Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Expert Opin Ther Targets ; 26(9): 811-822, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36424892

RESUMEN

INTRODUCTION: The Helping to End Addiction Long-termSM Initiative supports a wide range of programs to develop new or improved prevention and opioid addiction treatment strategies. An essential component of this effort is to accelerate development of non-opioid pain therapeutics. In all fields of medicine, therapeutics development is an arduous process and late-stage translational efforts such as clinical trials to validate targets are particularly complex and costly. While there are plentiful novel targets for pain treatment, successful clinical validation is rare. It is therefore crucial to develop processes whereby therapeutic targets can be reasonably 'de-risked' prior to substantial late-stage validation efforts. Such rigorous validation of novel therapeutic targets in the preclinical space will give potential private sector partners the confidence to pursue clinical validation of promising therapeutic concepts and compounds. AREAS COVERED: In 2020, the National Institutes of Health (NIH) held the Target Validation for Non-Addictive Therapeutics Development for Pain workshop to gather insights from key opinion leaders in academia, industry, and venture-financing. EXPERT OPINION: The result was a roadmap for pain target validation focusing on three modalities: 1) human evidence; 2) assay development in vitro; 3) assay development in vivo.


Asunto(s)
Trastornos Relacionados con Opioides , Dolor , Humanos , Dolor/tratamiento farmacológico , Trastornos Relacionados con Opioides/tratamiento farmacológico
2.
ACS Chem Biol ; 17(4): 918-929, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35274923

RESUMEN

Liver fibrosis progression in chronic liver disease leads to cirrhosis, liver failure, or hepatocellular carcinoma and often ends in liver transplantation. Even with an increased understanding of liver fibrogenesis and many attempts to generate therapeutics specifically targeting fibrosis, there is no approved treatment for liver fibrosis. To further understand and characterize the driving mechanisms of liver fibrosis, we developed a high-throughput genome-wide CRISPR/Cas9 screening platform to identify hepatic stellate cell (HSC)-derived mediators of transforming growth factor (TGF)-ß-induced liver fibrosis. The functional genomics phenotypic screening platform described here revealed the novel biology of TGF-ß-induced fibrogenesis and potential drug targets for liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas , Factor de Crecimiento Transformador beta , Fibrosis , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/efectos adversos , Factor de Crecimiento Transformador beta/metabolismo
3.
J Med Chem ; 64(6): 3427-3438, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33715378

RESUMEN

Inhibition of the pituitary adenylate cyclase 1 receptor (PAC1R) is a novel mechanism that could be used for abortive treatment of acute migraine. Our research began with comparative analysis of known PAC1R ligand scaffolds, PACAP38 and Maxadilan, which resulted in the selection of des(24-42) Maxadilan, 6, as a starting point. C-terminal modifications of 6 improved the peptide metabolic stability in vitro and in vivo. SAR investigations identified synergistic combinations of amino acid replacements that significantly increased the in vitro PAC1R inhibitory activity of the analogs to the pM IC90 range. Our modifications further enabled deletion of up to six residues without impacting potency, thus improving peptide ligand binding efficiency. Analogs 17 and 18 exhibited robust in vivo efficacy in the rat Maxadilan-induced increase in blood flow (MIIBF) pharmacodynamic model at 0.3 mg/kg subcutaneous dosing. The first cocrystal structure of a PAC1R antagonist peptide (18) with PAC1R extracellular domain is reported.


Asunto(s)
Circulación Sanguínea/efectos de los fármacos , Péptidos/química , Péptidos/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Animales , Humanos , Proteínas de Insectos/farmacología , Masculino , Ratones , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/fisiopatología , Simulación del Acoplamiento Molecular , Péptidos/farmacocinética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/química , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Vasodilatadores/farmacología
4.
Sci Rep ; 9(1): 19655, 2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31873179

