Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G887-98, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27012770

ABSTRACT

The Na(+)-glucose cotransporter 1 (SGLT1/SLC5A1) is predominantly expressed in the small intestine. It transports glucose and galactose across the apical membrane in a process driven by a Na(+) gradient created by Na(+)-K(+)-ATPase. SGLT2 is the major form found in the kidney, and SGLT2-selective inhibitors are a new class of treatment for type 2 diabetes mellitus (T2DM). Recent data from patients treated with dual SGLT1/2 inhibitors or SGLT2-selective drugs such as canagliflozin (SGLT1 IC50 = 663 nM) warrant evaluation of SGLT1 inhibition for T2DM. SGLT1 activity is highly dynamic, with modulation by multiple mechanisms to ensure maximal uptake of carbohydrates (CHOs). Intestinal SGLT1 inhibition lowers and delays the glucose excursion following CHO ingestion and augments glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) secretion. The latter is likely due to increased glucose exposure of the colonic microbiota and formation of metabolites such as L cell secretagogues. GLP-1 and PYY secretion suppresses food intake, enhances the ileal brake, and has an incretin effect. An increase in colonic microbial production of propionate could contribute to intestinal gluconeogenesis and mediate positive metabolic effects. On the other hand, a threshold of SGLT1 inhibition that could lead to gastrointestinal intolerability is unclear. Altered Na(+) homeostasis and increased colonic CHO may result in diarrhea and adverse gastrointestinal effects. This review considers the potential mechanisms contributing to positive metabolic and negative intestinal effects. Compounds that inhibit SGLT1 must balance the modulation of these mechanisms to achieve therapeutic efficacy for metabolic diseases.


Subject(s)
Intestinal Mucosa/metabolism , Metabolic Diseases/metabolism , Sodium-Glucose Transporter 1/metabolism , Animals , Enteroendocrine Cells/metabolism , Gastrointestinal Motility , Humans , Intestinal Absorption , Metabolic Diseases/drug therapy , Metabolic Diseases/genetics , Metabolic Diseases/microbiology , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 1/genetics
2.
J Pharmacol Exp Ther ; 347(2): 298-309, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008333

ABSTRACT

The GPRC6A receptor is a recently "deorphanized" class C G protein-coupled receptor. We and others have shown that this receptor is coactivated by basic l-α-amino acids and divalent cations, whereas other groups have also suggested osteocalcin and testosterone to be agonists. Likewise, the GPRC6A receptor has been suggested to couple to multiple G protein classes albeit via indirect methods. Thus, the exact ligand preferences and signaling pathways are yet to be elucidated. In the present study, we generated a Chinese hamster ovary (CHO) cell line that stably expresses mouse GPRC6A. In an effort to establish fully the signaling properties of the receptor, we tested representatives of four previously reported GPRC6A agonist classes for activity in the Gq, Gs, Gi, and extracellular-signal regulated kinase signaling pathways. Our results confirm that GPRC6A is activated by basic l-α-amino acids and divalent cations, and for the first time, we conclusively show that these responses are mediated through the Gq pathway. We were not able to confirm previously published data demonstrating Gi- and Gs-mediated signaling; neither could we detect agonistic activity of testosterone and osteocalcin. Generation of the stable CHO cell line with robust receptor responsiveness and optimization of the highly sensitive homogeneous time resolved fluorescence technology allow fast assessment of Gq activation without previous manipulations like cotransfection of mutated G proteins. This cell-based assay system for GPRC6A is thus useful in high-throughput screening for novel pharmacological tool compounds, which are necessary to unravel the physiologic function of the receptor.


Subject(s)
High-Throughput Screening Assays/methods , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , CHO Cells , Cricetulus , HEK293 Cells , Humans , Ligands , Mice , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects
3.
Spine (Phila Pa 1976) ; 45(24): E1636-E1644, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32947496

ABSTRACT

STUDY DESIGN: Preclinical studies: Efficacy and toxicological studies on lactic acid (LA)-induced sclerozation in pig lumbar discs. Clinical study: Prospective, randomized, double-blinded, placebo-controlled, single ascending dose study investigating the safety and local tolerability of LA. OBJECTIVE: To determine if LA produces sclerozation of the porcine nucleus pulposus (NP) followed by a phase Ib study to evaluate preliminary safety, tolerability, and efficacy of LA in patients with chronic discogenic low back pain. SUMMARY OF BACKGROUND DATA: Surgical stabilization of a motion segment harboring a painful degenerated disc often affords symptomatic relief. In the present study, the hypothesis was tested that LA can produce sclerozation and stabilization of the NP. METHODS: LA (0.2 mL; 60, 120, or 240 mg/mL) or vehicle was injected into the NP or close to the extra spinal region of spinal nerves of young female pigs. The size of the NP, MRI changes, flexural stiffness, and histology of the disc was studied after up to 84 days of survival. Fifteen patients injected intra discally with placebo (iohexol, 1.5 mL, n = 6) or iohexol plus LA (30, 60, or 120 mg/mL; three patients in each group) were followed for up to 12 months. RESULTS: Injection of LA in the pig reproducibly induced sclerozation of the NP and increased flexural rigidity. Histological changes included generation of connective tissue and increased expression of collagen I. No safety concerns were raised. Adverse events in patients were limited to transiently increased low back pain with no obvious difference between treatment groups. There was indication of lower water content of NP injected with the two highest doses of LA. CONCLUSION: LA has a sclerozing effect on the NP in pigs and patients and is therefore a candidate for further clinical studies powered to determine its potential as a treatment of chronic discogenic low back pain. LEVEL OF EVIDENCE: 2.


Subject(s)
Biological Products/administration & dosage , Intervertebral Disc/diagnostic imaging , Lactic Acid/administration & dosage , Low Back Pain/diagnostic imaging , Low Back Pain/drug therapy , Translational Research, Biomedical/methods , Animals , Biological Products/metabolism , Double-Blind Method , Female , Humans , Intervertebral Disc/drug effects , Intervertebral Disc/metabolism , Lactic Acid/metabolism , Low Back Pain/metabolism , Magnetic Resonance Imaging/methods , Male , Middle Aged , Nucleus Pulposus/diagnostic imaging , Nucleus Pulposus/drug effects , Nucleus Pulposus/metabolism , Prospective Studies , Swine , Treatment Outcome
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.
PLoS One ; 12(1): e0168226, 2017.
Article in English | MEDLINE | ID: mdl-28107353

ABSTRACT

Diet-induced obesity (DIO) resulting from consumption of a high fat diet (HFD) attenuates normal neuronal responses to leptin and may contribute to the metabolic defense of an acquired higher body weight in humans; the molecular bases for the persistence of this defense are unknown. We measured the responses of 23 brain regions to exogenous leptin in 4 different groups of weight- and/or diet-perturbed mice. Responses to leptin were assessed by quantifying pSTAT3 levels in brain nuclei 30 minutes following 3 mg/kg intraperitoneal leptin. HFD attenuated leptin sensing throughout the brain, but weight loss did not restore central leptin signaling to control levels in several brain regions important in energy homeostasis, including the arcuate and dorsomedial hypothalamic nuclei. Effects of diet on leptin signaling varied by brain region, with results dependent on the method of weight loss (restriction of calories of HFD, ad lib intake of standard mouse chow). High fat diet attenuates leptin signaling throughout the brain, but some brain regions maintain their ability to sense leptin. Weight loss restores leptin sensing to some degree in most (but not all) brain regions, while other brain regions display hypersensitivity to leptin following weight loss. Normal leptin sensing was restored in several brain regions, with the pattern of restoration dependent on the method of weight loss.


Subject(s)
Body Weight , Brain/metabolism , Leptin/metabolism , Signal Transduction , Animals , Blood Glucose/metabolism , Body Composition , Diet , Energy Intake , Energy Metabolism , Homeostasis , Insulin/blood , Male , Mice , Mice, Inbred C57BL
6.
Clin Ther ; 38(4): 946-60, 2016 04.
Article in English | MEDLINE | ID: mdl-26947796

ABSTRACT

PURPOSE: Lesogaberan, a γ-aminobutyric acid (GABA)B receptor agonist, was developed for the treatment of gastroesophageal reflux disease in patients with a partial response to proton pump inhibitor therapy. A high prevalence of paresthesia was observed in healthy individuals after dosing with lesogaberan in early-phase clinical trials. The aim of this review was to gain further insight into paresthesia caused by lesogaberan by summarizing the relevant preclinical and clinical data. METHODS: This study was a narrative review of the literature and unpublished data. FINDINGS: The occurrence of paresthesia may depend on the route or rate of drug administration; several studies were conducted to test this hypothesis, and formulations were developed to minimize the occurrence of paresthesia. Phase I clinical studies showed that, in healthy individuals, paresthesia occurred soon after administration of lesogaberan in a dose-dependent manner regardless of the route of administration. The occurrence of paresthesia could be decreased by fractionating the dose or reducing the rate of administration. These findings suggest that the initial rate of absorption plays an important part in the development of paresthesia. Modified-release formulations minimize the occurrence of paresthesia while retaining the anti-reflux activity of the drug, as measured by esophageal pH and the number of transient lower esophageal sphincter relaxations. IMPLICATIONS: The development of lesogaberan was halted because the effect on gastroesophageal reflux disease symptoms observed in Phase II studies was not considered clinically meaningful in the target patient population. Nevertheless, it is an example of successful formulation development designed to minimize the occurrence of a compound's adverse effect while retaining its pharmacodynamic action.


Subject(s)
GABA-A Receptor Agonists/adverse effects , Paresthesia/chemically induced , Phosphinic Acids/adverse effects , Propylamines/adverse effects , GABA-A Receptor Agonists/therapeutic use , Gastroesophageal Reflux/drug therapy , Humans , Phosphinic Acids/therapeutic use , Propylamines/therapeutic use
7.
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
8.
Eur J Pharmacol ; 519(1-2): 154-7, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16102747

ABSTRACT

Transient lower esophageal sphincter relaxation is the major mechanism for gastroesophageal reflux. The present study was initiated to investigate the potential effect of the metabotropic glutamate 5 (mGlu5) receptor antagonist, 2-methyl-6-(phenylethynyl)-pyridine (MPEP), on transient lower esophageal sphincter relaxations in the conscious dog. MPEP (1.4-8.7 micromol/kg i.v.) produced a dose-dependent inhibition of transient lower esophageal sphincter relaxations (59+/-11% inhibition at 8.7 micromol/kg). In addition, there was a reduction of the number of reflux episodes and an increase in latency time to the occurrence of the first transient lower esophageal sphincter relaxation. No effect was seen on basal lower esophageal sphincter pressure or on swallowing. It is concluded that the mGlu5 receptor antagonist MPEP potently inhibits transient lower esophageal sphincter relaxations and that the mGlu5 receptor is a potential target for treatment of gastroesophageal reflux disease.


Subject(s)
Esophageal Sphincter, Lower/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Muscle Relaxation/drug effects , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Binding Sites , Binding, Competitive , Dogs , Dose-Response Relationship, Drug , Esophageal Sphincter, Lower/physiology , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Female , Gastroesophageal Reflux/physiopathology , Gastroesophageal Reflux/prevention & control , Injections, Intravenous , Male , Pyridines/administration & dosage , Pyridines/metabolism , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism , Tritium
9.
Eur J Neurosci ; 2(7): 620-628, 1990.
Article in English | MEDLINE | ID: mdl-12106296

ABSTRACT

Glutamate (glu) is a major excitatory transmitter and a toxin in the brain. In the present study, the immature rat hippocampal slice was used to determine the morphology, topography, ionic mediation and receptor specificity of glu toxicity. Slices were exposed to glu for 30 min, and the damage was evaluated after 3 h of recovery in regular medium. The effects on glu toxicity of changes of [Ca2+], [Cl-] and [Na+] were determined. The receptor preference of glu was assessed by using the N-methyl-D-aspartate (NMDA) antagonist MK-801 and the kainate (KA)/quisqualate (QA) antagonist DNQX, alone or in combination. Further, to see whether glu produces cytotoxicity via osmolysis, the effects of hyperosmolal sucrose on glu toxicity were studied. Glu toxicity was similar to the previously described NMDA toxicity with regard to cytopathology, but differed in some aspects from that caused by KA and QA. The severity of the lesion was determined by the proximity of neurons to the incubation fluid, probably as a consequence of cellular accumulation of the amino acid. Omission of Ca2+ abolished glu toxicity in all neurons except the granule cells of the outer blade. This population was completely protected when Ca2+ was omitted and [Cl-] was reduced. Elevation of [Ca2+] markedly aggravated the lesion caused by glu. Substitution of isethionate for Cl- worsened the glu-induced damage, whilst the amino acid produced qualitatively different neuropathology when choline substituted for Na+. Apparently glu did not damage hippocampal nerve cells through an osmolytic mechanism as medium supplemented with 100 mM sucrose increased the toxicity of glu. Since the lesion produced by glu was more widespread in the presence of high [Ca2+], the effects of receptor antagonists were studied under this condition. MK-801 inhibited glu toxicity whereas DNQX had no effect. Combination of MK-801 and DNQX did not offer better protection than did MK-801 alone. The results suggest that Ca2+ is the main (but not single) determinant of glu toxicity in the immature hippocampal slice. The ionic requirements of glu neurotoxicity are identical to those of NMDA, but differ from those of KA and QA. The notion that glu is a selective NMDA agonist in the present model was confirmed by the protection of MK-801, and by the lack of an effect of DNQX. This is the first report demonstrating that the toxicity of glu is mediated by NMDA receptors in brain tissue which has developed normally. The findings indicate that specific blockade of NMDA receptors may be the most rational strategy in the prevention of glu-related neuronal death occurring in certain neurological anomalies.

10.
Br J Pharmacol ; 140(2): 315-22, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970075

ABSTRACT

1. Activation of GABA(B) receptors evokes hypothermia in wildtype (GABA(B(1))+/+) but not in GABA(B) receptor knockout (GABA(B(1))-/-) mice. The aim of the present study was to determine the hypothermic and behavioural effects of the putative GABA(B) receptor agonist gamma-hydroxybutyrate (GHB), and of the GABA(A) receptor agonist muscimol. In addition, basal body temperature was determined in GABA(B(1))+/+, GABA(B(1))+/- and GABA(B(1))-/- mice. 2. GABA(B(1))-/- mice were generated by homologous recombination in embryonic stem cells. Correct gene targeting was assessed by Southern blotting, PCR and Western blotting. GABA(B) receptor-binding sites were quantified with radioligand binding. Measurement of body temperature was done using subcutaneous temperature-sensitive chips, and behavioural changes after drug administration were scored according to a semiquantitative scale. 3. GABA(B(1))-/- mice had a short lifespan, probably caused by generalised seizure activity. No histopathological or blood chemistry changes were seen, but the expression of GABA(B(2)) receptor protein was below the detection limit in brains from GABA(B(1))-/- mice, in the absence of changes in mRNA levels. 4. GABA(B) receptor-binding sites were absent in brain membranes from GABA(B(1))-/- mice. 5. GABA(B(1))-/- mice were hypothermic by approximately 1 degrees C compared to GABA(B(1))+/+ and GABA(B(1))+/- mice. 6. Injection of baclofen (9.6 mg kg-1) produced a large reduction in body temperature and behavioural effects in GABA(B(1))+/+ and in GABA(B(1))+/- mice, but GABA(B(1))-/- mice were unaffected. The same pattern was seen after administration of GHB (400 mg kg-1). The GABA(A) receptor agonist muscimol (2 mg kg-1), on the other hand, produced a more pronounced hypothermia in GABA(B(1))-/-mice. In GABA(B(1))+/+ and GABA(B(1))+/- mice, muscimol induced sedation and reduced locomotor activity. However, when given to GABA(B(1))-/- mice, muscimol triggered periods of intense jumping and wild running. 7. It is concluded that hypothermia should be added to the characteristics of the GABAB(1)-/-phenotype. Using this model, GHB was shown to be a selective GABAB receptor agonist. In addition, GABAB(1)-/- mice are hypersensitive to GABAA receptor stimulation, indicating that GABAB tone normally balances GABAA-mediated effects.


Subject(s)
Body Temperature Regulation/drug effects , GABA Agonists/pharmacology , GABA-B Receptor Agonists , Animals , Baclofen/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Cloning, Molecular , GABA-A Receptor Agonists , Gene Expression Regulation/drug effects , Genotype , Hypothermia/chemically induced , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Muscimol/pharmacology , Phenotype , Protein Subunits/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Receptors, GABA-B/genetics , Receptors, GABA-B/physiology , Sodium Oxybate/pharmacology
11.
Curr Opin Investig Drugs ; 5(7): 760-5, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15298074
12.
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
13.
Obesity (Silver Spring) ; 22(5): 1287-95, 2014 May.
Article in English | MEDLINE | ID: mdl-24318934

ABSTRACT

OBJECTIVE: The physiology of the weight-reduced (WR) state suggests that pharmacologic agents affecting energy homeostasis may have greater efficacy in WR individuals. Our aim was to establish a protocol that allows for evaluation of efficacy of weight maintenance agents and to assess the effectiveness of AZD2820, a novel melanocortin 4 receptor (MC4R) agonist in such a paradigm. METHODS: MC4R agonist was administered in stratified doses to mice who were either fed high-fat diet ad libitum (AL) throughout the study; or stabilized at a 20% reduced body weight (BW), administered the drug for 4 weeks, and thereafter released from caloric restriction while continuing to receive the drug (WR). RESULTS: After release of WR mice to AL feeding, the high-dose group (53.4 nmol/day) regained 12.4% less BW than their vehicle-treated controls since the beginning of drug treatment. In WR mice, 10.8 nmol/day of the agonist was sufficient to maintain these animals at 95.1% of initial BW versus 53.4 nmol/day required to maintain the BW of AL animals (94.5%). CONCLUSIONS: In the WR state, the MC4R agonist was comparably efficacious to a five-fold higher dose in the AL state. This protocol provides a model for evaluating the mechanisms and quantitative efficacy of weight-maintenance strategies and agents.


Subject(s)
Body Weight/drug effects , Obesity/drug therapy , Receptor, Melanocortin, Type 4/agonists , Animals , Blood Glucose/metabolism , Body Composition , Caloric Restriction , Calorimetry, Indirect , Diet, High-Fat/adverse effects , Energy Metabolism , Hormones/blood , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Weight Loss
14.
J Med Chem ; 56(1): 220-40, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23227781

ABSTRACT

Agonists of the cannabinoid receptor 1 (CB1) have been suggested as possible treatments for a range of medical disorders including gastroesophageal reflux disease (GERD). While centrally acting cannabinoid agonists are known to produce psychotropic effects, it has been suggested that the CB1 receptors in the periphery could play a significant role in reducing reflux. A moderately potent and highly lipophilic series of 2-aminobenzamides was identified through focused screening of GPCR libraries. Development of this series focused on improving potency and efficacy at the CB1 receptor, reducing lipophilicity and limiting the central nervous system (CNS) exposure while maintaining good oral absorption. Improvement of the series led to compounds having excellent potency at the CB1 receptor and high levels of agonism, good physical and pharmacokinetic properties, and low penetration into the CNS. A range of compounds demonstrated a dose-dependent inhibition of transient lower esophageal sphincter relaxations in a dog model.


Subject(s)
Benzamides/chemical synthesis , Brain/metabolism , Gastroesophageal Reflux/drug therapy , Receptor, Cannabinoid, CB1/agonists , Administration, Oral , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Benzamides/pharmacokinetics , Benzamides/pharmacology , Biological Availability , Cell Line , Cricetinae , Cricetulus , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Dogs , ERG1 Potassium Channel , Esophageal Sphincter, Lower/drug effects , Esophageal Sphincter, Lower/physiology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , High-Throughput Screening Assays , Humans , Muscle Relaxation/drug effects , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Solubility , Structure-Activity Relationship , Sulfoxides/chemical synthesis , Sulfoxides/pharmacokinetics , Sulfoxides/pharmacology , Triazoles/chemical synthesis , Triazoles/pharmacokinetics , Triazoles/pharmacology
15.
Cough ; 8(1): 7, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23025757

ABSTRACT

BACKGROUND: Gastroesophageal reflux disease (GERD) is a common cause of chronic cough. Both acid and nonacid reflux is thought to play a role in the initiation of coughing and cough hypersensitivity. The GABAB receptor agonist lesogaberan was developed as a peripherally restricted anti-reflux therapy that reduces the frequency of transient lower esophageal sphincter relaxations (TLESR; the major cause of reflux) in animals and in patients with GERD. GABAB receptor agonists have also been shown to possess antitussive effects in patients and in animals independent of their effects on TLESR, suggesting that lesogaberan may be a promising treatment for chronic cough. METHODS: We have assessed the direct antitussive effects of lesogaberan (AZD3355). The effects of other GABAB receptor agonists were also determined. Coughing was evoked in awake guinea pigs using aerosol challenges with citric acid. RESULTS: Lesogaberan dose-dependently inhibited citric acid evoked coughing in guinea pigs. Comparable effects of the GABAB receptor agonists baclofen and 3-aminopropylphosphinic acid (3-APPiA) on cough were also observed. Baclofen produced obvious signs of sedation and respiratory depression. By contrast, both lesogaberan and 3-APPiA (both inactivated centrally by GABA transporters) were devoid of sedative effects and did not alter respiratory rate. CONCLUSIONS: Together, the data suggest that lesogaberan and related GABAB receptor agonists may hold promise as safe and effective antitussive agents largely devoid of CNS side effects.

16.
Curr Opin Pharmacol ; 11(6): 630-3, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22036168

ABSTRACT

The development of the novel γ-aminobutyric acid type-B receptor (GABAB) agonist lesogaberan is presented as an example of a partly successful translational strategy in the field of gastroenterology. Data on transient lower esophageal sphincter relaxations (TLESRs) and gastroesophageal reflux inhibition from preclinical models translated well to clinical studies in healthy volunteers and patients with gastroesophageal reflux disease (GERD). Animal models have also been used successfully to predict the effect of other target mechanisms on TLESRs in humans. However, while translation of physiology to symptomatology in patients with GERD was achieved, the effect size was too small to be of clinical significance. A deeper understanding of the cause of symptoms in different patient categories is therefore required.


Subject(s)
Esophageal Sphincter, Lower/drug effects , GABA-B Receptor Agonists/therapeutic use , Gastroesophageal Reflux/drug therapy , Gastrointestinal Agents/therapeutic use , Phosphinic Acids/therapeutic use , Propylamines/therapeutic use , Translational Research, Biomedical , Animals , Baclofen/pharmacology , Baclofen/therapeutic use , Dogs , Drug Resistance , Esophageal Sphincter, Lower/physiopathology , GABA-B Receptor Agonists/pharmacology , Gastroesophageal Reflux/physiopathology , Gastrointestinal Agents/pharmacology , Humans , Muscle Relaxation/drug effects , Neuromuscular Agents/pharmacology , Neuromuscular Agents/therapeutic use , Phosphinic Acids/pharmacology , Propylamines/pharmacology , Species Specificity
17.
PLoS One ; 6(4): e19095, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21552517

ABSTRACT

Gastro-esophageal reflux disease (GERD) is partly caused by genetic factors. The underlying susceptibility genes are currently unknown, with the exception of COL3A1. We used three independent GERD patient cohorts to identify GERD susceptibility genes. Thirty-six families, demonstrating dominant transmission of GERD were subjected to whole genome microsatellite genotyping and linkage analysis. Five linked regions were identified. Two families shared a linked region (LOD 3.9 and 2.0) on chromosome 16. We used two additional independent GERD patient cohorts, one consisting of 219 trios (affected child with parents) and the other an adult GERD case control cohort consisting of 256 cases and 485 controls, to validate individual genes in the linked region through association analysis. Sixty six single nucleotide polymorphism (SNP) markers distributed over the nine genes present in the linked region were genotyped in the independent GERD trio cohort. Transmission disequilibrium test analysis followed by multiple testing adjustments revealed a significant genetic association for one SNP located in an intron of the gene 4-aminobutyrate aminotransferase (ABAT) (P(adj) = 0.027). This association did not replicate in the adult case-control cohort, possibly due to the differences in ethnicity between the cohorts. Finally, using the selective ABAT inhibitor vigabatrin (γ-vinyl GABA) in a dog study, we were able to show a reduction of transient lower esophageal sphincter relaxations (TLESRs) by 57.3 ± 11.4 % (p = 0.007) and the reflux events from 3.1 ± 0.4 to 0.8 ± 0.4 (p = 0.007). Our results demonstrate the direct involvement of ABAT in pathways affecting lower esophageal sphincter (LES) control and identifies ABAT as a genetic risk factor for GERD.


Subject(s)
4-Aminobutyrate Transaminase/genetics , Gastroesophageal Reflux/drug therapy , Gastroesophageal Reflux/genetics , Adolescent , Adult , Animals , Case-Control Studies , Dogs , Esophageal Sphincter, Lower/metabolism , Esophageal Sphincter, Lower/physiopathology , Female , Gastroesophageal Reflux/physiopathology , Genetic Linkage , Genetic Predisposition to Disease , Humans , Male , Reproducibility of Results , Sequence Analysis, DNA
19.
Adv Pharmacol ; 58: 287-313, 2010.
Article in English | MEDLINE | ID: mdl-20655487

ABSTRACT

Defined pharmacologically by its insensitivity to the GABA(A) antagonist bicuculline and sensitivity to the GABA analogue baclofen, the G protein-linked gamma-aminobutyric acid type B (GABA(B)) receptor couples to adenylyl cyclase, voltage-gated calcium channels, and inwardly-rectifying potassium channels. On the basis of a wealth of preclinical data in conjunction with early clinical observations that baclofen improves symptoms of gastroesophageal reflux disease (GERD), the GABA(B) receptor has been proposed as a therapeutic target for a number of diseases including GERD. Subsequently, there has been a significant effort to develop a peripherally-restricted GABA(B) agonist that is devoid of the central nervous system side effects that are observed with baclofen. In this article we review the in vitro and in vivo pharmacology of the peripherally-restricted GABA(B) receptor agonists and the preclinical and clinical development of lesogaberan (AZD3355, (R)-(3-amino-2-fluoropropyl) phosphinic acid), a potent and predominately peripherally-restricted GABA(B) receptor agonist with a preclinical therapeutic window superior to baclofen.


Subject(s)
GABA Agonists/therapeutic use , GABA-B Receptor Agonists , Gastroesophageal Reflux/drug therapy , Animals , Drug Evaluation, Preclinical , GABA Agonists/pharmacology , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/physiopathology , Humans , Receptors, GABA-B/chemistry
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL