Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Spine (Phila Pa 1976) ; 45(24): E1636-E1644, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-32947496

RESUMO

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.


Assuntos
Produtos Biológicos/administração & dosagem , Disco Intervertebral/diagnóstico por imagem , Ácido Láctico/administração & dosagem , Dor Lombar/diagnóstico por imagem , Dor Lombar/tratamento farmacológico , Pesquisa Translacional Biomédica/métodos , Animais , Produtos Biológicos/metabolismo , Método Duplo-Cego , Feminino , Humanos , Disco Intervertebral/efeitos dos fármacos , Disco Intervertebral/metabolismo , Ácido Láctico/metabolismo , Dor Lombar/metabolismo , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Núcleo Pulposo/diagnóstico por imagem , Núcleo Pulposo/efeitos dos fármacos , Núcleo Pulposo/metabolismo , Estudos Prospectivos , Suínos , Resultado do Tratamento
2.
PLoS One ; 12(1): e0168226, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28107353

RESUMO

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.


Assuntos
Peso Corporal , Encéfalo/metabolismo , Leptina/metabolismo , Transdução de Sinais , Animais , Glicemia/metabolismo , Composição Corporal , Dieta , Ingestão de Energia , Metabolismo Energético , Homeostase , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G887-98, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27012770

RESUMO

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.


Assuntos
Mucosa Intestinal/metabolismo , Doenças Metabólicas/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Células Enteroendócrinas/metabolismo , Motilidade Gastrointestinal , Humanos , Absorção Intestinal , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/genética , Doenças Metabólicas/microbiologia , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/genética
4.
Clin Ther ; 38(4): 946-60, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26947796

RESUMO

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.


Assuntos
Agonistas de Receptores de GABA-A/efeitos adversos , Parestesia/induzido quimicamente , Ácidos Fosfínicos/efeitos adversos , Propilaminas/efeitos adversos , Agonistas de Receptores de GABA-A/uso terapêutico , Refluxo Gastroesofágico/tratamento farmacológico , Humanos , Ácidos Fosfínicos/uso terapêutico , Propilaminas/uso terapêutico
6.
Obesity (Silver Spring) ; 22(5): 1287-95, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24318934

RESUMO

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.


Assuntos
Peso Corporal/efeitos dos fármacos , Obesidade/tratamento farmacológico , Receptor Tipo 4 de Melanocortina/agonistas , Animais , Glicemia/metabolismo , Composição Corporal , Restrição Calórica , Calorimetria Indireta , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Hormônios/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Redução de Peso
7.
J Pharmacol Exp Ther ; 347(2): 298-309, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24008333

RESUMO

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.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Animais , Células CHO , Cricetulus , Células HEK293 , Humanos , Ligantes , Camundongos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos
8.
J Med Chem ; 56(1): 220-40, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23227781

RESUMO

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.


Assuntos
Benzamidas/síntese química , Encéfalo/metabolismo , Refluxo Gastroesofágico/tratamento farmacológico , Receptor CB1 de Canabinoide/agonistas , Administração Oral , Animais , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Benzamidas/farmacocinética , Benzamidas/farmacologia , Disponibilidade Biológica , Linhagem Celular , Cricetinae , Cricetulus , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A , Inibidores do Citocromo P-450 CYP3A , Cães , Canal de Potássio ERG1 , Esfíncter Esofágico Inferior/efeitos dos fármacos , Esfíncter Esofágico Inferior/fisiologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Ensaios de Triagem em Larga Escala , Humanos , Relaxamento Muscular/efeitos dos fármacos , Pirazinas/síntese química , Pirazinas/farmacocinética , Pirazinas/farmacologia , Piridinas/síntese química , Piridinas/farmacocinética , Piridinas/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Solubilidade , Relação Estrutura-Atividade , Sulfóxidos/síntese química , Sulfóxidos/farmacocinética , Sulfóxidos/farmacologia , Triazóis/síntese química , Triazóis/farmacocinética , Triazóis/farmacologia
9.
Cough ; 8(1): 7, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23025757

RESUMO

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.

10.
Curr Opin Pharmacol ; 11(6): 630-3, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22036168

RESUMO

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.


