-G329, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28684460

ABSTRACT

Inflammation plays a role in abdominal surgery (AS)-induced intestinal ileus that is alleviated by electrical vagal stimulation. Intracisternal injection of RX-77368, the stable thyrotropin-releasing hormone agonist, activates dorsal motor nucleus neurons and gastric vagal efferent discharges. We investigated the gastric inflammation induced by AS and the modulation by intracisternal RX-77368 in rats. RX-77368 (50 ng/rat) or saline was injected followed, 1 h later, by laparotomy and small intestinal/cecal manipulation. The sham group had anesthesia alone. After 6 h, gastric emptying (GE) and the inflammation in gastric corpus were determined. AS inhibited GE by 72% vs. control and doubled the number of M1-like macrophage immunoreactive for major histocompatibility complex class II (MHCII; M1 marker) but not for cluster of differentiation 206 (CD206; M2 marker) (MHCII+/CD206-) while there was no change in M2-like macrophages (MHCII-/CD206+). AS increased mRNA levels of interleukin-1Ɵ (IL-1Ɵ) and tumor necrosis factor α (TNF-α) by 1.7- and 1.5-fold, respectively, in the gastric submucosa plus muscle layers and the infiltration of neutrophils labeled by myeloperoxidase by 9.5-fold in the muscularis externa. RX-77368 inhibited AS-related gastric changes while not altering these parameters in the sham group. There was a significant negative correlation between GE and IL-1Ɵ (r = -0.46), TNF-α (r = -0.44), M1 macrophage (r = -0.82), and neutrophils (r = -0.91). The M2-like macrophages and IL-10 expression were unchanged by AS with intracisternal saline or RX-77368. These data indicate that AS activates gastric M1 macrophages and increases proinflammatory cytokines expression, which are prevented by central vagal activation and may contribute to the correlated dampening of postoperative gastric ileus.NEW & NOTEWORTHY MHCII+/CD206- (M1) and MHCII-/CD206+ (M2) constitute two distinct populations of macrophages that are in close apposition to the cholinergic neurons in the rat gastric myenteric plexus (MP). Abdominal surgery (6 h) activates M1 macrophage leading to inflammation in the gastric MP correlated with the delayed gastric emptying, which was abolished by central vagal stimulation via intracisternal injection of RX-77368. Vagal stimulation linked with the cephalic phase may have potential beneficial effects to curtail postoperative gastric ileus.


Subject(s)
Enterostomy/adverse effects , Gastrointestinal Motility/immunology , Intestinal Pseudo-Obstruction/immunology , Intestinal Pseudo-Obstruction/prevention & control , Macrophage Activation/immunology , Myenteric Plexus/physiopathology , Vagus Nerve/physiopathology , Animals , Gastrointestinal Motility/drug effects , Intestinal Pseudo-Obstruction/etiology , Macrophage Activation/drug effects , Male , Myenteric Plexus/drug effects , Myoelectric Complex, Migrating/drug effects , Myoelectric Complex, Migrating/immunology , Pyrrolidonecarboxylic Acid/administration & dosage , Pyrrolidonecarboxylic Acid/analogs & derivatives , Rats , Rats, Sprague-Dawley , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/analogs & derivatives , Treatment Outcome , Vagus Nerve/drug effects
5.
Pharmacol Res ; 124: 1-8, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28720519

ABSTRACT

Fatigue affects most cancer patients and has numerous potential causes, including cancer itself and cancer treatment. Cancer-related fatigue (CRF) is not relieved by rest, can decrease quality of life, and has no FDA-approved therapy. Thyrotropin-releasing hormone (TRH) has been proposed as a potential novel treatment for CRF, but its efficacy against CRF remains largely untested. Thus, we tested the TRH analog, taltirelin (TAL), in mouse models of CRF. To model fatigue, we used a mouse model of chemotherapy, a mouse model of radiation therapy, and mice bearing colon 26 carcinoma tumors. We used the treadmill fatigue test to assess fatigue-like behavior after treatment with TAL. Additionally, we used wild-type and TRH receptor knockout mice to determine which TRH receptor was necessary for the actions of TAL. Tumor-bearing mice displayed muscle wasting and all models caused fatigue-like behavior, with mice running a shorter distance in the treadmill fatigue test than controls. TAL reversed fatigue-like behavior in all three models and the mouse TRH1 receptor was necessary for the effects of TAL. These data suggest that TAL may be useful in alleviating fatigue in all cancer patients and provide further support for evaluating TAL as a potential therapy for CRF in humans.