RESUMEN

Multiple genome-wide association studies have identified non-coding single-nucleotide variants (SNVs) near (e.g., rs10166942[C]) or within (rs17862920[T]) the TRPM8 gene that encodes a cold thermosensor is associated with reduced migraine risk. Furthermore, rs10166942[C]) and rs10166942[T]) are more prevalent in populations that reside in hotter and colder climates, respectively. Here we assessed whether these alleles affect TRPM8 expression in humans and human physiologic responses to cold challenge. Here we show that TRPM8 expression is decreased from the chromosome harboring the rs10166942[C] allele in the human dorsal root ganglia. Moreover, carriers of rs10166942[C] required significantly lower temperatures and longer duration of exposure to reach a cold pain threshold (CPTh), which correlated with decreased TRPM8 expression expected in the carriers. This study provides evidence for a genotype-dependent influence on cold pain sensation suggesting that carriers of the reduced migraine risk allele have reduced sensitivity to cold stimuli and that TRPM8 acts as a cold thermosensor and cold pain transducer in humans. Reduced TRPM8 expression and function underpins the migraine protection in carriers of rs10166942[C]; thus, the evaluation of TRPM8 antagonists as migraine therapeutics is warranted. Furthermore, these results provide mechanistic insights for evolutionary positive selection of rs10166942[T] allele in adaptation along latitudinal cline to colder climates.


Asunto(s)
Frío , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Trastornos Migrañosos , Percepción del Dolor , Polimorfismo de Nucleótido Simple , Canales Catiónicos TRPM , Alelos , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Trastornos Migrañosos/genética , Trastornos Migrañosos/metabolismo , Canales Catiónicos TRPM/biosíntesis , Canales Catiónicos TRPM/genética
5.
J Med Chem ; 61(18): 8186-8201, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30148953

RESUMEN

Transient-receptor-potential melastatin 8 (TRPM8), the predominant mammalian cold-temperature thermosensor, is a nonselective cation channel expressed in a subpopulation of sensory neurons in the peripheral nervous system, including nerve circuitry implicated in migraine pathogenesis: the trigeminal and pterygopalatine ganglia. Genomewide association studies have identified an association between TRPM8 and reduced risk of migraine. This disclosure focuses on medicinal-chemistry efforts to improve the druglike properties of initial leads, particularly removal of CYP3A4-induction liability and improvement of pharmacokinetic properties. A novel series of biarylmethanamide TRPM8 antagonists was developed, and a subset of leads were evaluated in preclinical toxicology studies to identify a clinical candidate with an acceptable preclinical safety profile leading to clinical candidate AMG 333, a potent and highly selective antagonist of TRPM8 that was evaluated in human clinical trials.


Asunto(s)
Anticonvulsivantes/farmacología , Descubrimiento de Drogas , Trastornos Migrañosos/prevención & control , Niacina/química , Convulsiones/tratamiento farmacológico , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Anticonvulsivantes/química , Agonistas de los Canales de Calcio/toxicidad , Humanos , Masculino , Microsomas Hepáticos/efectos de los fármacos , Modelos Moleculares , Estructura Molecular , Pirimidinonas/toxicidad , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
6.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27899696

RESUMEN

The transient receptor potential ankyrin 1 (TRPA1) channel has been implicated in pathophysiological processes that include asthma, cough, and inflammatory pain. Agonists of TRPA1 such as mustard oil and its key component allyl isothiocyanate (AITC) cause pain and neurogenic inflammation in humans and rodents, and TRPA1 antagonists have been reported to be effective in rodent models of pain. In our pursuit of TRPA1 antagonists as potential therapeutics, we generated AMG0902, a potent (IC90 of 300 nM against rat TRPA1), selective, brain penetrant (brain to plasma ratio of 0.2), and orally bioavailable small molecule TRPA1 antagonist. AMG0902 reduced mechanically evoked C-fiber action potential firing in a skin-nerve preparation from mice previously injected with complete Freund's adjuvant, supporting the role of TRPA1 in inflammatory mechanosensation. In vivo target coverage of TRPA1 by AMG0902 was demonstrated by the prevention of AITC-induced flinching/licking in rats. However, oral administration of AMG0902 to rats resulted in little to no efficacy in models of inflammatory, mechanically evoked hypersensitivity; and no efficacy was observed in a neuropathic pain model. Unbound plasma concentrations achieved in pain models were about 4-fold higher than the IC90 concentration in the AITC target coverage model, suggesting that either greater target coverage is required for efficacy in the pain models studied or TRPA1 may not contribute significantly to the underlying mechanisms.