Assuntos
Esfíncter Esofágico Inferior/efeitos dos fármacos , Agonistas dos Receptores de GABA-B/uso terapêutico , Refluxo Gastroesofágico/tratamento farmacológico , Fármacos Gastrointestinais/uso terapêutico , Ácidos Fosfínicos/uso terapêutico , Propilaminas/uso terapêutico , Pesquisa Translacional Biomédica , Animais , Baclofeno/farmacologia , Baclofeno/uso terapêutico , Cães , Resistência a Medicamentos , Esfíncter Esofágico Inferior/fisiopatologia , Agonistas dos Receptores de GABA-B/farmacologia , Refluxo Gastroesofágico/fisiopatologia , Fármacos Gastrointestinais/farmacologia , Humanos , Relaxamento Muscular/efeitos dos fármacos , Fármacos Neuromusculares/farmacologia , Fármacos Neuromusculares/uso terapêutico , Ácidos Fosfínicos/farmacologia , Propilaminas/farmacologia , Especificidade da Espécie
11.
PLoS One ; 6(4): e19095, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21552517

RESUMO

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.


Assuntos
4-Aminobutirato Transaminase/genética , Refluxo Gastroesofágico/tratamento farmacológico , Refluxo Gastroesofágico/genética , Adolescente , Adulto , Animais , Estudos de Casos e Controles , Cães , Esfíncter Esofágico Inferior/metabolismo , Esfíncter Esofágico Inferior/fisiopatologia , Feminino , Refluxo Gastroesofágico/fisiopatologia , Ligação Genética , Predisposição Genética para Doença , Humanos , Masculino , Reprodutibilidade dos Testes , Análise de Sequência de DNA
12.
Adv Pharmacol ; 58: 287-313, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20655487

RESUMO

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.


Assuntos
Agonistas GABAérgicos/uso terapêutico , Agonistas dos Receptores de GABA-B , Refluxo Gastroesofágico/tratamento farmacológico , Animais , Avaliação Pré-Clínica de Medicamentos , Agonistas GABAérgicos/farmacologia , Refluxo Gastroesofágico/metabolismo , Refluxo Gastroesofágico/fisiopatologia , Humanos , Receptores de GABA-B/química
13.
Eur J Pharmacol ; 634(1-3): 138-41, 2010 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-20176012

RESUMO

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.


Assuntos
Esôfago/efeitos dos fármacos , Agonistas GABAérgicos/uso terapêutico , Agonistas dos Receptores de GABA-B , Refluxo Gastroesofágico/metabolismo , Refluxo Gastroesofágico/prevenção & controle , Ácidos Fosfínicos/uso terapêutico , Propilaminas/uso terapêutico , Animais , Baclofeno/administração & dosagem , Baclofeno/farmacologia , Cães , Esôfago/fisiologia , Feminino , Agonistas GABAérgicos/farmacologia , Ácido Gástrico/metabolismo , Refluxo Gastroesofágico/fisiopatologia , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Masculino , Manometria/métodos , Ácidos Fosfínicos/farmacologia , Projetos Piloto , Propilaminas/farmacologia , Receptores de GABA-B/fisiologia , Fatores de Tempo
14.
J Pharmacol Exp Ther ; 331(2): 504-12, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19648470

RESUMO

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.


Assuntos
Esfíncter Esofágico Inferior/efeitos dos fármacos , Agonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B , Nervos Periféricos/efeitos dos fármacos , Ácidos Fosfínicos/farmacologia , Propilaminas/farmacologia , Animais , Autorradiografia , Baclofeno/farmacologia , Ligação Competitiva/efeitos dos fármacos , Cálcio/metabolismo , Cães , Relação Dose-Resposta a Droga , Esfíncter Esofágico Inferior/inervação , Feminino , Furões/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Humanos , Hipotermia/induzido quimicamente , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Relaxamento Muscular/efeitos dos fármacos , Ligação Proteica , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de GABA-B/metabolismo , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
15.
Pharmacol Ther ; 122(3): 239-45, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19303900

RESUMO

For many years, acid-suppressive therapy has been at the forefront of treating gastroesophageal reflux disease (GERD), yet despite the advent of the proton pump inhibitors (PPIs) some patients continue to experience persistent GERD symptoms. Therapeutic (non-surgical) options for such patients are currently limited. To tackle this clinical issue, research efforts have begun to focus on 'reflux inhibition' as a potential therapeutic target - i.e. inhibition of transient lower esophageal relaxations (TLESRs), the predominant mechanism of gastroesophageal reflux. Preclinical research has identified a number of drug targets through which TLESRs can be modulated, and the gamma-aminobutyric acid (GABA) type B (GABA(B)) receptor has emerged as one of the most promising. Studies with baclofen, a well-known agonist of this receptor, have demonstrated that reflux inhibition is a valid concept in the clinical setting in that reducing the incidence of TLESRs improves GERD symptoms. But baclofen is associated with significant central nervous system (CNS) side effects, rendering it undesirable for use as a treatment for GERD. Further development work has yielded a number of novel GABA(B) receptor agonists with reduced CNS side effect profiles, and clinical trials are currently being performed with several agents. Compounds that target TLESRs may therefore present a new add-on treatment for patients with persistent GERD symptoms despite PPI therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Agonistas GABAérgicos/administração & dosagem , Refluxo Gastroesofágico/tratamento farmacológico , Fármacos Gastrointestinais/administração & dosagem , Receptores de GABA-B , Animais , Refluxo Gastroesofágico/fisiopatologia , Humanos , Receptores de GABA-B/fisiologia
16.
J Med Chem ; 51(14): 4315-20, 2008 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-18578471

RESUMO

We have previously demonstrated that the prototypical GABA B receptor agonist baclofen inhibits transient lower esophageal sphincter relaxations (TLESRs), the most important mechanism for gastroesophageal reflux. Thus, GABA B agonists could be exploited for the treatment of gastroesophageal reflux disease. However, baclofen, which is used as an antispastic agent, and other previously known GABA B agonists can produce CNS side effects such as sedation, dizziness, nausea, and vomiting at higher doses. We now report the discovery of atypical GABA B agonists devoid of classical GABA B agonist related CNS side effects at therapeutic doses and the optimization of this type of compound for inhibition of TLESRs, which has resulted in a candidate drug ( R)- 7 (AZD3355) that is presently being evaluated in man.


Assuntos
Agonistas GABAérgicos/química , Agonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B , Refluxo Gastroesofágico/tratamento farmacológico , Animais , Relação Dose-Resposta a Droga , Agonistas GABAérgicos/uso terapêutico , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas de Bombardeamento Rápido de Átomos
17.
Gastroenterology ; 129(3): 995-1004, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16143137

RESUMO

BACKGROUND & AIMS: Transient lower esophageal sphincter relaxation (TLESR) is the major mechanism of gastroesophageal acid reflux. TLESR is mediated via vagal pathways, which may be modulated by metabotropic glutamate receptors (mGluRs). Group I mGluRs (mGluR1 and 5) have excitatory effects on neurons, whereas group II (mGluR2 and 3) and group III (mGluR4, 6, 7, and 8) are inhibitory. This study determined the effect of mGluRs on triggering of TLESR and reflux in an established conscious ferret model. METHODS: Esophageal manometric/pH studies were performed in ferrets with chronic esophagostomies. TLESR were induced by a gastric load of 25 mL glucose (pH 3.5) and 30 mL air. RESULTS: In control treated animals, gastric load induced 3.52 +/- 0.46 TLESRs per 47-minute study, 89.7% of which were associated with reflux episodes (n = 16). The mGluR5 antagonist MPEP inhibited TLESR dose dependently, with maximal 71% +/- 7% inhibition at 35 micromol/kg (n = 9; P < .0001). MPEP also significantly reduced reflux episodes (P < .001) and increased basal lower esophageal sphincter pressure (P < .05). MPEP inhibited swallowing dose dependently, suggesting a common action on trigger mechanisms for swallowing and TLESR. The more selective analogue, MTEP, had more potent effects (90% +/- 6% inhibition TLESR at 40 micromol/kg; n = 8; P < .0001). In contrast, the group I agonist DHPG tended to increase TLESR. The group II agonist (2R, 4R)-APDC was ineffective, whereas the group III agonist L-(AP4 slightly reduced TLESR (33% at 11 micromol/kg; P < .05). The selective mGluR8 agonist (S)-3, 4-DCPG inhibited TLESR by 54% at 15 micromol/kg (P < .01). CONCLUSIONS: mGluR5 antagonists potently inhibit TLESR and reflux in ferrets, implicating mGluR5 in the mechanism of TLESR. mGluR5 antagonists are therefore promising as therapy for patients with GERD.


Assuntos
Esôfago/fisiologia , Refluxo Gastroesofágico/prevenção & controle , Receptores de Glutamato Metabotrópico/fisiologia , Animais , Deglutição/fisiologia , Modelos Animais de Doenças , Feminino , Furões , Ligantes , Relaxamento Muscular , Músculo Liso/fisiologia , Pressão , Piridinas/farmacologia , Piridinas/uso terapêutico , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Tiazóis/farmacologia
18.
Eur J Pharmacol ; 519(1-2): 154-7, 2005 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16102747

RESUMO

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.


Assuntos
Esfíncter Esofágico Inferior/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Relaxamento Muscular/efeitos dos fármacos , Piridinas/farmacologia , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Animais , Sítios de Ligação , Ligação Competitiva , Cães , Relação Dose-Resposta a Droga , Esfíncter Esofágico Inferior/fisiologia , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/metabolismo , Feminino , Refluxo Gastroesofágico/fisiopatologia , Refluxo Gastroesofágico/prevenção & controle , Injeções Intravenosas , Masculino , Piridinas/administração & dosagem , Piridinas/metabolismo , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/metabolismo , Trítio
19.
Br J Pharmacol ; 146(1): 89-97, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15980875

RESUMO

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.


Assuntos
Esfíncter Esofágico Inferior/efeitos dos fármacos , Agonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B , Relaxamento Muscular/efeitos dos fármacos , Ácidos Sulfínicos/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , Deglutição/efeitos dos fármacos , Cães , Esfíncter Esofágico Inferior/fisiologia , Feminino , Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-B , Hipotermia/induzido quimicamente , Camundongos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de GABA-A/metabolismo , Receptores de GABA-B/metabolismo , Ácidos Sulfínicos/efeitos adversos , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo
20.
Gastroenterology ; 128(2): 402-10, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15685551

RESUMO

BACKGROUND AND AIMS: Inhibitory G-protein-coupled receptors have demonstrated potential in treatment of gastroesophageal reflux disease (GERD) through actions on vagal afferent signaling. Metabotropic glutamate receptors (mGluR) belong to this receptor family and have great pharmacologic and molecular diversity, with 8 subtypes. We investigated mGluR in the vagal system of humans and other species. METHODS: Expression of mGluR1-8 in human, dog, ferret, and rodent nodose ganglia was investigated by reverse-transcription polymerase chain reaction. mGluR1-8 immunohistochemistry was performed in combination with retrograde tracing of vagal afferents from ferret proximal stomach to nodose ganglia. Transport of mGluR peripherally was investigated by vagal ligation, followed by immunohistochemistry. Glutamate receptor pharmacology of ferret and rodent gastroesophageal vagal afferents was investigated by testing single fiber responses to graded mechanical stimuli during drug application to their peripheral endings. RESULTS: Messenger RNA for several mGluR was detected in the nodose ganglia of all species. Retrograde tracing indicated that ferret gastric vagal afferents express mGluR protein. Accumulation of immunoreactivity proximal to a ligature showed that mGluR were transported peripherally in the vagus nerves. Glutamate (1-30 mumol/L with kynurenate 0.1 mmol/L) concentration dependently inhibited vagal afferent mechanosensitivity. This was mimicked by selective group II and III mGluR agonists but not by a group I agonist. Conversely, a group III mGluR antagonist increased mechanosensitivity to intense stimuli. CONCLUSIONS: Both exogenous and endogenous glutamate inhibits mechanosensitivity of vagal afferents. Group II (mGluR2 and 3) and group III mGluR (mGluR4, 6, 7, 8) are novel targets for inhibition of vagal signaling with therapeutic potential in, for example, GERD.


Assuntos
Mecanorreceptores/fisiologia , Neurônios Aferentes/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/fisiologia , Animais , Sequência de Bases , Primers do DNA , Cães , Humanos , Gânglio Nodoso/fisiologia , Reação em Cadeia da Polimerase , Ratos , Receptores de Glutamato Metabotrópico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...