Subject(s)
Fatigue/drug therapy , Nootropic Agents/therapeutic use , Thyrotropin-Releasing Hormone/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/adverse effects , Cell Line, Tumor , Colonic Neoplasms/complications , Colonic Neoplasms/pathology , Disease Models, Animal , Fatigue/etiology , Female , Fluorouracil/adverse effects , Gamma Rays/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/therapeutic use
6.
J Physiol ; 593(2): 415-30, 2015 01 15.
Article in English | MEDLINE | ID: mdl-25630262

ABSTRACT

RaphƩ-derived serotonin (5-HT) and thyrotropin-releasing hormone (TRH) play important roles in fundamental, homeostatic control systems such as breathing and specifically the ventilatory CO2 chemoreflex. Brown Norway (BN) rats exhibit an inherent and severe ventilatory insensitivity to hypercapnia but also exhibit relatively normal ventilation at rest and during other conditions, similar to multiple genetic models of 5-HT system dysfunction in mice. Herein, we tested the hypothesis that the ventilatory insensitivity to hypercapnia in BN rats is due to altered raphƩ gene expression and the consequent deficiencies in raphƩ-derived neuromodulators such as TRH. Medullary raphƩ transcriptome comparisons revealed lower expression of multiple 5-HT neuron-specific genes in BN compared to control Dahl salt-sensitive rats, predictive of reduced central nervous system monoamines by bioinformatics analyses and confirmed by high-performance liquid chromatography measurements. In particular, raphƩ Trh mRNA and peptide levels were significantly reduced in BN rats, and injections of the stable TRH analogue Taltirelin (TAL) stimulated breathing dose-dependently, with greater effects in BN versus control Sprague-Dawley rats. Importantly, TAL also effectively normalized the ventilatory CO2 chemoreflex in BN rats, but TAL did not affect CO2 sensitivity in control Sprague-Dawley rats. These data establish a molecular basis of the neuromodulatory deficiency in BN rats, and further suggest an important functional role for TRH signalling in the mammalian CO2 chemoreflex.


Subject(s)
Hypercapnia/metabolism , Raphe Nuclei/metabolism , Thyrotropin-Releasing Hormone/metabolism , Transcriptome , Animals , Carbon Dioxide/pharmacology , Hypercapnia/genetics , Neurotransmitter Agents/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raphe Nuclei/drug effects , Rats , Rats, Inbred BN , Rats, Inbred Dahl , Rats, Sprague-Dawley , Reflex , Serotonin/metabolism , Species Specificity , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/pharmacology
7.
J Mol Recognit ; 27(10): 609-17, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25178856

ABSTRACT

The present study involves molecular docking, molecular dynamics (MD) simulation studies, and Caco-2 cell monolayer permeability assay to investigate the effect of structural modifications on PepT1-mediated transport of thyrotropin releasing hormone (TRH) analogs. Molecular docking of four TRH analogs was performed using a homology model of human PepT1 followed by subsequent MD simulation studies. Caco-2 cell monolayer permeability studies of four TRH analogs were performed at apical to basolateral and basolateral to apical directions. Inhibition experiments were carried out using Gly-Sar, a typical PepT1 substrate, to confirm the PepT1-mediated transport mechanism of TRH analogs. Papp of the four analogs follows the order: NP-1894 < NP-2378 < NP-1896 < NP-1895. Higher absorptive transport was observed in the case of TRH analogs, indicating the possibility of a carrier-mediated transport mechanism. Further, the significant inhibition of the uptake of Gly-Sar by TRH analogs confirmed the PepT1-mediated transport mechanism. Glide docking scores of all the four analogues were in good agreement with their transport rates, suggesting the role of substrate binding affinity in the PepT1-mediated transport of TRH analogs. MD simulation studies revealed that the polar interactions with amino acid residues present in the active site are primarily responsible for substrate binding, and a downward trend was observed with the increase in bulkiness at the N-histidyl moiety of TRH analogs.


Subject(s)
Intestinal Mucosa/metabolism , Models, Molecular , Symporters/chemistry , Thyrotropin-Releasing Hormone/chemistry , Binding Sites , Biological Transport , Caco-2 Cells , Chromatography, High Pressure Liquid , Computational Biology , Computer Simulation , Humans , Molecular Dynamics Simulation , Peptide Transporter 1 , Permeability , Symporters/metabolism , Symporters/physiology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacokinetics
8.
Peptides ; 175: 171181, 2024 May.
Article in English | MEDLINE | ID: mdl-38423212