Asunto(s)
Hiperalgesia/metabolismo , Inflamación/complicaciones , Ciática/complicaciones , Canales Catiónicos TRPC/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Aminas/uso terapéutico , Analgésicos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/farmacología , Células CHO , Cricetulus , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Conducta Exploratoria/efectos de los fármacos , Adyuvante de Freund/toxicidad , Gabapentina , Hiperalgesia/tratamiento farmacológico , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Naproxeno/farmacología , Fibras Nerviosas Amielínicas/efectos de los fármacos , Fibras Nerviosas Amielínicas/fisiología , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ciática/tratamiento farmacológico , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Ácido gamma-Aminobutírico/uso terapéutico
7.
J Med Chem ; 59(6): 2794-809, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26942860

RESUMEN

There has been significant interest in developing a transient receptor potential A1 (TRPA1) antagonist for the treatment of pain due to a wealth of data implicating its role in pain pathways. Despite this, identification of a potent small molecule tool possessing pharmacokinetic properties allowing for robust in vivo target coverage has been challenging. Here we describe the optimization of a potent, selective series of quinazolinone-based TRPA1 antagonists. High-throughput screening identified 4, which possessed promising potency and selectivity. A strategy focused on optimizing potency while increasing polarity in order to improve intrinsic clearance culminated with the discovery of purinone 27 (AM-0902), which is a potent, selective antagonist of TRPA1 with pharmacokinetic properties allowing for >30-fold coverage of the rat TRPA1 IC50 in vivo. Compound 27 demonstrated dose-dependent inhibition of AITC-induced flinching in rats, validating its utility as a tool for interrogating the role of TRPA1 in in vivo pain models.


Asunto(s)
Proteínas del Tejido Nervioso/antagonistas & inhibidores , Oxadiazoles/síntesis química , Oxadiazoles/farmacología , Purinas/síntesis química , Purinas/farmacología , Quinazolinas/síntesis química , Quinazolinas/farmacología , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Animales , Transporte Biológico Activo , Células CHO , Canales de Calcio , Cricetulus , Perros , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro , Células de Riñón Canino Madin Darby , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Dimensión del Dolor/efectos de los fármacos , Ratas , Relación Estructura-Actividad , Canal Catiónico TRPA1
8.
Cephalalgia ; 36(2): 185-93, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25944818

RESUMEN

BACKGROUND: Migraine headache is a neurological disorder affecting millions worldwide. However, little is known about the mechanisms contributing to migraine. Recent genome-wide association studies have found single nucleotide polymorphisms in the gene encoding transient receptor potential channel M8. Transient receptor potential channel M8 is generally known as a cold receptor but it has been implicated in pain signaling and may play a role in migraine pain. METHODS: In order to investigate whether transient receptor potential channel M8 may contribute to the pain of migraine, the transient receptor potential channel M8 activator icilin was applied to the dura mater using a rat behavioral model of headache. Cutaneous allodynia was measured for 5 hours using Von Frey filaments. RESULTS: Dural application of icilin produced cutaneous facial and hind paw allodynia that was attenuated by systemic pretreatment with the transient receptor potential channel M8-selective antagonist AMG1161 (10 mg/kg p.o.). Further, the anti-migraine agent sumatriptan (0.6 mg/kg s.c.) or the non-selective NOS inhibitor L-NAME (20 mg/kg i.p.) also attenuated allodynia when given as a pretreatment. CONCLUSIONS: These data indicate that transient receptor potential channel M8 activation in the meninges produces behaviors in rats that are consistent with migraine and that are sensitive to pharmacological mechanisms known to have efficacy for migraine in humans. The findings suggest that activation of meningeal transient receptor potential channel M8 may contribute to the pain of migraine.


Asunto(s)
Hiperalgesia/fisiopatología , Trastornos Migrañosos/fisiopatología , Canales Catiónicos TRPM/metabolismo , Animales , Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Masculino , Trastornos Migrañosos/metabolismo , Pirimidinonas/farmacología , Ratas , Ratas Sprague-Dawley
9.
Naunyn Schmiedebergs Arch Pharmacol ; 388(4): 465-76, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25662185

RESUMEN

TRPM8 has been implicated in pain and migraine based on dorsal root- and trigeminal ganglion-enriched expression, upregulation in preclinical models of pain, knockout mouse studies, and human genetics. Here, we evaluated the therapeutic potential in pain of AMG2850 ((R)-8-(4-(trifluoromethyl)phenyl)-N-((S)-1,1,1-trifluoropropan-2-yl)-5,6-dihydro-1,7-naphthyridine-7(8H)-carboxamide), a small molecule antagonist of TRPM8 by in vitro and in vivo characterization. AMG2850 is potent in vitro at rat TRPM8 (IC90 against icilin activation of 204 ± 28 nM), highly selective (>100-fold IC90 over TRPV1 and TRPA1 channels), and orally bioavailable (F po > 40 %). When tested in a skin-nerve preparation, AMG2850 blocked menthol-induced action potentials but not mechanical activation in C fibers. AMG2850 exhibited significant target coverage in vivo in a TRPM8-mediated icilin-induced wet-dog shake (WDS) model in rats (at 10 mg/kg p.o.). However, AMG2850 did not produce a significant therapeutic effect in rat models of inflammatory mechanical hypersensitivity or neuropathic tactile allodynia at doses up to 100 mg/kg. The lack of efficacy suggests that either TRPM8 does not play a role in mediating pain in these models or that a higher level of target coverage is required. The potential of TRPM8 antagonists as migraine therapeutics is yet to be determined.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Naftiridinas/farmacología , Naftiridinas/uso terapéutico , Canales Catiónicos TRPM/antagonistas & inhibidores , Potenciales de Acción/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Encéfalo/metabolismo , Células CHO , Calcio/metabolismo , Frío , Cricetinae , Cricetulus , Adyuvante de Freund , Humanos , Masculino , Mentol/farmacología , Ratones Endogámicos C57BL , Dolor/tratamiento farmacológico , Pirimidinonas , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones
10.
PLoS One ; 9(9): e107151, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25203266

RESUMEN

The mammalian transient receptor potential melastatin channel 8 (TRPM8) is highly expressed in trigeminal and dorsal root ganglia. TRPM8 is activated by cold temperature or compounds that cause a cooling sensation, such as menthol or icilin. TRPM8 may play a role in cold hypersensitivity and hyperalgesia in various pain syndromes. Therefore, TRPM8 antagonists are pursued as therapeutics. In this study we explored the feasibility of blocking TRPM8 activation with antibodies. We report the functional characterization of a rabbit polyclonal antibody, ACC-049, directed against the third extracellular loop near the pore region of the human TRPM8 channel. ACC-049 acted as a full antagonist at recombinantly expressed human and rodent TRPM8 channels in cell based agonist-induced 45Ca2+ uptake assays. Further, several poly-and monoclonal antibodies that recognize the same region also blocked icilin activation of not only recombinantly expressed TRPM8, but also endogenous TRPM8 expressed in rat dorsal root ganglion neurons revealing the feasibility of generating monoclonal antibody antagonists. We conclude that antagonist antibodies are valuable tools to investigate TRPM8 function and may ultimately pave the way for development of therapeutic antibodies.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Ganglios Espinales/efectos de los fármacos , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular , Frío , Cricetulus , Ganglios Espinales/metabolismo , Humanos , Hiperalgesia/metabolismo , Masculino , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Bioorg Med Chem Lett ; 24(15): 3464-8, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24953819

RESUMEN

The transient receptor potential ankyrin 1 (TRPA1) channel is activated by noxious stimuli including chemical irritants and endogenous inflammatory mediators. Antagonists of this channel are currently being investigated for use as therapeutic agents for treating pain, airway disorders, and itch. A novel azabenzofuran series was developed that demonstrated in vitro inhibition of allyl isothiocyanate (AITC)-induced (45)Ca(2+) uptake with nanomolar potencies against both human and rat TRPA1. From this series, compound 10 demonstrated in vivo target coverage in an AITC-induced flinching model in rats while providing unbound plasma concentrations up to 16-fold higher than the TRPA1 rat IC50.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Diseño de Fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Animales , Bloqueadores de los Canales de Calcio/síntesis química , Bloqueadores de los Canales de Calcio/química , Canales de Calcio/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Compuestos Heterocíclicos con 3 Anillos/química , Humanos , Isotiocianatos/antagonistas & inhibidores , Estructura Molecular , Proteínas del Tejido Nervioso/metabolismo , Ratas , Relación Estructura-Actividad , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo
12.
J Pharmacol Exp Ther ; 350(2): 223-31, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24893987

RESUMEN

The transient receptor potential ankyrin 1 (TRPA1) channel has been implicated in different pathophysiologies that include asthma, cough, itch, and inflammatory pain. Agonists of TRPA1 such as mustard oil and its key component allyl isothiocyanate (AITC) cause pain and neurogenic inflammation in humans and pain behaviors in rodents. Hence, TRPA1 antagonists are being pursued as potential therapeutics. With the goal of generating monoclonal antibodies (mAbs) to human TRPA1 that could act as selective antagonists, we immunized mice with a variety of antigens expressing TRPA1 channels. After generation of hybridomas, the hybridoma conditioned media were screened to identify the mAbs that bind TRPA1 channels by a flow cytometry assay utilizing U2OS or Chinese hamster ovary (CHO) cells stably expressing TRPA1. The purified IgGs from the hybridomas that showed selective binding to TRPA1 were evaluated for antagonism in agonist-induced (45)Ca(2+) uptake assays using CHO-TRPA1 cells. Several of the mAbs showed concentration-dependent inhibition of AITC and cold (4°C) activation of TRPA1. The most potent mAb, 2B10, had IC50 values of approximately 260 and 90 nM in the two assays, respectively. These antagonist mAbs also blocked osmotically activated TRPA1 as well as activation by an endogenous agonist (4-oxo-2-nonenal). In summary, we generated mouse mAbs against TRPA1 that act as antagonists of multiple modes of TRPA1 activation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Aldehídos/farmacología , Secuencia de Aminoácidos , Animales , Células CHO , Calcio/metabolismo , Canales de Calcio , Cricetulus , Humanos , Ratones , Datos de Secuencia Molecular , Canal Catiónico TRPA1
13.
J Med Chem ; 57(7): 2989-3004, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24597733

RESUMEN

Transient receptor potential melastatin 8 (TRPM8) is a nonselective cation channel expressed in a subpopulation of sensory neurons in the peripheral nervous system. TRPM8 is the predominant mammalian cold temperature thermosensor and is activated by cold temperatures ranging from 8 to 25 °C and cooling compounds such as menthol or icilin. TRPM8 antagonists are being pursued as potential therapeutics for treatment of pain and bladder disorders. This manuscript outlines new developments in the SAR of a lead series of 1,2,3,4-tetrahydroisoquinoline derivatives with emphasis on strategies to improve pharmacokinetic properties and potency. Selected compounds were profiled in two TRPM8 target-specific in vivo coverage models in rats (the icilin-induced wet dog shake model and the cold pressor test). Compound 45 demonstrated robust efficacy in both pharmacodynamic models with ED90 values <3 mg/kg.


Asunto(s)
Conducta Animal/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Canales Catiónicos TRPM/antagonistas & inhibidores , Tetrahidroisoquinolinas/farmacocinética , Animales , Dicroismo Circular , Frío , Perros , Humanos , Masculino , Microsomas Hepáticos/metabolismo , Pirimidinonas/farmacología , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Estereoisomerismo , Canales Catiónicos TRPM/metabolismo , Tetrahidroisoquinolinas/química , Tetrahidroisoquinolinas/farmacología , Distribución Tisular
14.
J Neurosci ; 34(13): 4445-52, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24671991

RESUMEN

The rodent transient receptor potential ankyrin-1 (TRPA1) channel has been hypothesized to serve as a temperature sensor for thermoregulation in the cold. We tested this hypothesis by using deletion of the Trpa1 gene in mice and pharmacological blockade of the TRPA1 channel in rats. In both Trpa1(-/-) and Trpa1(+/+) mice, severe cold exposure (8°C) resulted in decreases of skin and deep body temperatures to ∼8°C and 13°C, respectively, both temperatures being below the reported 17°C threshold temperature for TRPA1 activation. Under these conditions, Trpa1(-/-) mice had the same dynamics of body temperature as Trpa1(+/+) mice and showed no weakness in the tail skin vasoconstriction response or thermogenic response to cold. In rats, the effects of pharmacological blockade were studied by using two chemically unrelated TRPA1 antagonists: the highly potent and selective compound A967079, which had been characterized earlier, and the relatively new compound 43 ((4R)-1,2,3,4-tetrahydro-4-[3-(3-methoxypropoxy)phenyl]-2-thioxo-5H-indeno[1,2-d]pyrimidin-5-one), which we further characterized in the present study and found to be highly potent (IC50 against cold of ∼8 nm) and selective. Intragastric administration of either antagonist at 30 mg/kg before severe (3°C) cold exposure did not affect the thermoregulatory responses (deep body and tail skin temperatures) of rats, even though plasma concentrations of both antagonists well exceeded their IC50 value at the end of the experiment. In the same experimental setup, blocking the melastatin-8 (TRPM8) channel with AMG2850 (30 mg/kg) attenuated cold-defense mechanisms and led to hypothermia. We conclude that TRPA1 channels do not drive autonomic thermoregulatory responses to cold in rodents.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Regulación de la Temperatura Corporal/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Sensación Térmica/genética , Animales , Regulación de la Temperatura Corporal/efectos de los fármacos , Células CHO , Frío , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas HSP90 de Choque Térmico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Transgénicos , Oximas/sangre , Oximas/farmacología , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Temperatura Cutánea/efectos de los fármacos , Canales Catiónicos TRPM/antagonistas & inhibidores , Sensación Térmica/efectos de los fármacos
15.
Mol Pain ; 8: 36, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22571355

RESUMEN

BACKGROUND: Transient receptor potential cation channel subfamily M member 8 (TRPM8) is activated by cold temperature in vitro and has been demonstrated to act as a 'cold temperature sensor' in vivo. Although it is known that agonists of this 'cold temperature sensor', such as menthol and icilin, cause a transient increase in body temperature (Tb), it is not known if TRPM8 plays a role in Tb regulation. Since TRPM8 has been considered as a potential target for chronic pain therapeutics, we have investigated the role of TRPM8 in Tb regulation. RESULTS: We characterized five chemically distinct compounds (AMG0635, AMG2850, AMG8788, AMG9678, and Compound 496) as potent and selective antagonists of TRPM8 and tested their effects on Tb in rats and mice implanted with radiotelemetry probes. All five antagonists used in the study caused a transient decrease in Tb (maximum decrease of 0.98°C). Since thermoregulation is a homeostatic process that maintains Tb about 37°C, we further evaluated whether repeated administration of an antagonist attenuated the decrease in Tb. Indeed, repeated daily administration of AMG9678 for four consecutive days showed a reduction in the magnitude of the Tb decrease Day 2 onwards. CONCLUSIONS: The data reported here demonstrate that TRPM8 channels play a role in Tb regulation. Further, a reduction of magnitude in Tb decrease after repeated dosing of an antagonist suggests that TRPM8's role in Tb maintenance may not pose an issue for developing TRPM8 antagonists as therapeutics.


Asunto(s)
Regulación de la Temperatura Corporal , Naftiridinas/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Regulación de la Temperatura Corporal/efectos de los fármacos , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , Concentración 50 Inhibidora , Activación del Canal Iónico/efectos de los fármacos , Masculino , Moduladores del Transporte de Membrana/química , Moduladores del Transporte de Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPM/antagonistas & inhibidores
16.
J Neurosci ; 32(6): 2086-99, 2012 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-22323721

RESUMEN

We studied N-(2-aminoethyl)-N-(4-(benzyloxy)-3-methoxybenzyl)thiophene-2-carboxamide hydrochloride (M8-B), a selective and potent antagonist of the transient receptor potential melastatin-8 (TRPM8) channel. In vitro, M8-B blocked cold-induced and TRPM8-agonist-induced activation of rat, human, and murine TRPM8 channels, including those on primary sensory neurons. In vivo, M8-B decreased deep body temperature (T(b)) in Trpm8(+/+) mice and rats, but not in Trpm8(-/-) mice, thus suggesting an on-target action. Intravenous administration of M8-B was more effective in decreasing T(b) in rats than intrathecal or intracerebroventricular administration, indicating a peripheral action. M8-B attenuated cold-induced c-Fos expression in the lateral parabrachial nucleus, thus indicating a site of action within the cutaneous cooling neural pathway to thermoeffectors, presumably on sensory neurons. A low intravenous dose of M8-B did not affect T(b) at either a constantly high or a constantly low ambient temperature (T(a)), but the same dose readily decreased T(b) if rats were kept at a high T(a) during the M8-B infusion and transferred to a low T(a) immediately thereafter. These data suggest that both a successful delivery of M8-B to the skin (high cutaneous perfusion) and the activation of cutaneous TRPM8 channels (by cold) are required for the hypothermic action of M8-B. At tail-skin temperatures <23°C, the magnitude of the M8-B-induced decrease in T(b) was inversely related to skin temperature, thus suggesting that M8-B blocks thermal (cold) activation of TRPM8. M8-B affected all thermoeffectors studied (thermopreferendum, tail-skin vasoconstriction, and brown fat thermogenesis), thus suggesting that TRPM8 is a universal cold receptor in the thermoregulation system.


Asunto(s)
Temperatura Corporal/fisiología , Frío , Ganglios Espinales/fisiología , Tiritona/fisiología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/deficiencia , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/fisiología , Conducta Animal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Ganglios Espinales/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Preparaciones Farmacéuticas/administración & dosificación , Ratas , Ratas Wistar , Tiritona/efectos de los fármacos , Tiofenos/farmacología
17.
J Med Chem ; 55(4): 1593-611, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22329507

RESUMEN

The transient receptor potential melastatin type 8 (TRPM8) is a nonselective cation channel primarily expressed in a subpopulation of sensory neurons that can be activated by a wide range of stimuli, including menthol, icilin, and cold temperatures (<25 °C). Antagonism of TRPM8 is currently under investigation as a new approach for the treatment of pain. As a result of our screening efforts, we identified tetrahydrothienopyridine 4 as an inhibitor of icilin-induced calcium influx in CHO cells expressing recombinant rat TRPM8. Exploration of the structure-activity relationships of 4 led to the identification of a potent and orally bioavailable TRPM8 antagonist, tetrahydroisoquinoline 87. Compound 87 demonstrated target coverage in vivo after oral administration in a rat pharmacodynamic model measuring the prevention of icilin-induced wet-dog shakes (WDS).


Asunto(s)
Analgésicos/síntesis química , Piperidinas/síntesis química , Canales Catiónicos TRPM/antagonistas & inhibidores , Administración Oral , Analgésicos/química , Analgésicos/farmacología , Animales , Células CHO , Calcio/metabolismo , Cricetinae , Cricetulus , Técnicas In Vitro , Masculino , Microsomas Hepáticos/metabolismo , Piperidinas/química , Piperidinas/farmacología , Pirimidinonas/farmacología , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad
18.
Cell Cycle ; 11(2): 343-9, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22214765

RESUMEN

Studies in young rodents have shown that the transient receptor potential vanilloid-1 (TRPV1) channel plays a suppressive role in the systemic inflammatory response syndrome (SIRS) by inhibiting production of tumor necrosis factor (TNF)α and possibly by other mechanisms. We asked whether the anti-inflammatory role of TRPV1 changes with age. First, we studied the effect of AMG517, a selective and potent TRPV1 antagonist, on aseptic, lipopolysaccharide (LPS)-induced SIRS in young (12 wk) mice. In agreement with previous studies, AMG517 increased LPS-induced mortality in the young. We then studied the effects of TRPV1 antagonism (AMG517 or genetic deletion of TRPV1) on SIRS in middle-aged (43-44 wk) mice. Both types of TRPV1 antagonism delayed and decreased LPS-induced mortality, indicating a reversal of the anti-inflammatory role of TRPV1 with aging. In addition, deletion of TRPV1 decreased the serum TNFα response to LPS, suggesting that the suppressive control of TRPV1 on TNFα production is also reversed with aging. In contrast to aseptic SIRS, polymicrobial sepsis (induced by cecal ligation and puncture) caused accelerated mortality in aged TRPV1-deficient mice as compared with wild-type littermates. The recovery of TRPV1-deficient mice from hypothermia associated with the cecal ligation and puncture procedure was delayed. Hence, the reversal of the anti-inflammatory role of TRPV1 found in the aged and their decreased systemic inflammatory response are coupled with suppressed defense against microbial infection. These results caution that TRPV1 antagonists, widely viewed as new-generation painkillers, may decrease the resistance of older patients to infection and sepsis.


Asunto(s)
Envejecimiento , Mediadores de Inflamación/metabolismo , Sepsis/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Canales Catiónicos TRPV/fisiología , Factores de Edad , Animales , Benzotiazoles/farmacología , Benzotiazoles/uso terapéutico , Femenino , Técnicas de Inactivación de Genes , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos de Riesgos Proporcionales , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Sepsis/inmunología , Sepsis/prevención & control , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Factor de Necrosis Tumoral alfa/sangre
19.
J Physiol ; 589(Pt 9): 2415-31, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21486787

RESUMEN

Hypothermia occurs in the most severe cases of systemic inflammation, but the mechanisms involved are poorly understood. This study evaluated whether the hypothermic response to bacterial lipopolysaccharide (LPS) is modulated by the endocannabinoid anandamide(AEA) and its receptors: cannabinoid-1 (CB1), cannabinoid-2 (CB2) and transient receptor potential vanilloid-1 (TRPV1). In rats exposed to an ambient temperature of 22◦C, a moderate dose of LPS (25 - 100 µg kg−1 I.V.) induced a fall in body temperature with a nadir at ∼100 minpostinjection. This response was not affected by desensitization of intra-abdominal TRPV1 receptors with resiniferatoxin (20 µg kg - 1 I.P.), by systemic TRPV1 antagonism with capsazepine(40mg kg−1 I.P.), or by systemic CB2 receptor antagonism with SR144528 (1.4 mg kg−1 I.P.).However, CB1 receptor antagonism by rimonabant (4.6mg kg−1 I.P.) or SLV319 (15mg kg−1 I.P.)blocked LPS hypothermia. The effect of rimonabant was further studied. Rimonabant blocked LPS hypothermia when administered I.C.V. at a dose (4.6 µg) that was too low to produce systemic effects. The blockade of LPS hypothermia by I.C.V. rimonabant was associated with suppression of the circulating level of tumour necrosis factor-α. In contrast to rimonabant,the I.C.V. administration of AEA (50 µg) enhanced LPS hypothermia. Importantly, I.C.V. AEAdid not evoke hypothermia in rats not treated with LPS, thus indicating that AEA modulates LPS-activated pathways in the brain rather than thermo effector pathways. In conclusion, the present study reveals a novel, critical role of brain CB1 receptors in LPS hypothermia. Brain CB1 receptors may constitute a new therapeutic target in systemic inflammation and sepsis.


Asunto(s)
Regulación de la Temperatura Corporal , Encéfalo/metabolismo , Hipotermia/metabolismo , Lipopolisacáridos , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Canales Catiónicos TRPV/metabolismo , Análisis de Varianza , Animales , Ácidos Araquidónicos/metabolismo , Regulación de la Temperatura Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Canfanos/administración & dosificación , Capsaicina/administración & dosificación , Capsaicina/análogos & derivados , Modelos Animales de Enfermedad , Diterpenos/administración & dosificación , Endocannabinoides , Femenino , Hipotermia/inducido químicamente , Hipotermia/fisiopatología , Hipotermia/prevención & control , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Intraventriculares , Masculino , Piperidinas/administración & dosificación , Alcamidas Poliinsaturadas/metabolismo , Pirazoles/administración & dosificación , Ratas , Ratas Long-Evans , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB2/antagonistas & inhibidores , Rimonabant , Transducción de Señal , Sulfonamidas/administración & dosificación , Canales Catiónicos TRPV/antagonistas & inhibidores , Factores de Tiempo
20.
J Neurosci ; 31(5): 1721-33, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21289181

RESUMEN

This study aimed at determining the thermoregulatory phenotype of mice lacking transient receptor potential vanilloid-1 (TRPV1) channels. We used Trpv1 knockout (KO) mice and their genetically unaltered littermates to study diurnal variations in deep body temperature (T(b)) and thermoeffector activities under basal conditions, as well as thermoregulatory responses to severe heat and cold. Only subtle alterations were found in the basal T(b) of Trpv1 KO mice or in their T(b) responses to thermal challenges. The main thermoregulatory abnormality of Trpv1 KO mice was a different pattern of thermoeffectors used to regulate T(b). On the autonomic side, Trpv1 KO mice were hypometabolic (had a lower oxygen consumption) and hypervasoconstricted (had a lower tail skin temperature). In agreement with the enhanced skin vasoconstriction, Trpv1 KO mice had a higher thermoneutral zone. On the behavioral side, Trpv1 KO mice preferred a lower ambient temperature and expressed a higher locomotor activity. Experiments with pharmacological TRPV1 agonists (resiniferatoxin and anandamide) and a TRPV1 antagonist (AMG0347) confirmed that TRPV1 channels located outside the brain tonically inhibit locomotor activity. With age (observed for up to 14 months), the body mass of Trpv1 KO mice exceeded that of controls, sometimes approaching 60 g. In summary, Trpv1 KO mice possess a distinct thermoregulatory phenotype, which is coupled with a predisposition to age-associated overweight and includes hypometabolism, enhanced skin vasoconstriction, decreased thermopreferendum, and hyperkinesis. The latter may be one of the primary deficiencies in Trpv1 KO mice. We propose that TRPV1-mediated signals from the periphery tonically suppress the general locomotor activity.


Asunto(s)
Envejecimiento/metabolismo , Regulación de la Temperatura Corporal/genética , Hipercinesia/metabolismo , Sobrepeso/metabolismo , Canales Catiónicos TRPV/deficiencia , Acrilamidas/farmacología , Animales , Ácidos Araquidónicos/farmacología , Temperatura Corporal/genética , Frío , Diterpenos/farmacología , Endocannabinoides , Femenino , Calor , Hipercinesia/genética , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Pruebas Neuropsicológicas , Consumo de Oxígeno , Fenotipo , Reacción en Cadena de la Polimerasa , Alcamidas Poliinsaturadas/farmacología , Piridinas/farmacología , Piel/irrigación sanguínea , Temperatura Cutánea/genética , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Vasoconstricción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA