Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.025
1.
Int J Toxicol ; 42(4): 334-344, 2023.
Article En | MEDLINE | ID: mdl-36634266

Thyrotropin-releasing hormone (TRH) and TRH-like peptides carry a therapeutic potential for neurological conditions. Nanoparticles (NP) made of the biodegradable polymer, Poly(Sebacic Anhydride) (PSA), have been developed to carry TRH, intended for intranasal administration to patients. There is limited information on the safety of biodegradable polymers when given intranasally, and therefore, we have performed two preclinical safety and toxicity studies in cynomolgus monkeys and rats using TRH-PSA nanoparticles. The rats and monkeys were dosed intranasally for 42 days or 28 days, respectively, and several animals were followed for additional 14 days. Animals received either placebo, vehicle (PSA), or different concentrations of TRH-PSA. No systemic adverse effects were seen. Changes in T3 or T4 concentrations were observed in some TRH-PSA-treated animals, which did not have clinical or microscopic correlates. No effect was seen on TSH or prolactin concentrations. In the monkey study, microscopic changes in the nasal turbinates were observed, which were attributed to incidental mechanical trauma caused during administration. Taken together, the TRH-loaded PSA NPs have proven to be safe, with no local or systemic adverse effects attributed to the drug loaded nanoparticles. These findings provide additional support to the growing evidence of the safety of peptide-loaded NPs for intranasal delivery and pave the way for future clinical trials in humans.


Nanoparticles , Thyrotropin-Releasing Hormone , Thyrotropin-Releasing Hormone/administration & dosage , Animals , Rats , Macaca fascicularis , Administration, Intranasal , Male , Female
2.
Chem Pharm Bull (Tokyo) ; 69(4): 314-324, 2021.
Article En | MEDLINE | ID: mdl-33790077

We explored orally effective thyrotropin-releasing hormone (TRH) mimetics, which show high central nervous system effects in structure-activity relationship studies based on in vivo antagonistic activity on reserpine-induced hypothermia (anti-hypothermic effect) in mice starting from TRH. This led us to the TRH mimetic: [(4S,5S)-(5-methyl-2-oxooxazolidine-4-yl)carbonyl]-[3-(thiazol-4-yl)-L-alanyl]-L-prolinamide 1, which shows a higher anti-hypothermic effect compared with that of TRH after oral administration. We next attempted further chemical modification of the N- and C-terminus of 1 to find more orally effective TRH mimetics. As a result, we obtained several N- and C-terminus modified TRH mimetics which showed high anti-hypothermic effects.


Hypothermia/drug therapy , Proline/analogs & derivatives , Thyrotropin-Releasing Hormone/chemical synthesis , Thyrotropin-Releasing Hormone/pharmacology , Administration, Oral , Animals , Body Temperature Regulation/drug effects , Male , Proline/administration & dosage , Proline/chemical synthesis , Proline/chemistry , Proline/pharmacology , Rats, Sprague-Dawley , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/chemistry
3.
Cochrane Database Syst Rev ; 5: CD011505, 2020 05 18.
Article En | MEDLINE | ID: mdl-32421208

BACKGROUND: Many women express concern about their ability to produce enough milk, and insufficient milk is frequently cited as the reason for supplementation and early termination of breastfeeding. When addressing this concern, it is important first to consider the influence of maternal and neonatal health, infant suck, proper latch, and feeding frequency on milk production, and that steps be taken to correct or compensate for any contributing issues. Oral galactagogues are substances that stimulate milk production. They may be pharmacological or non-pharmacological (natural). Natural galactagogues are usually botanical or other food agents. The choice between pharmacological or natural galactagogues is often influenced by familiarity and local customs. Evidence for the possible benefits and harms of galactagogues is important for making an informed decision on their use. OBJECTIVES: To assess the effect of oral galactagogues for increasing milk production in non-hospitalised breastfeeding mother-term infant pairs. SEARCH METHODS: We searched the Cochrane Pregnancy and Childbirth Group's Trials Register, ClinicalTrials.gov, the World Health Organization (WHO) International Clinical Trials Registry Platform (ICTRP), Health Research and Development Network - Phillippines (HERDIN), Natural Products Alert (Napralert), the personal reference collection of author LM, and reference lists of retrieved studies (4 November 2019). SELECTION CRITERIA: We included randomised controlled trials (RCTs) and quasi-RCTs (including published abstracts) comparing oral galactagogues with placebo, no treatment, or another oral galactagogue in mothers breastfeeding healthy term infants. We also included cluster-randomised trials but excluded cross-over trials. DATA COLLECTION AND ANALYSIS: We used standard Cochrane Pregnancy and Childbirth methods for data collection and analysis. Two to four review authors independently selected the studies, assessed the risk of bias, extracted data for analysis and checked accuracy. Where necessary, we contacted the study authors for clarification. MAIN RESULTS: Forty-one RCTs involving 3005 mothers and 3006 infants from at least 17 countries met the inclusion criteria. Studies were conducted either in hospitals immediately postpartum or in the community. There was considerable variation in mothers, particularly in parity and whether or not they had lactation insufficiency. Infants' ages at commencement of the studies ranged from newborn to 6 months. The overall certainty of evidence was low to very low because of high risk of biases (mainly due to lack of blinding), substantial clinical and statistical heterogeneity, and imprecision of measurements. Pharmacological galactagogues Nine studies compared a pharmacological galactagogue (domperidone, metoclopramide, sulpiride, thyrotropin-releasing hormone) with placebo or no treatment. The primary outcome of proportion of mothers who continued breastfeeding at 3, 4 and 6 months was not reported. Only one study (metoclopramide) reported on the outcome of infant weight, finding little or no difference (mean difference (MD) 23.0 grams, 95% confidence interval (CI) -47.71 to 93.71; 1 study, 20 participants; low-certainty evidence). Three studies (metoclopramide, domperidone, sulpiride) reported on milk volume, finding pharmacological galactagogues may increase milk volume (MD 63.82 mL, 95% CI 25.91 to 101.72; I² = 34%; 3 studies, 151 participants; low-certainty evidence). Subgroup analysis indicates there may be increased milk volume with each drug, but with varying CIs. There was limited reporting of adverse effects, none of which could be meta-analysed. Where reported, they were limited to minor complaints, such as tiredness, nausea, headache and dry mouth (very low-certainty evidence). No adverse effects were reported for infants. Natural galactagogues Twenty-seven studies compared natural oral galactagogues (banana flower, fennel, fenugreek, ginger, ixbut, levant cotton, moringa, palm dates, pork knuckle, shatavari, silymarin, torbangun leaves or other natural mixtures) with placebo or no treatment. One study (Mother's Milk Tea) reported breastfeeding rates at six months with a concluding statement of "no significant difference" (no data and no measure of significance provided, 60 participants, very low-certainty evidence). Three studies (fennel, fenugreek, moringa, mixed botanical tea) reported infant weight but could not be meta-analysed due to substantial clinical and statistical heterogeneity (I2 = 60%, 275 participants, very low-certainty evidence). Subgroup analysis shows we are very uncertain whether fennel or fenugreek improves infant weight, whereas moringa and mixed botanical tea may increase infant weight compared to placebo. Thirteen studies (Bu Xue Sheng Ru, Chanbao, Cui Ru, banana flower, fenugreek, ginger, moringa, fenugreek, ginger and turmeric mix, ixbut, mixed botanical tea, Sheng Ru He Ji, silymarin, Xian Tong Ru, palm dates; 962 participants) reported on milk volume, but meta-analysis was not possible due to substantial heterogeneity (I2 = 99%). The subgroup analysis for each intervention suggested either benefit or little or no difference (very low-certainty evidence). There was limited reporting of adverse effects, none of which could be meta-analysed. Where reported, they were limited to minor complaints such as mothers with urine that smelled like maple syrup and urticaria in infants (very low-certainty evidence). Galactagogue versus galactagogue Eight studies (Chanbao; Bue Xue Sheng Ru, domperidone, moringa, fenugreek, palm dates, torbangun, moloco, Mu Er Wu You, Kun Yuan Tong Ru) compared one oral galactagogue with another. We were unable to perform meta-analysis because there was only one small study for each match-up, so we do not know if one galactagogue is better than another for any outcome. AUTHORS' CONCLUSIONS: Due to extremely limited, very low certainty evidence, we do not know whether galactagogues have any effect on proportion of mothers who continued breastfeeding at 3, 4 and 6 months. There is low-certainty evidence that pharmacological galactagogues may increase milk volume. There is some evidence from subgroup analyses that natural galactagogues may benefit infant weight and milk volume in mothers with healthy, term infants, but due to substantial heterogeneity of the studies, imprecision of measurements and incomplete reporting, we are very uncertain about the magnitude of the effect. We are also uncertain if one galactagogue performs better than another. With limited data on adverse effects, we are uncertain if there are any concerning adverse effects with any particular galactagogue; those reported were minor complaints. High-quality RCTs on the efficacy and safety of galactagogues are urgently needed. A set of core outcomes to standardise infant weight and milk volume measurement is also needed, as well as a strong basis for the dose and dosage form used.


Galactogogues/administration & dosage , Lactation/drug effects , Milk, Human , Phytotherapy/methods , Plant Extracts/administration & dosage , Administration, Oral , Body Weight/drug effects , Breast Feeding , Domperidone/administration & dosage , Domperidone/adverse effects , Female , Galactogogues/adverse effects , Humans , Infant , Infant, Newborn , Metoclopramide/administration & dosage , Metoclopramide/adverse effects , Milk, Human/drug effects , Mothers , Phytotherapy/adverse effects , Plant Extracts/adverse effects , Randomized Controlled Trials as Topic , Sulpiride/administration & dosage , Sulpiride/adverse effects , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/adverse effects
4.
J Vet Intern Med ; 34(3): 1350-1356, 2020 May.
Article En | MEDLINE | ID: mdl-32255541

BACKGROUND: Determination of plasma adrenocotrophic hormone (ACTH) concentration (endogenous or thyrotropin-releasing hormone [TRH] stimulation test) is the most commonly used diagnostic test for pituitary pars intermedia dysfunction (PPID) in horses. Because ACTH is unstable, samples often are frozen to be shipped to laboratories or to allow for batch analysis of research samples. However, the effect of multiple freeze-thaw cycles on equine ACTH is unknown. OBJECTIVE: To determine the effects of multiple freeze-thaw cycles on immunoreactive ACTH concentration. ANIMALS: Twenty-eight horses ranging from 10 to 27 years of age were used. METHODS: Prospective study. Horses were divided into 4 groups: group 1, PPID-negative, without TRH stimulation; group 2, PPID-negative, with TRH stimulation; group 3, PPID-positive, without TRH stimulation; and group 4, PPID-positive, with TRH stimulation. Whole blood was collected from each horse at baseline or 30 minutes after TRH stimulation. Immunoreactive plasma ACTH concentration was determined using a chemiluminescence assay. Plasma samples then were frozen at -80°C >24 hours, thawed at 4°C and reanalyzed for 5 freeze-thaw cycles. Changes in plasma ACTH concentration were analyzed using a linear mixed-effect model. RESULTS: Significant effects of freeze-thaw cycles (P = .001) and PPID status (P = .04) on plasma ACTH concentration were observed, but no significant effect of TRH stimulation was identified. CONCLUSIONS AND CLINICAL IMPORTANCE: The plasma ACTH concentration is altered by freeze-thaw cycles, and the effect is observed sooner in horses with PPID. To diagnose PPID, multiple freeze-thaw cycles should be avoided when measuring plasma ACTH concentration.


Adrenocorticotropic Hormone/blood , Freezing/adverse effects , Horse Diseases/diagnosis , Pituitary Diseases/veterinary , Specimen Handling/veterinary , Animals , Diagnostic Tests, Routine , Female , Horse Diseases/blood , Horses , Luminescent Measurements/veterinary , Male , Pituitary Diseases/blood , Pituitary Diseases/diagnosis , Prospective Studies , Specimen Handling/methods , Thyrotropin-Releasing Hormone/administration & dosage
6.
J Vet Intern Med ; 33(5): 2249-2256, 2019 Sep.
Article En | MEDLINE | ID: mdl-31498947

BACKGROUND: The thyrotropin-releasing hormone (TRH) stimulation test and the 2-step insulin sensitivity test are commonly used methods to diagnose, respectively, pituitary pars intermedia dysfunction (PPID) and insulin dysregulation (ID). OBJECTIVES: To investigate the diagnostic value of combining the TRH stimulation test and the 2-step insulin sensitivity test to diagnose PPID and ID simultaneously. ANIMALS: Twenty-seven adult horses, 10 control horses without PPID or ID, 5 horses with PPID only, 5 horses with ID only, and 7 horses with PPID and ID. METHODS: Randomized prospective study. Horses underwent a TRH stimulation test alone, a 2-step insulin sensitivity test alone, and combined testing with simultaneous TRH and insulin injection in the same syringe. Data were compared by 2-way repeated measures analysis of variance and 2 1-sided tests to demonstrate equivalence. Bland-Altman plots were generated to visualize agreement between combined and independent testing. RESULTS: The effect of combined testing on plasma adrenocorticotropic hormone, blood glucose concentration, or percentage decrease in blood glucose concentration was not significantly different from the effect obtained with independent testing. One control horse appeared falsely positive for PPID, 2 PPID-only horses appeared falsely positive for ID, and 1 PPID and ID horse appeared falsely negative for ID when tests were performed simultaneously. Bland-Altman plots supported the agreement between combined and independent testing. CONCLUSIONS AND CLINICAL IMPORTANCE: Combining the TRH stimulation test and the 2-step insulin sensitivity test appears to be a useful diagnostic tool for equine practitioners in the field, allowing testing of a horse for both PPID and ID simultaneously.


Adrenocorticotropic Hormone/blood , Horse Diseases/diagnosis , Insulin Resistance , Pituitary Diseases/veterinary , Animals , Blood Glucose , Female , Horse Diseases/blood , Horse Diseases/metabolism , Horses , Male , Pituitary Diseases/blood , Pituitary Diseases/diagnosis , Pituitary Gland, Intermediate/pathology , Prospective Studies , Random Allocation , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/pharmacology
7.
Domest Anim Endocrinol ; 68: 135-141, 2019 07.
Article En | MEDLINE | ID: mdl-31082785

Published information on the pharmacokinetic and pharmacodynamic properties of pergolide is limited. The aim of this study was to investigate the pharmacokinetic and pharmacodynamic properties of oral pergolide in horses with pituitary pars intermedia dysfunction (PPID). The study design was a nonrandomized clinical trial. Six horses with PPID diagnosed by thyrotropin-releasing hormone (TRH) stimulation tests received pergolide at 4 µg/kg for 18 d. Plasma samples for determination of pergolide and ACTH concentration were collected 0.5 h before and 2 and 12 h after each administration of pergolide. Maximum plasma concentrations after the first oral dose of pergolide (0.104-0.684 ng/mL; median 0.261 ng/mL; interquartile range [IQR] 0.184-0.416 ng/mL) were not significantly different to the maximum steady-state concentration at day 18 (0.197-0.628 ng/mL; median 0.274; IQR 0.232-0.458 ng/mL). Chronic administration was not associated with drug accumulation (R = 1.09) and pergolide concentration reached steady state within 3 d. Throughout, concentrations of pergolide fluctuated considerably, with median plasma peak concentrations more than four times higher than median trough concentrations. Plasma ACTH concentration reduced significantly within 12 h of administration with further reductions occurring up to 10 d after the initiation of treatment. Although there were parallel fluctuations in the concentrations of pergolide and ACTH, timing of ACTH measurement in relation to the administration of pergolide did not have a significant effect. Alterations in the response to TRH were identified at 8 d with no further change being identified at 18 d. A small number of horses were studied. Oral pergolide results in significant suppression of pars intermedia activity within hours. Pergolide and ACTH concentrations fluctuated in tandem although correlation was poor. Fluctuations in pergolide concentration were consistent with a terminal elimination half-life of less than 12 h. To reduce the level of fluctuation of ACTH, twice-daily dosing of pergolide may be more appropriate.


Horse Diseases/drug therapy , Pergolide/pharmacokinetics , Pituitary Diseases/veterinary , Pituitary Gland, Intermediate/drug effects , Administration, Oral , Adrenocorticotropic Hormone/blood , Animals , Area Under Curve , Horses , Pergolide/administration & dosage , Pergolide/blood , Pergolide/therapeutic use , Pituitary Diseases/drug therapy , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/pharmacology
8.
J Control Release ; 295: 278-289, 2019 02 10.
Article En | MEDLINE | ID: mdl-30610951

Nose-to-brain delivery of drugs is affected by nanoparticles (NPs) deposited on the olfactory surface and absorbed directly into the brain. Thyrotropin releasing hormone (TRH), a water soluble drug used for treating suicidal patients, was incorporated into a fast degrading poly(sebacic anhydride) (PSA) NPs. NPs were prepared by a solvent-antisolvent process under strict anhydrous environment to obtain high TRH loading and to avoid premature PSA degradation and TRH release. PSA and TRH were dissolved in a mixture of dichloromethane and ethanol and added dropwise to a dispersion of mannitol particles in heptane as an antisolvent. Mannitol powder was included in the antisolvent, so that formed NPs adhered to the mannitol microparticles for easy isolation and immediate dispersion in water prior to use. The size, surface charge, and morphology of the TRH-PSA NPs were determined using dynamic light scattering (DLS), zeta-potential, and Scanning Electron Microscopy (SEM), respectively. The NPs prepared were uniform and spherical of ~250 nm. Further, the in vitro release profile of TRH from NPs lasted for 12 h with most TRH released within the first hour in water. Concentration dependent cell toxicity studies revealed low toxicity level at low concentrations of the NPs. Surface adsorption of the NPs was also uniform on the cell surface as examined through the odyssey near infrared fluorescence (NIR) images using Indocyanine green (ICG). The NPs are designed to enable direct delivery to the olfactory epithelium using a refillable nasal atomizer that deposits mist onto the olfactory neuro-epithelium.


Anhydrides/chemistry , Decanoic Acids/chemistry , Drug Carriers/chemistry , Hormones/administration & dosage , Nanoparticles/chemistry , Thyrotropin-Releasing Hormone/administration & dosage , Administration, Intranasal , Cell Line, Tumor , Drug Liberation , Hormones/pharmacokinetics , Humans , Thyrotropin-Releasing Hormone/pharmacokinetics
9.
Vet Q ; 38(1): 72-78, 2018 Dec.
Article En | MEDLINE | ID: mdl-30362899

BACKGROUND: In dogs, spontaneous Cushing's syndrome is most often pituitary-dependent and caused by hypersecretion of adrenocorticotropic hormone (ACTH), resulting in increased adrenocortical glucocorticoid secretion similar to horses. In horses with Cushing's syndrome (or pituitary pars intermedia dysfunction [PPID]) a thyrotropin-releasing hormone (TRH) stimulation test can be used for diagnosis, as TRH administration results in increased circulating ACTH and cortisol concentrations in affected horses. OBJECTIVE: The aim of this study was to investigate the effect of TRH administration on the circulating ACTH and cortisol concentrations in dogs with pituitary-dependent hypercortisolism (PDH). METHODS: Ten clinically normal control dogs and 10 dogs with PDH, all client owned, underwent a TRH stimulation test with measurement of plasma concentrations of ACTH and cortisol, before and after intravenous administration of 10 µg TRH/kg bodyweight. RESULTS: Plasma ACTH concentration did not rise significantly after TRH stimulation, neither in PDH dogs nor in clinically normal dogs. In contrast, the plasma cortisol concentration did increase significantly after TRH stimulation in both groups (p = .003 in PDH and p < .001 in control). Immunohistochemistry of normal adrenal glands demonstrated the presence of TRH receptors in the whole adrenal cortex. CONCLUSIONS: The results of this study demonstrate that the TRH stimulation test should be rejected as a tool to diagnose PDH in dogs. The observed TRH-induced increase in plasma cortisol concentration without a significant rise in plasma ACTH concentration may be explained by a direct effect of TRH on adrenocortical cells mediated by adrenocortical TRH receptors.


Adrenocorticotropic Hormone/analysis , Dog Diseases/diagnosis , Hydrocortisone/analysis , Pituitary ACTH Hypersecretion/veterinary , Thyrotropin-Releasing Hormone/administration & dosage , Adrenocorticotropic Hormone/urine , Analysis of Variance , Animals , Case-Control Studies , Cushing Syndrome , Dog Diseases/blood , Dog Diseases/urine , Dogs , Female , Hydrocortisone/blood , Immunohistochemistry/veterinary , Male , Pituitary ACTH Hypersecretion/diagnosis
10.
J Pharmacol Exp Ther ; 366(1): 105-112, 2018 07.
Article En | MEDLINE | ID: mdl-29674333

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% ± 6% to 131% ± 7% and 45% ± 6% to 168% ± 13%, respectively (percent baseline; n = 4 ± S.E.M.), primarily through increased breathing rates (from 76% ± 9% to 260% ± 14% and 66% ± 8% to 318% ± 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% ± 5% to 126% ± 12% and 64% ± 7% to 116% ± 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.


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
11.
Am J Physiol Gastrointest Liver Physiol ; 313(4): G320-G329, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28684460

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-1ß (IL-1ß) 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-1ß (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.


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
12.
Anim Sci J ; 88(10): 1588-1594, 2017 Oct.
Article En | MEDLINE | ID: mdl-28556305

The aim of the present study was to clarify the relationship between hypothalamic dopamine (DA) and salsolinol (SAL) for the secretion of prolactin (PRL) in goats. SAL or thyrotropin-releasing hormone (TRH) was intravenously injected into female goats treated with or without the D2 DA receptor antagonist haloperidol (Hal), which crosses the blood-brain barrier, and the PRL-releasing response to SAL was compared with that to TRH. PRL-releasing responses to SAL, Hal, and Hal plus SAL were also examined after a pretreatment to augment central DA using carbidopa (Carbi) and L-dopa. The PRL-releasing response to Hal alone was greater than that to SAL or TRH alone. The PRL-releasing response to Hal plus SAL was similar to that of Hal alone. In contrast, the PRL-releasing response to Hal plus TRH was greater than that to TRH or Hal alone. The treatment with Carbi plus L-dopa inhibited SAL- and Hal-induced PRL secretion. The inhibition of the PRL-releasing response to SAL disappeared when SAL was injected with Hal. These results indicate that the mechanisms underlying the SAL-induced PRL response differ from those of TRH, and suggest that hypothalamic DA and its synthesis is associated in part with SAL-induced PRL secretion in goats.


Dopamine/physiology , Goats/metabolism , Goats/physiology , Hypothalamus , Isoquinolines/pharmacology , Prolactin/metabolism , Animals , Carbidopa/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Drug Combinations , Female , Haloperidol/pharmacology , Injections, Intravenous , Isoquinolines/administration & dosage , Levodopa/pharmacology , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/pharmacology
13.
J Nanosci Nanotechnol ; 16(5): 5324-32, 2016 May.
Article En | MEDLINE | ID: mdl-27483926

Thyrotropin-Releasing Hormone (TRH), a tripeptide amide with molecular formula L-pGlu-L-His-L- Pro-NH2, is used in the treatment of brain/spinal injury and certain central nervous system (CNS) disorders, including schizophrenia, Alzheimer's disease, epilepsy, depression, shock and ischemia due to its profound effects on the CNS. However, TRH's therapeutic activity is severely hampered because of instability and hydrophilicity owing to its peptidic nature which results into ineffective penetration into the blood brain barrier. In the present study, we report the synthesis and stability studies of novel chitosan engineered TRH encapsulated poly(lactide-co-glycolide) (PLGA) based nanoformulation. The aim of such an encapsulation is to allow effective delivery of TRH in biological systems as the peptidase degrade naked TRH. The synthesis of TRH was carried out manually in solution phase followed by its encapsulation using PLGA to form polymeric nanoparticles (NPs) via nanoprecipitation technique. Different parameters such as type of organic phase, concentration of stabilizer, ratio of organic phase and aqueous phase, rate of addition of organic phase were optimized, tested and evaluated for particle size, encapsulation efficiency, and stability of NPs. The TRH-PLGA NPs were then surface modified with chitosan to achieve positive surface charge rendering them potential membrane penetrating agents. PLGA, PLGA-TRH, Chitosan-PLGA and Chitosan-PLGA-TRH NPs were characterized and analyzed using Dynamic Light Scattering (DLS), Transmissiom Electron Microscopy (TEM) and Infra-red spectroscopic techniques.


Chitosan/chemistry , Delayed-Action Preparations/chemical synthesis , Lactic Acid/chemistry , Nanocapsules/chemistry , Polyglycolic Acid/chemistry , Thyrotropin-Releasing Hormone/chemistry , Absorption, Physicochemical , Delayed-Action Preparations/administration & dosage , Diffusion , Drug Compounding/methods , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Neuropeptides/administration & dosage , Neuropeptides/chemistry , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Thyrotropin-Releasing Hormone/administration & dosage
14.
Gen Comp Endocrinol ; 230-231: 76-86, 2016 05 01.
Article En | MEDLINE | ID: mdl-27044512

Growth hormone (GH), together with thyroid hormones (TH), regulates growth and development, and has critical effects on vertebrate metabolism. In ectotherms, these physiological processes are strongly influenced by environmental temperature. In reptiles, however, little is known about the direct influences of this factor on the somatotropic and thyroid axes. Therefore, the aim of this study was to describe the effects of both acute (48h) and chronic (2weeks) exposure to sub-optimal temperatures (25 and 18°C) upon somatotropic and thyroid axis function of the green iguana, in comparison to the control temperature (30-35°C). We found a significant increase in GH release (2.0-fold at 25°C and 1.9-fold at 18°C) and GH mRNA expression (up to 3.7-fold), mainly under chronic exposure conditions. The serum concentration of insulin-like growth factor-I (IGF-I) was significantly greater after chronic exposure (18.5±2.3 at 25°C; 15.92±3.4 at 18°C; vs. 9.3±1.21ng/ml at 35°C), while hepatic IGF-I mRNA expression increased up to 6.8-fold. Somatotropic axis may be regulated, under acute conditions, by thyrotropin-releasing hormone (TRH) that significantly increased its hypothalamic concentration (1.45 times) and mRNA expression (0.9-fold above control), respectively; and somatostatin (mRNA expression increased 1.0-1.2 times above control); and under chronic treatment, by pituitary adenylate cyclase-activating peptide (PACAP mRNA expression was increased from 0.4 to 0.6 times). Also, it was shown that, under control conditions, injection of TRH stimulated a significant increase in circulating GH. On the other hand, while there was a significant rise in the hypothalamic content of TRH and its mRNA expression, this hormone did not appear to influence the thyroid axis activity, which showed a severe diminution in all conditions of cold exposure, as indicated by the decreases in thyrotropin (TSH) mRNA expression (up to one-eight of the control), serum T4 (from 11.6±1.09 to 5.3±0.58ng/ml, after 2weeks at 18°C) and T3 (from 0.87±0.09 to 0.05±0.01ng/ml, under chronic conditions at 25°C), and Type-2 deiodinase (D2) activity (from 992.5±224 to 213.6±26.4fmolI(125)T4/mgh). The reduction in thyroid activity correlates with the down-regulation of metabolism as suggested by the decrease in the serum glucose and free fatty acid levels. These changes apparently were independent of a possible stress response, at least under acute exposure to both temperatures and in chronic treatment to 25°C, since serum corticosterone had no significant changes in these conditions, while at chronic 18°C exposure, a slight increase (0.38 times above control) was found. Thus, these data suggest that the reptilian somatotropic and thyroid axes have differential responses to cold exposure, and that GH and TRH may play important roles associated to adaptation mechanisms that support temperature acclimation in the green iguana.


Growth Hormone/metabolism , Iguanas/metabolism , Temperature , Thyroid Gland/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Blood Glucose/analysis , Corticosterone/blood , Growth Hormone/genetics , Hypothalamus/drug effects , Hypothalamus/metabolism , Iguanas/blood , Iguanas/genetics , Insulin-Like Growth Factor I/genetics , Iodide Peroxidase/metabolism , Liver/drug effects , Liver/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/blood , Somatostatin/genetics , Thyroid Gland/drug effects , Thyroid Hormones/blood , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Thyrotropin/genetics , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/pharmacology
15.
Anim Sci J ; 87(12): 1522-1527, 2016 Dec.
Article En | MEDLINE | ID: mdl-26997651

The aim of the present study was to clarify the effect of extracerebral dopamine (DA) on salsolinol (SAL)-induced prolactin (PRL) secretion in goats. An intravenous injection of SAL or thyrotropin-releasing hormone (TRH) was given to female goats before and after treatment with an extracerebral DA receptor antagonist, domperidone (DOM), and the PRL-releasing response to SAL was compared with that to TRH. DOM alone increased plasma PRL concentrations and the PRL-releasing response to DOM alone was greater than that to either SAL alone or TRH alone. The PRL-releasing response to DOM plus SAL was similar to that to DOM alone, and no additive effect of DOM and SAL on the secretion of PRL was observed. In contrast, the PRL-releasing response to DOM plus TRH was greater than that to either TRH alone or DOM alone and DOM synergistically increased TRH-induced PRL secretion. The present results demonstrate that the mechanism involved in PRL secretion by SAL differs from that by TRH, and suggest that the extracerebral DA might be associated in part with the modulation of SAL-induced PRL secretion in goats.


Dopamine/physiology , Goats/physiology , Isoquinolines/pharmacology , Prolactin/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Animals , Domperidone/pharmacology , Dopamine Antagonists/pharmacology , Female , Injections, Intravenous , Isoquinolines/administration & dosage , Stimulation, Chemical , Thyrotropin-Releasing Hormone/administration & dosage
16.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1120-7, 2016 06 01.
Article En | MEDLINE | ID: mdl-27009047

In rat paraventricular thalamic nucleus (PVT) neurons, activation of thyrotropin-releasing hormone (TRH) receptors enhances neuronal excitability via concurrent decrease in a G protein-coupled inwardly rectifying K (GIRK)-like conductance and opening of a cannabinoid receptor-sensitive transient receptor potential canonical (TRPC)-like conductance. Here, we investigated the calcium (Ca(2+)) contribution to the components of this TRH-induced response. TRH-induced membrane depolarization was reduced in the presence of intracellular BAPTA, also in media containing nominally zero [Ca(2+)]o, suggesting a critical role for both intracellular Ca(2+) release and Ca(2+) influx. TRH-induced inward current was unchanged by T-type Ca(2+) channel blockade, but was decreased by blockade of high-voltage-activated Ca(2+) channels (HVACCs). Both the pharmacologically isolated GIRK-like and the TRPC-like components of the TRH-induced response were decreased by nifedipine and increased by BayK8644, implying Ca(2+) influx via L-type Ca(2+) channels. Only the TRPC-like conductance was reduced by either thapsigargin or dantrolene, suggesting a role for ryanodine receptors and Ca(2+)-induced Ca(2+) release in this component of the TRH-induced response. In pituitary and other cell lines, TRH stimulates MAPK. In PVT neurons, only the GIRK-like component of the TRH-induced current was selectively decreased in the presence of PD98059, a MAPK inhibitor. Collectively, the data imply that TRH-induced depolarization and inward current in PVT neurons involve both a dependency on extracellular Ca(2+) influx via opening of L-type Ca(2+) channels, a sensitivity of a TRPC-like component to intracellular Ca(2+) release via ryanodine channels, and a modulation by MAPK of a GIRK-like conductance component.


Calcium Channels, L-Type/metabolism , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Midline Thalamic Nuclei/physiology , Neurons/physiology , Thyrotropin-Releasing Hormone/administration & dosage , Animals , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Female , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Male , Midline Thalamic Nuclei/drug effects , Neurons/drug effects , Rats , Rats, Wistar
17.
Neuroscience ; 311: 81-91, 2015 Dec 17.
Article En | MEDLINE | ID: mdl-26470810

In rat thalamic paraventricular nucleus of thalamus (PVT) neurons, activation of thyrotropin-releasing hormone (TRH) receptors enhances excitability via concurrent decrease in G protein-coupled inwardly-rectifying potassium (GIRK)-like and activation of transient receptor potential cation (TRPC)4/5-like cationic conductances. An exploration of intracellular signaling pathways revealed the TRH-induced current to be insensitive to phosphatidylinositol-specific phospholipase C (PI-PLC) inhibitors, but reduced by D609, an inhibitor of phosphatidylcholine-specific PLC (PC-PLC). A corresponding change in the I-V relationship implied suppression of the cationic component of the TRH-induced current. Diacylglycerol (DAG) is a product of the hydrolysis of PC. Studies focused on the isolated cationic component of the TRH-induced response revealed a reduction by RHC80267, an inhibitor of DAG lipase, the enzyme involved in the hydrolysis of DAG to the endocannabinoid 2-arachidonoylglycerol (2-AG). Further investigation revealed enhancement of the cationic component in the presence of either JZL184 or WWL70, inhibitors of enzymes involved in the hydrolysis of 2-AG. A decrease in the TRH-induced response was noted in the presence of rimonabant or SR144528, membrane permeable CB1 and CB2 receptor antagonists, respectively. A decrease in the TRH-induced current by intracellular, but not by bath application of the membrane impermeable peptide hemopressin, selective for CB1 receptors, suggests a postsynaptic intracellular localization of these receptors. The TRH-induced current was increased in the presence of arachidonyl-2'-chloroethylamide (ACEA) or JWH133, CB1 and CB2 receptor agonists, respectively. The PI3-kinase inhibitor LY294002, known to inhibit TRPC translocation, decreased the response to TRH. In addition, a TRH-induced enhancement of the low-threshold spike was prevented by both rimonabant, and SR144528. TRH had no influence on excitatory or inhibitory miniature postsynaptic currents, suggesting presynaptic CB receptors are not involved in this situation. Collectively, the data imply that activation of TRH receptors in these midline thalamic neurons engages novel signaling pathways that include postsynaptic intracellular CB1 and CB2 receptors in the activation of TRPC4/5-like channels.


Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , TRPC Cation Channels/metabolism , Animals , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Patch-Clamp Techniques , Rats, Wistar , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/metabolism , Tissue Culture Techniques
18.
Eur J Pharmacol ; 761: 413-22, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-26142830

Rovatirelin ([1-[-[(4S,5S)-(5-methyl-2-oxo oxazolidin-4-yl) carbonyl]-3-(thiazol-4-yl)-l-alanyl]-(2R)-2-methylpyrrolidine) is a novel synthetic agent that mimics the actions of thyrotropin-releasing hormone (TRH). The aim of this study was to investigate the electrophysiological and pharmacological effects of rovatirelin on the central noradrenergic system and to compare the results with those of another TRH mimetic agent, taltirelin, which is approved for the treatment of spinocerebellar degeneration (SCD) in Japan. Rovatirelin binds to the human TRH receptor with higher affinity (Ki=702nM) than taltirelin (Ki=3877nM). Rovatirelin increased the spontaneous firing of action potentials in the acutely isolated noradrenergic neurons of rat locus coeruleus (LC). The facilitatory action of rovatirelin on the firing rate in the LC neurons was inhibited by the TRH receptor antagonist, chlordiazepoxide. Reduction of the extracellular pH increased the spontaneous firing of LC neurons and rovatirelin failed to increase the firing frequency further, indicating an involvement of acid-sensitive K+ channels in the rovatirelin action. In in vivo studies, oral administration of rovatirelin increased both c-Fos expression in the LC and extracellular levels of noradrenaline (NA) in the medial prefrontal cortex (mPFC) of rats. Furthermore, rovatirelin increased locomotor activity. The increase in NA level and locomotor activity by rovatirelin was more potent and longer acting than those by taltirelin. These results indicate that rovatirelin exerts a central nervous system (CNS)-mediated action through the central noradrenergic system, which is more potent than taltirelin. Thus, rovatirelin may have an orally effective therapeutic potential in patients with SCD.


Adrenergic Neurons/drug effects , Locus Coeruleus/drug effects , Oxazolidinones/pharmacology , Prefrontal Cortex/drug effects , Pyrrolidines/pharmacology , Thyrotropin-Releasing Hormone/pharmacology , Action Potentials , Administration, Oral , Adrenergic Neurons/metabolism , Animals , Dose-Response Relationship, Drug , Ligands , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Male , Microdialysis , Motor Activity/drug effects , Norepinephrine/metabolism , Oxazolidinones/administration & dosage , Oxazolidinones/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Pyrrolidines/administration & dosage , Pyrrolidines/metabolism , Radioligand Assay , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Time Factors
19.
J Perinatol ; 35(9): 725-8, 2015 Sep.
Article En | MEDLINE | ID: mdl-26110495

OBJECTIVE: Whether hormone supplementation is necessary for infants with transient hypothyroxinemia of prematurity (THOP) remains controversial, and further analysis of the hypothalamus-pituitary-thyroid axis of infants with THOP is necessary. STUDY DESIGN: Thyrotropin-releasing hormone (TRH) stimulation tests were performed at 2 weeks of age in 50 infants with a gestational age of 30 weeks or less, and the data were analyzed retrospectively. RESULT: Subjects were divided into three groups; group A consisted of euthyroid infants, group B consisted of infants with THOP and group C consisted of hypothyroid infants. The basal and peak thyroid-stimulating hormone level of group C in response to TRH stimulation tests was significantly higher than the others, but no differences were observed between groups A and B. CONCLUSION: The response of infants with THOP to the TRH stimulation test was not different from that of euthyroid infants, which suggested that their hypothalamic-pituitary-thyroid axis was appropriately regulated in infants with THOP.


Hormone Replacement Therapy/methods , Hypothyroidism , Infant, Premature, Diseases , Infant, Premature/blood , Thyrotropin-Releasing Hormone , Thyroxine/blood , Female , Hormones/administration & dosage , Hormones/metabolism , Humans , Hypothyroidism/blood , Hypothyroidism/diagnosis , Hypothyroidism/etiology , Hypothyroidism/therapy , Infant , Infant, Newborn , Infant, Premature, Diseases/blood , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/etiology , Infant, Premature, Diseases/therapy , Japan , Male , Monitoring, Physiologic/methods , Retrospective Studies , Stimulation, Chemical , Thyrotropin/analysis , Thyrotropin/metabolism , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/metabolism
20.
Psychoneuroendocrinology ; 59: 71-80, 2015 Sep.
Article En | MEDLINE | ID: mdl-26036452

BACKGROUND: We previously demonstrated that the difference between 2300h and 0800h TSH response to protirelin (TRH) tests on the same day (ΔΔTSH test) is an improved measure in detecting hypothalamic-pituitary-thyroid (HPT) axis dysregulation in depression. This chronobiological index (1) is reduced in about three quarters of major depressed inpatients, and (2) is normalized after successful antidepressant treatment. In the present study, we examined whether early changes in HPT axis activity during the first 2 weeks of antidepressant treatment could be associated with subsequent outcome. METHODS: The ΔΔTSH test was performed in 50 drug-free DSM-IV euthyroid major depressed inpatients and 50 hospitalized controls. After 2 weeks of antidepressant treatment the ΔΔTSH test was repeated in all inpatients. Antidepressant response was evaluated after 6 weeks of treatment. RESULTS: At baseline, ΔΔTSH values were significantly lower in patients compared to controls and 38 patients (76%) showed reduced ΔΔTSH values (i.e., <2.5mU/L). After 2 weeks of antidepressant treatment, 20 patients showed ΔΔTSH normalization (among them 18 were subsequent remitters), while 18 patients did not normalize their ΔΔTSH (among them 15 were non-remitters) (p<0.00001). Among the 12 patients who had normal ΔΔTSH values at baseline, 8 out 9 who had still normal values after 2 weeks of treatment were remitters, while the 3 with worsening HPT axis function (i.e., reduced ΔΔTSH value after 2 weeks of treatment) were non-remitters (p<0.02). A logistic regression analysis revealed that ΔΔTSH levels after 2 weeks of treatment could predict the probability of remission (odds ratio [OR]=2.11, 95% confidence interval [CI]=1.31-3.41). CONCLUSIONS: Our results suggest that after 2 weeks of antidepressant treatment: (1) chronobiological restoration of the HPT axis activity precedes clinical remission, and (2) alteration of the HPT axis is associated with treatment resistance.


Antidepressive Agents/administration & dosage , Depressive Disorder, Major/drug therapy , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Adult , Case-Control Studies , Chronobiology Phenomena/drug effects , Depressive Disorder, Major/blood , Depressive Disorder, Major/metabolism , Female , Humans , Hypothalamo-Hypophyseal System/metabolism , Male , Middle Aged , Pituitary-Adrenal System/metabolism , Predictive Value of Tests , Thyrotropin/blood , Thyrotropin/metabolism , Thyrotropin-Releasing Hormone/administration & dosage , Treatment Outcome
...