ABSTRACT

Thyrotropin-releasing hormone (TRH) acts centrally to exert pleiotropic actions independently from its endocrine function, including antinociceptive effects against somatic pain in rodents. Whether exogenous or endogenous activation of TRH signaling in the brain modulates visceral pain is unknown. Adult male Sprague-Dawley rats received an intracerebroventricular (ICV) injection of the stable TRH analog, RX-77368 (10, 30 and 100Ć¢Ā€ĀÆng/rat) or saline (5Ć¢Ā€ĀÆĀµl) or were semi-restrained and exposed to cold (4Ā°C) for 45Ć¢Ā€ĀÆmin. The visceromotor response (VMR) to graded phasic colorectal distensions (CRD) was monitored using non-invasive intracolonic pressure manometry. Naloxone (1Ć¢Ā€ĀÆmg/kg) was injected subcutaneously 10Ć¢Ā€ĀÆmin before ICV RX-77368 or saline. Fecal pellet output was monitored for 1Ć¢Ā€ĀÆh after ICV injection. RX-77368 ICV (10, 30 and 100Ć¢Ā€ĀÆng/rat) reduced significantly the VMR by 56.7%, 67.1% and 81.1% at 40Ć¢Ā€ĀÆmmHg and by 30.3%, 58.9% and 87.4% at 60Ć¢Ā€ĀÆmmHg respectively vs ICV saline. Naloxone reduced RX-77368 (30 and 100Ć¢Ā€ĀÆng, ICV) analgesic response by 51% and 28% at 40Ć¢Ā€ĀÆmmHg and by 30% and 33% at 60Ć¢Ā€ĀÆmmHg respectively, but had no effect per se. The visceral analgesia was mimicked by the acute exposure to cold. At the doses of 30 and 100Ć¢Ā€ĀÆng, ICV RX-77368 induced defecation within 30Ć¢Ā€ĀÆmin. These data established the antinociceptive action of RX-77368 injected ICV in a model of visceral pain induced by colonic distension through recruitment of both opioid and non-opioid dependent mechanisms.


Subject(s)
Colorectal Neoplasms , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/analogs & derivatives , Visceral Pain , Rats , Male , Animals , Rats, Sprague-Dawley , Visceral Pain/drug therapy , Analgesics/pharmacology , Naloxone/pharmacology
9.
Mol Pharm ; 9(9): 2458-68, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22779445

ABSTRACT

The present study, in general, is aimed to uncover the properties of the transport mechanism or mechanisms responsible for the uptake of NP-647 into Caco-2 cells and, in particular, to understand whether it is a substrate for the intestinal oligopeptide transporter, PEPT1 (SLC15A1). NP-647 showed a carrier-mediated, saturable transport with Michaelis-Menten parameters K(m) = 1.2 mM and V(max) = 2.2 ĀµM/min. The effect of pH, sodium ion (Na(+)), glycylsarcosine and amoxicillin (substrates of PEPT1), and sodium azide (Na(+)/K(+)-ATPase inhibitor) on the flux rate of NP-647 was determined. Molecular docking and molecular dynamics simulation studies were carried out to investigate molecular interactions of NP-647 with transporter using homology model of human PEPT1. The permeability coefficient (P(appCaco-2)) of NP-647 (32.5 Ɨ 10(-6) cm/s) was found to be four times higher than that of TRH. Results indicate that NP-647 is transported into Caco-2 cells by means of a carrier-mediated, proton-dependent mechanism that is inhibited by Gly-Sar and amoxicillin. In turn, NP-647 also inhibits the uptake of Gly-Sar into Caco-2 cells and, together, this evidence suggests that PEPT1 is involved in the process. Docking and molecular dynamics simulation studies indicate high affinity of NP-647 toward PEPT1 binding site as compared to TRH. High permeability of NP-647 over TRH is attributed to its increased hydrophobicity which increases its affinity toward PEPT1 by interacting with the hydrophobic pocket of the transporter through hydrophobic forces.


Subject(s)
Anticonvulsants/pharmacokinetics , Symporters/metabolism , Thyrotropin-Releasing Hormone/analogs & derivatives , Amoxicillin/pharmacology , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Biological Transport/drug effects , Caco-2 Cells , Cell Line, Tumor , Dipeptides/pharmacology , Drug Stability , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Models, Molecular , Molecular Dynamics Simulation , Peptide Transporter 1 , Sodium/metabolism , Sodium Azide/metabolism , Symporters/chemistry , Thyrotropin-Releasing Hormone/chemistry , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/pharmacokinetics
10.
J Recept Signal Transduct Res ; 31(6): 416-22, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22044177

