Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Sci ; 162(1): 301-308, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29182718

ABSTRACT

Respiratory tract toxicity represents a significant cause of attrition of inhaled drug candidates targeting respiratory diseases. One of the key issues to allow early detection of respiratory toxicities is the lack of reliable and predictive in vitro systems. Here, the relevance and value of a physiologically relevant 3D human airway in vitro model (MucilAir) were explored by repeated administration of a set of compounds with (n = 8) or without (n = 7) respiratory toxicity following inhalation in vivo. Predictability for respiratory toxicity was evaluated by readout of cytotoxicity, barrier integrity, viability, morphology, ciliary beating frequency, mucociliary clearance and cytokine release. Interestingly, the data show that in vivo toxicity can be predicted in vitro by studying cell barrier integrity by transepithelial electrical resistance (TEER), and cell viability determined by the Resazurin method. Both read-outs had 88% sensitivity and 100% specificity, respectively, while the former was more accurate with receiver operating characteristic (ROC) AUC of 0.98 (p = .0018) compared with ROC AUC of 0.90 (p = .0092). The loss of cell barrier integrity could mainly, but not fully, be attributed to a loss of cell coverage in 6 out of 7 compounds with reduced TEER. Notably, these effects occurred only at 400 µM, at concentration levels significantly above primary target cell potency, suggesting that greater attention to high local lung concentrations should be taken into account in safety assessment of inhaled drugs. Thus, prediction of respiratory toxicity in 3D human airway in vitro models may result in improved animal welfare and reduced attrition in inhaled drug discovery projects.


Subject(s)
Drugs, Investigational/toxicity , Epithelial Cells/drug effects , Inhalation Exposure/adverse effects , Models, Biological , Respiratory System/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Electric Impedance , Epithelial Cells/pathology , Humans , Predictive Value of Tests , Respiratory System/pathology
2.
Autoimmun Rev ; 15(11): 1062-1070, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27491564

ABSTRACT

RORγ is a nuclear hormone receptor which controls polarization of naive CD4+ T-cells into proinflammatory Th17 cells. Pharmacological antagonism of RORγ has therapeutic potential for autoimmune diseases; however, this mechanism may potentially carry target-related safety risks, as mice deficient in Rorc, the gene encoding RORγ, develop T-cell lymphoma with 50% frequency. Due to the requirement of RORγ during development, the Rorc knockout (KO) animals lack secondary lymphoid organs and have a dysregulation in the generation of CD4+ and CD8+ T cells. We wanted to extend the evaluation of RORγ deficiency to address the question whether lymphomas, similar to those observed in the Rorc KO, would develop in an animal with an otherwise intact adult immune system. Accordingly, we designed a conditional RORγ knockout mouse (Rorc CKO) where the Rorc locus could be deleted in adult animals. Based on these studies we can confirm that these animals also develop lymphoma in a similar time frame as embryonic Rorc knockouts. This study also suggests that in animals where the gene deletion is incomplete, the thymus undergoes a rapid selection process replacing Rorc deficient cells with remnant thymocytes carrying a functional Rorc locus and that subsequently, these animals do not develop lymphoblastic lymphoma.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Autoimmune Diseases/therapy , Male , Mice , Mice, Knockout , Th17 Cells/immunology
3.
Eur J Pharmacol ; 634(1-3): 138-41, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20176012

ABSTRACT

While patients with symptoms of gastroesophageal reflux disease generally respond well to proton pump inhibitors, 20-30% continue to experience troublesome symptoms. In such cases, agents that target transient lower esophageal sphincter (LES) relaxation may be useful as add-on therapy to proton pump inhibitors. The GABAB receptor agonist baclofen inhibits transient LES relaxation but it is not an ideal agent due to central nervous system activity. Lesogaberan (AZD3355) is a peripherally restricted GABAB receptor agonist with limited central nervous system activity that inhibits transient LES relaxation in dogs. In the present study, the comparative effects of lesogaberan (7 micromol/kg) and baclofen (2.8 micromol/kg) on reflux were studied in dogs using 24-h pHmetry. Drugs (or vehicle control) were administered orally prior to the first meal of the day, and the number of reflux episodes (pH<4 for > or = 5 s) and acid exposure time were computed for the 24-h monitoring period. The mean (S.E.M.) number of reflux episodes/24 h was 4.6 (0.4) and 6.4 (0.6) for lesogaberan and baclofen, respectively, versus 10.7 (0.5) for control (P<0.0001 for both). Acid exposure time was 51.2 (4.5) min for control versus 23.6 (3.8) min for lesogaberan (P<0.0001) and 35.4 (6.5) min with baclofen (P=0.05). It is concluded that lesogaberan significantly reduces acid reflux in dogs, with comparable efficacy to baclofen.


Subject(s)
Esophagus/drug effects , GABA Agonists/therapeutic use , GABA-B Receptor Agonists , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/prevention & control , Phosphinic Acids/therapeutic use , Propylamines/therapeutic use , Animals , Baclofen/administration & dosage , Baclofen/pharmacology , Dogs , Esophagus/physiology , Female , GABA Agonists/pharmacology , Gastric Acid/metabolism , Gastroesophageal Reflux/physiopathology , Hydrogen-Ion Concentration/drug effects , Male , Manometry/methods , Phosphinic Acids/pharmacology , Pilot Projects , Propylamines/pharmacology , Receptors, GABA-B/physiology , Time Factors
4.
J Pharmacol Exp Ther ; 331(2): 504-12, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19648470

ABSTRACT

Gastroesophageal reflux disease (GERD) affects >10% of the Western population. Conventionally, GERD is treated by reducing gastric acid secretion, which is effective in most patients but inadequate in a significant minority. We describe a new therapeutic approach for GERD, based on inhibition of transient lower esophageal sphincter relaxation (TLESR) with a proposed peripherally acting GABA(B) receptor agonist, (R)-(3-amino-2-fluoropropyl)phosphinic acid (AZD3355). AZD3355 potently stimulated recombinant human GABA(B) receptors and inhibited TLESR in dogs, with a biphasic dose-response curve. In mice, AZD3355 produced considerably less central side effects than the prototypical GABA(B) receptor agonist baclofen but evoked hypothermia at very high doses (blocked by a GABA(B) receptor antagonist and absent in GABA(B)-/- mice). AZD3355 and baclofen differed markedly in their distribution in rat brain; AZD3355, but not baclofen, was concentrated in circumventricular organs as a result of active uptake (shown by avid intracellular sequestration) and related to binding of AZD3355 to native GABA transporters in rat cerebrocortical membranes. AZD3355 was also shown to be transported by all four recombinant human GABA transporters. AR-H061719 [(R/S)-(3-amino-2-fluoropropyl)phosphinic acid], (the racemate of AZD3355) inhibited the response of ferret mechanoreceptors to gastric distension, further supporting its peripheral site of action on TLESR. In summary, AZD3355 probably inhibits TLESR through stimulation of peripheral GABA(B) receptors and may offer a potential new approach to treatment of GERD.


Subject(s)
Esophageal Sphincter, Lower/drug effects , GABA Agonists/pharmacology , GABA-B Receptor Agonists , Peripheral Nerves/drug effects , Phosphinic Acids/pharmacology , Propylamines/pharmacology , Animals , Autoradiography , Baclofen/pharmacology , Binding, Competitive/drug effects , Calcium/metabolism , Dogs , Dose-Response Relationship, Drug , Esophageal Sphincter, Lower/innervation , Female , Ferrets/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Humans , Hypothermia/chemically induced , In Vitro Techniques , Membrane Potentials/drug effects , Mice , Muscle Relaxation/drug effects , Protein Binding , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/metabolism , Vagus Nerve/drug effects , Vagus Nerve/physiology
5.
Br J Pharmacol ; 146(1): 89-97, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15980875

ABSTRACT

The effects of the novel GABA analogue (2R)-(3-amino-2-fluoropropyl)sulphinic acid (AFPSiA) on transient lower oesophageal sphincter relaxations (TLOSRs) were studied in the dog. In addition, the GABA(A)/GABA(B) selectivity was determined in vitro and in vivo, and the pharmacokinetics and the metabolism of the compound were studied in the dog and rat. TLOSRs were reduced by 55 +/- 8% after intragastric administration of AFPSiA at 14 mumol kg(-1) and did not decrease further at higher doses. When evaluated 2 and 4 h after administration, the effect declined to 37 +/- 6 and 16 +/- 9%, respectively. Spontaneous swallowing was only significantly inhibited at 100 micromol kg(-1). The oral availability of AFPSiA was 52 +/- 17 and 71 +/- 4% in the dog and rat, respectively. A fraction of AFPSiA was oxidised to the corresponding sulphonate, (2R)-(3-amino-2-fluoropropyl)sulphonic acid (AFPSoA) after oral administration to the rat and dog. In rat brain membranes, AFPSiA was found to have ten times higher affinity for rat brain GABA(B) (K(i) =47 +/- 4.4 nM) compared to GABA(A) (K(i) = 430 +/- 46 nM) binding sites. The compound was a full agonist at human recombinant GABA(B(1a,2)) receptors (EC(50) = 130 +/- 10 nM). In contrast, the metabolite AFPSoA was considerably more selective for binding to rat brain GABA(A) (K(i) = 37 +/- 3.1 nM) vs GABA(B) (K(i) = 6800 +/- 280 nM) receptors. In the mouse, high doses (1-8 mmol kg(-1)) of AFPSiA induced a rapid and mild hypothermia followed by a profound and sustained hypothermia at the higher doses tested (6 and 8 mmol kg(-1)). This effect was unaffected by the selective GABA(B) receptor antagonist CGP62349. AFPSoA (1 and 2 mmol kg(-1)) produced transient and moderate hypothermia while the hypothermic response was considerably larger at 4 mmol kg(-1).It is concluded that AFPSiA inhibits but does not abolish TLOSRs in the dog. High doses of the compound induce hypothermia in the mouse, which probably is attributable to activation of the GABA(A) receptor. The latter effect may be caused both by AFPSiA and its oxidised sulphonic acid metabolite AFPSoA.


Subject(s)
Esophageal Sphincter, Lower/drug effects , GABA Agonists/pharmacology , GABA-B Receptor Agonists , Muscle Relaxation/drug effects , Sulfinic Acids/pharmacology , Animals , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Deglutition/drug effects , Dogs , Esophageal Sphincter, Lower/physiology , Female , GABA-A Receptor Agonists , GABA-B Receptor Antagonists , Hypothermia/chemically induced , Mice , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Sulfinic Acids/adverse effects , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism
6.
Gastroenterology ; 123(4): 1129-34, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12360475

ABSTRACT

BACKGROUND & AIMS: Transient lower esophageal sphincter relaxations (TLESRs) are the major cause of gastroesophageal acid reflux, and are triggered by postprandial gastric distention. Stimulation of GABA(B) receptors potently inhibits triggering of TLESR by gastric loads. The functional similarity between GABA(B) and cannabinoid receptors (CBRs) prompted us to study the role of CBRs on mechanisms of gastric distention-induced TLESRs. METHODS: Gastric nutrient infusion and air insufflation was performed during gastroesophageal manometry in conscious dogs. The effects of the CBR agonist WIN 55,212-2 were assessed alone and in combination with the CBR1 antagonist SR141716A or the CBR2 antagonist SR144528. The effects of WIN 55,212-2 were also studied on firing of gastric vagal mechanosensitive afferents in an isolated preparation of ferret stomach. RESULTS: WIN 55,212-2 (57 nmol/kg) inhibited the occurrence of TLESR after gastric loads by 80% (P < 0.01). The latency to the first TLESR after the load was prolonged (P < 0.001), and the occurrence of swallowing was reduced (P < 0.05). The CBR1 antagonist SR141716A reversed the effects of WIN 55,212-2, whereas the CBR2 antagonist SR144528 did not. The CBR1 antagonist alone increased occurrence of TLESR (P < 0.05). The responses of gastric vagal mechanoreceptors to distention were unaffected by WIN 55,212-2 at a concentration of 3 micromol/L. CONCLUSIONS: Exogenous and endogenous activation of the CBR1 receptor inhibits TLESRs. The effects of CBR1 are not mediated peripherally on gastric vagal afferents, and therefore are most likely in the brain stem.


Subject(s)
Esophagogastric Junction/physiology , Gastroesophageal Reflux/drug therapy , Gastroesophageal Reflux/physiopathology , Receptors, Drug/agonists , Analgesics/pharmacology , Animals , Benzoxazines , Dogs , Esophagogastric Junction/innervation , Female , Ferrets , GABA-B Receptor Agonists , Ligands , Male , Mechanoreceptors/drug effects , Mechanoreceptors/physiology , Morpholines/pharmacology , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Naphthalenes/pharmacology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Rimonabant , Vagus Nerve/cytology , Vagus Nerve/physiology
7.
Eur J Pharmacol ; 448(1): 67-70, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12126973

ABSTRACT

The effects of the GABA(B) receptor agonists baclofen (1.4 and 7 micromol/kg i.v.) and CGP 44532 ([(2S)-3-amino-2-hydroxypropyl]methyl phosphinic acid], 0.2 and 0.7 micromol/kg i.v.) on transient lower esophageal sphincter relaxations and spontaneous and pharyngeally stimulated swallowing were investigated in conscious dogs. Both compounds inhibited transient lower esophageal sphincter relaxations dose-dependently, CGP 44532 being approximately fivefold more potent. In experiments designed to measure transient lower esophageal sphincter relaxations, spontaneous swallowing was suppressed by both compounds. When swallowing was evoked by intrapharyngeal water injection, both baclofen and CGP 44532 reduced the occurrence of primary peristalsis. It is concluded that centrally acting GABA(B) receptor agonists inhibit spontaneous and stimulated swallowing probably through an action in the central pattern generator for swallowing.


Subject(s)
Baclofen/pharmacology , Deglutition/drug effects , Esophagogastric Junction/drug effects , Organophosphonates/pharmacology , Receptors, GABA-B , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology , Animals , Dogs , GABA-B Receptor Agonists , Muscle Relaxation/drug effects , Phosphinic Acids
SELECTION OF CITATIONS
SEARCH DETAIL
...