ABSTRACT

We investigated the effect of taltirelin hydrate ((−)-N-[(S)-hexahydro-1-methyl- 2,6-dioxo-4-pyrimidinyl-carbonyl]-L-histidyl-L-prolinamide tetrahydrate; taltirelin), a metabolically stable thyrotropin-releasing hormone (TRH) analog, on circulatory function, respiratory function, and viable time after bleeding in urethane-anesthetized rats. Massive volume-controlled bleeding caused marked reductions in mean arterial pressure (MAP) and respiratory rate (RR). The vital signs of control rats were lost within an average of 23 min after bleeding. Intravenous administration of taltirelin (0.03−0.3 mg/kg) and TRH (1 and 3 mg/kg) immediately after bleeding accelerated recovery of MAP and RR, and prolonged viable time in a dose-dependent manner. The potency of taltirelin in accelerating MAP and RR recovery and prolonging viable time was higher when compared with that of TRH. In addition, recovery of MAP and RR and the extension of viable time by taltirelin were inhibited by preintraperitoneal administration of atropine sulfate, which is a centrally acting muscarinic antagonist, but not by that of atropine methylbromide, which is a peripherally acting muscarinic antagonist. Taltirelin also recovered decreased arterial pH, bicarbonate ions, and base excess, and prevented a decrease in arterial oxygen saturation. In conclusion, the anti-shock effect of taltirelin was more potent than that of TRH. Taltirelin activity was mediated by the central muscarinic cholinergic system. In addition, taltirelin also corrected metabolic acidosis. These results suggest that taltirelin could be useful in the treatment of hypovolemic shock.


Subject(s)
Hypovolemia/prevention & control , Shock, Hemorrhagic/drug therapy , Thyrotropin-Releasing Hormone/analogs & derivatives , Animals , Atropine/administration & dosage , Atropine/pharmacology , Blood Gas Analysis , Blood Pressure/drug effects , Hypovolemia/etiology , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/drug effects , Receptors, Thyrotropin-Releasing Hormone/metabolism , Respiratory Rate/drug effects , Shock, Hemorrhagic/complications , Thyrotropin-Releasing Hormone/therapeutic use
11.
Eur J Pharmacol ; 882: 173271, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32534077

ABSTRACT

Rovatirelin is a newly synthetized thyrotropin-releasing hormone (TRH) analog. This study aimed to investigate the effect of rovatirelin on motor function using rolling mouse Nagoya (RMN), a mouse model of hereditary ataxia, and compare it with that of taltirelin, which is clinically used to treat spinocerebellar degeneration in Japan. We also examined the effect of rovatirelin on glucose metabolism in various brain regions of RMN using autoradiography (ARG). Rovatirelin (1, 3, 10, and 30Ā mg/kg) dose-dependently reduced the fall index in RMN, and its effect was more potent than that of taltirelin (3, 10, 30, and 100Ā mg/kg). No attenuation of the effect was observed by repeated daily administration for 2 weeks. Furthermore, the reduction in the fall index by rovatirelin persisted for 2 weeks after completing treatment. In the ARG study, rovatirelin induced a significantly elevated uptake of glucose in the prefrontal cortex, nucleus accumbens shell, nucleus accumbens core, striatum, anterior cingulate cortex, secondary motor area, pretectal area, ventral tegmental area, black pars compacta, locus coeruleus, nucleus cerebellaris middle nucleus, medial nucleus of the vestibular nerve, fourth/fifth lobule, and third lobule. Furthermore, rovatirelin increased cerebellar mRNA level of brain derived neurotrophic factor. These results suggest that rovatirelin activates the cerebellum and other parts of the central nervous system to improve motor function in spinocerebellar ataxia (SCA) model animals, and its action is more potent than that of taltirelin. Therefore, rovatirelin can be a potential alternative to the traditionally used therapeutics for SCA.


Subject(s)
Ataxia/drug therapy , Oxazolidinones/therapeutic use , Pyrrolidines/therapeutic use , Spinocerebellar Degenerations/drug therapy , Animals , Ataxia/genetics , Ataxia/metabolism , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Female , Glucose/metabolism , Male , Mice , Oxazolidinones/pharmacology , Pyrrolidines/pharmacology , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/metabolism , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/therapeutic use
12.
J Clin Pharmacol ; 60(10): 1314-1323, 2020 10.
Article in English | MEDLINE | ID: mdl-32459872

ABSTRACT

The effects of itraconazole on the pharmacokinetics of rovatirelin were investigated in an open-label, single-sequence drug-drug interaction study in 16 healthy subjects. Subjects were administered a single oral dose of rovatirelin (1.6 mg) on day 1 and day 15. From day 8 through 16, subjects received daily oral doses of itraconazole (200 mg/day). Concentrations of rovatirelin and (thiazolylalanyl)methylpyrrolidine (TAMP), the major metabolite of rovatirelin formed by cytochrome P450 (CYP) 3A4/5, were determined in plasma and urine. Pharmacokinetic parameters were used to evaluate the drug-drug interaction potential of rovatirelin as a victim. With coadministration, maximum concentration (Cmax ) and area under the concentration-time curve extrapolated to infinity (AUCinf ) of rovatirelin increased 3.05-fold and 2.82-fold, respectively, and the 90% confidence intervals of the ratios for Cmax (2.64-3.52) and AUCinf (2.47-3.23) did not fall within the 0.8-1.25 boundaries. Urinary excretion of rovatirelin increased at almost the same ratio as the AUCinf ratio with coadministration; however, renal clearance did not change. Cmax , AUCinf , and urinary excretion of TAMP were decreased by coadministration. Itraconazole has the potential to inhibit drug transport via intestinal P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP); therefore, substrate assessments of rovatirelin for the 2 transporters were evaluated using Caco-2 cell monolayers. In vitro studies showed that rovatirelin is a substrate for P-gp but not for BCRP. The current study shows that itraconazole's effect on rovatirelin pharmacokinetics is mediated through inhibition of CYP3A4/5 and intestinal P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Itraconazole/pharmacokinetics , Oxazolidinones/pharmacokinetics , Pyrrolidines/pharmacokinetics , Thyrotropin-Releasing Hormone/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Administration, Oral , Adult , Area Under Curve , Asian People , Biological Transport/drug effects , Caco-2 Cells , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Elimination Routes/drug effects , Healthy Volunteers , Hormones/blood , Humans , Itraconazole/administration & dosage , Itraconazole/pharmacology , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxazolidinones/administration & dosage , Oxazolidinones/adverse effects , Oxazolidinones/metabolism , Permeability/drug effects , Pyrrolidines/administration & dosage , Pyrrolidines/adverse effects , Pyrrolidines/metabolism , Young Adult
13.
J Physiol ; 587(Pt 6): 1179-86, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19204048

ABSTRACT

Central orexin/hypocretin neurones are critical for sustaining consciousness: their firing stimulates wakefulness and their destruction causes narcolepsy. We explored whether the activity of orexin cells is modulated by thyrotropin-releasing hormone (TRH), an endogenous stimulant of wakefulness and locomotor activity whose mechanism of action is not fully understood. Living orexin neurones were identified by targeted expression of green fluorescent protein (GFP) in acute brain slices of transgenic mice. Using whole-cell patch-clamp recordings, we found that TRH robustly increased the action potential firing rate of these neurones. TRH-induced excitation persisted under conditions of synaptic isolation, and involved a Na(+)-dependent depolarization and activation of a mixed cation current in the orexin cell membrane. By double-label immunohistochemistry, we found close appositions between TRH-immunoreactive nerve terminals and orexin-A-immunoreactive cell bodies. These results identify a new physiological modulator of orexin cell firing, and suggest that orexin cell excitation may contribute to the arousal-enhancing actions of TRH.


Subject(s)
Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Neuropeptides/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Action Potentials/drug effects , Animals , Green Fluorescent Proteins/genetics , Hypothalamus/cytology , Hypothalamus/drug effects , In Vitro Techniques , Membrane Potentials/drug effects , Mice , Mice, Inbred Strains , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Orexins , Patch-Clamp Techniques , Sodium/metabolism , Tetrodotoxin/pharmacology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism
14.
Epilepsy Behav ; 14(1): 48-53, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18952198

ABSTRACT

Thyrotropin-releasing hormone (TRH) and its analogs have a number of neurobiological functions and therapeutic uses in disorders of the central nervous system. In this study, the newly synthesized TRH analogs were evaluated for central nervous system activity in pentobarbital-induced sleeping in mice. The most potent TRH analog (L-pGlu-(2-propyl)-L-His-L-ProNH(2) coded as NP-647) was evaluated for its antiepileptic potential in various seizure models in mice in comparison with TRH. Intravenous pretreatment with NP-647 (10 and 20 micromol/kg body wt) significantly delayed the onset and reduced the frequency of convulsions in the pentylenetetrazole model, but not in the maximum electroshock seizure model. Also, it was found to be protective against picrotoxin- and kainic acid-induced seizures. However, NP-647 did not significantly affect theophylline-induced seizures. Further study of the effect of NP-647 on locomotor activity and a functional observational battery revealed that it did not significantly exhibit any undesirable effects as compared with vehicle and TRH. NP-647 did not significantly affect cerebral blood flow, whereas the native peptide TRH markedly increased cerebral blood flow. Furthermore, NP-647 exerted antiepileptic activity without significantly altering plasma thyroid-stimulating hormone levels and mean arterial blood pressure. This suggests that NP-647 is more selective for central nervous system activity and devoid of hormonal and cerebrovascular system effects. In contrast, TRH exhibited cardiac and endocrine effects as marked by significant elevation in mean arterial blood pressure and plasma thyroid-stimulating hormone levels. This study demonstrates that NP-647 has potential antiepileptic activity devoid of undesirable effects and, thus, can be exploited for the prevention and treatment of epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacology , Animals , Anticonvulsants/toxicity , Behavior, Animal/drug effects , Blood Pressure/drug effects , Cardiovascular System/drug effects , Central Nervous System Stimulants/pharmacology , Cerebrovascular Circulation/drug effects , Convulsants/antagonists & inhibitors , Electroshock , Hypnotics and Sedatives/antagonists & inhibitors , Hypnotics and Sedatives/pharmacology , Kainic Acid/antagonists & inhibitors , Male , Mice , Motor Activity/drug effects , Pentylenetetrazole/antagonists & inhibitors , Picrotoxin/antagonists & inhibitors , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/prevention & control , Sleep/drug effects , Theophylline/antagonists & inhibitors , Thyrotropin/blood , Thyrotropin-Releasing Hormone/therapeutic use , Thyrotropin-Releasing Hormone/toxicity
15.
Behav Brain Res ; 360: 255-261, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30529403

ABSTRACT

Fatigue is a common symptom in many diseases and disorders and can reduce quality of life, yet lacks an adequate pharmacological intervention. To identify and develop such interventions, and to better understand fatigue, additional preclinical research is necessary. However, despite numerous mouse behavioral assays reportedly detecting fatigue-like behavior, the assumption that fatigue-like behavior is detected in many assays has not been validated through a cross-assay study. Thus, we modeled fatigue in mice by administering 5-fluorouracil, a chemotherapy drug known to cause fatigue in humans and fatigue-like behavior in mice, then evaluated its effects via voluntary wheel running activity (VWRA), locomotor activity in the open field test (OFT), immobility in the forced swim test (FST), and distance run in the treadmill fatigue test (TFT) and treadmill exercise capacity test. Additionally, taltirelin or methylphenidate was administered to alleviate fatigue-like behavior. As a result of 5-fluorouracil treatment, VWRA and the TFT were markedly reduced, indicating fatigue. The OFT, FST, and treadmill exercise capacity test, however, failed to detect fatigue-like behavior. Interestingly, both taltirelin and methylphenidate alleviated fatigue-like behavior in TFT. These data suggest that, of the current assays, only the TFT and VWRA should be expected to detect fatigue-like behavior. Moreover, this study provides additional evidence that taltirelin may provide a novel treatment for chemotherapy-induced fatigue and warrants further evaluation as an anti-fatigue therapeutic.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Behavior, Animal/drug effects , Fatigue/chemically induced , Fatigue/drug therapy , Fluorouracil/toxicity , Nootropic Agents/therapeutic use , Analysis of Variance , Animals , Body Weight/drug effects , Central Nervous System Stimulants/therapeutic use , Disease Models, Animal , Exercise Test , Exploratory Behavior/drug effects , Female , Locomotion/drug effects , Methylphenidate/therapeutic use , Mice , Mice, Inbred C57BL , Swimming/psychology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/therapeutic use , Time Factors
16.
Curr Med Chem ; 15(26): 2718-33, 2008.
Article in English | MEDLINE | ID: mdl-18991632

ABSTRACT

Thyrotropin-releasing hormone (TRH), a hypothalamic orally active neuropeptide, has been manifested in a wide range of biological responses. Besides its central role in regulating the pituitary-thyroid axis by simulating the release of thyrotropin, TRH has considerable influence on the activity of a number of neurobiological systems. Due to the therapeutic potential of TRH to treat several CNS maladies, the development of CNS-selective and metabolically stable TRH analogs is an area of interest. TRH is known to elicit its biological response through two G-protein coupled receptors for TRH (namely, TRH-R1 and TRH-R2). The distinct distribution of TRH receptors in tissues has provided opportunity to discover receptor subtype-specific analogs, which would demonstrate high CNS activities, and are completely free of hormonal activities. In this review, an in-depth analysis of the chemistry and biology of TRH and its analogs is provided. Recent discoveries of TRH-R2 selective analogs, TRH super agonists, metabolically stable TRH analogs, and targeted delivery of TRH analogs have been also discussed.


Subject(s)
Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Animals , Homeostasis/drug effects , Humans , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/biosynthesis , Thyrotropin-Releasing Hormone/pharmacology
17.
Psychoneuroendocrinology ; 33(9): 1183-97, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18657370

ABSTRACT

Sustained abnormalities of glucocorticoid levels have been associated with neuropsychiatric illnesses such as major depression, posttraumatic stress disorder (PTSD), panic disorder, and obsessive compulsive disorder. The pathophysiological effects of glucocorticoids may depend not only on the amount of glucocorticoid exposure but also on its temporal pattern, since it is well established that hormone receptors are down-regulated by continuously elevated cognate hormones. We have previously reported that TRH (pGlu-His-Pro-NH2) and TRH-like peptides (pGlu-X-Pro-NH2) have endogenous antidepressant-like properties and mediate or modulate the acute effects of a single i.p. injection of high dose corticosterone (CORT) in rats. For these reasons, two accepted methods for inducing chronic hyperglucocorticoidemia have been compared for their effects on brain and peripheral tissue levels of TRH and TRH-like peptides in male, 250 g, Sprague-Dawley rats: (1) the dosing effect of CORT hemisuccinate in drinking water, and (2) s.c. slow-release pellets. Overall, there were 93% more significant changes in TRH and TRH-like peptide levels in brain and 111% more in peripheral tissues of those rats ingesting various doses of CORT in drinking water compared to those with 1-3 s.c. pellets. We conclude that providing rats with CORT in drinking water is a convenient model for the pathophysiological effects of hyperglucocorticoidemia in rodents.


Subject(s)
Brain/metabolism , Corticosterone/administration & dosage , Drug Implants , Thyrotropin-Releasing Hormone/metabolism , Administration, Oral , Analysis of Variance , Animals , Brain/drug effects , Corticosterone/blood , Dose-Response Relationship, Drug , Drinking , Drug Administration Schedule , Drug Delivery Systems , Epididymis/anatomy & histology , Male , Organ Size , Pancreas/anatomy & histology , Prostate/anatomy & histology , Rats , Rats, Sprague-Dawley , Testis/anatomy & histology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/genetics , Thyroxine/blood , Triiodothyronine/blood
18.
Neurosci Lett ; 431(1): 26-30, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18069127

ABSTRACT

Glp-Asn-Pro-D-Tyr-D-TrpNH(2) is a novel synthetic peptide that mimics and amplifies central actions of thyrotropin-releasing hormone (TRH) in rat without releasing TSH. The aim of this study was to compare the binding properties of this pentapeptide and its all-L counterpart (Glp-Asn-Pro-Tyr-TrpNH(2)) to TRH receptors in native rat brain tissue and cells expressing the two TRH receptor subtypes identified in rat to date, namely TRHR1 and TRHR2. Radioligand binding studies were carried out using [(3)H][3-Me-His(2)]TRH to label receptors in hippocampal, cortical and pituitary tissue, GH4 pituitary cells, as well as CHO cells expressing TRHR1 and/or TRHR2. In situ hybridization studies suggest that cortex expresses primarily TRHR2 mRNA, hippocampus primarily TRHR1 mRNA and pituitary exclusively TRHR1 mRNA. Competition experiments showed [3-Me-His(2)]TRH potently displaced [(3)H][3-Me-His(2)]TRH binding from all tissues/cells investigated. Glp-Asn-Pro-D-Tyr-D-TrpNH(2) in concentrations up to 10(-5)M did not displace [(3)H][3-Me-His(2)]TRH binding to membranes derived from GH4 cells or CHO-TRHR1 cells, consistent with its lack of binding to pituitary membranes and TSH-releasing activity. Similar results were obtained for the corresponding all-L peptide. In contrast, both pentapeptides displaced binding from rat hippocampal membranes (pIC(50) Glp-Asn-Pro-D-Tyr-D-TrpNH(2): 7.7+/-0.2; pIC(50) Glp-Asn-Pro-Tyr-TrpNH(2): 6.6+/-0.2), analogous to cortical membranes (pIC(50) Glp-Asn-Pro-D-Tyr-D-TrpNH(2): 7.8+/-0.2; pIC(50) Glp-Asn-Pro-Tyr-TrpNH(2): 6.6+/-0.2). Neither peptide, however, displaced [(3)H][3-Me-His(2)]TRH binding to CHO-TRHR2. Thus, this study reveals for the first time significant differences in the binding properties of native and heterologously expressed TRH receptors. Also, the results raise the possibility that Glp-Asn-Pro-D-Tyr-D-TrpNH(2) is not displacing [(3)H][3-Me-His(2)]TRH from a known TRH receptor in rat cortex, but rather a hitherto unidentified TRH receptor.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Oligopeptides/metabolism , Pituitary Gland/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Amino Acid Sequence/physiology , Animals , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , CHO Cells , Cerebral Cortex/drug effects , Cricetinae , Cricetulus , Hippocampus/drug effects , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Pituitary Gland/drug effects , Radioligand Assay , Rats , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/chemical synthesis , Thyrotropin-Releasing Hormone/pharmacology
19.
J Org Chem ; 73(21): 8591-3, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18826326

ABSTRACT

The synthesis of the phosphinic analogue of thyrotropin releasing hormone (TRH) GlpPsi[P(O)(OH)]HisProNH2, where the scissile peptide bond of TRH has been replaced by the hydrolytically stable phosphinic bond, has been achieved by a multistep synthetic strategy, providing thus one of the most potent synthetic inhibitors of pyroglutamyl peptidase II (PPII) reported to date (170 nM). The key synthetic step, an Ugi-type condensation reaction, produced directly the suitably protected for solid-phase peptide synthesis pseudodipeptidic block FmocGlu(OMe)Psi[P(O)(OH)]His(Tr)OH. Formation of the pyroglutamic ring was performed on solid phase, providing thus a general method for synthesizing pyroglutamyl phosphinic peptides on solid phase. Using this strategy, the phosphinic analogue of TRH has been synthesized for the first time.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Phosphinic Acids/chemistry , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/analogs & derivatives , Protease Inhibitors/chemical synthesis , Pyrrolidonecarboxylic Acid/antagonists & inhibitors , Thyrotropin-Releasing Hormone/chemical synthesis
20.
Br J Pharmacol ; 150(4): 403-14, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17220907

ABSTRACT

BACKGROUND AND PURPOSE: Exogenously administered thyrotropin-releasing hormone (TRH) is known to exert potent but short-acting centrally-mediated antinociceptive effects. We sought to investigate the mechanisms underlying these effects using the synthetic TRH analogue taltirelin, focusing on the descending monoaminergic systems in mice. EXPERIMENTAL APPROACH: The mice received systemic or local injections of taltirelin combined with either central noradrenaline (NA) or 5-hydroxytryptamine (5-HT) depletion by 6-hydroxydopamine (6-OHDA) or DL-p-chlorophenylalanine (PCPA), respectively, or blockade of their receptors. The degree of antinociception was determined using the tail flick and tail pressure tests. KEY RESULTS: Subcutaneously (s.c.) administered taltirelin exhibited dose-dependent antinociceptive effects in the tail flick and tail pressure tests. These effects appeared to be primarily supraspinally mediated, since intracerebroventricularly (i.c.v.) but not intrathecally (i.t.) injected taltirelin generated similar effects. Depletion of central NA abolished only the analgesic effect of taltirelin (s.c. and i.c.v.) on mechanical nociception. By contrast, depletion of central 5-HT abolished only its analgesic effect on thermal nociception. Intraperitoneal (i.p.) and i.t. injection of the alpha2-adrenoceptor antagonist yohimbine respectively reduced the analgesic effect of taltirelin (s.c. and i.c.v.) on mechanical nociception. By contrast, the 5-HT1A receptor antagonist WAY-100635 (i.p. and i.t.) reduced the effect of taltirelin (s.c. and i.c.v.) on thermal nociception. Neither the 5-HT2 receptor antagonist ketanserin nor the opioid receptor antagonist naloxone altered the antinociceptive effect of taltirelin. CONCLUSIONS AND IMPLICATIONS: These findings suggest that taltirelin activates the descending noradrenergic and serotonergic pain inhibitory systems, respectively, to exert its analgesic effects on mechanical and thermal nociception.


Subject(s)
Analgesics , Biogenic Monoamines/physiology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin/analogs & derivatives , Adrenergic alpha-Antagonists/pharmacology , Animals , Dose-Response Relationship, Drug , Fenclonine/pharmacology , Hot Temperature , Injections, Intraventricular , Injections, Spinal , Injections, Subcutaneous , Ketanserin/pharmacology , Male , Mice , Mice, Inbred ICR , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neural Pathways/drug effects , Norepinephrine/physiology , Oxidopamine/pharmacology , Pain Measurement/drug effects , Physical Stimulation , Reaction Time/drug effects , Serotonin/physiology , Serotonin Agents/pharmacology , Sympatholytics/pharmacology , Thyrotropin-Releasing Hormone/pharmacology , Yohimbine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL