Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 378(2): 70-76, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33986036

RESUMO

N-Acylethanolamine acid amidase (NAAA) is an N-terminal cysteine hydrolase that stops the physiologic actions of palmitoylethanolamide, an endogenous lipid messenger that activates the transcription factor, peroxisome proliferator-activated receptor-α We have previously reported that the compound ARN19702 [(2-ethylsulfonylphenyl)-[(2S)-4-(6-fluoro-1,3-benzothiazol-2-yl)-2-methylpiperazin-1-yl]methanone] is an orally active, reversible NAAA inhibitor (IC50 on human NAAA = 230 nM) that produces remarkable protective effects against multiple sclerosis in mice. In the present study, we assessed the profile of ARN19702 in mouse and rat models of acute and neuropathic pain. Oral administration in male mice attenuated in a dose-dependent manner the spontaneous nocifensive response elicited by intraplantar formalin injection and the hypersensitivity caused by intraplantar carrageenan injection, paw incision, or sciatic nerve ligation. In male rats, ARN19702 reduced nociception associated with paclitaxel-induced neuropathy without development of subacute antinociceptive tolerance. Finally, ARN19702 (30 mg/kg, oral) did not produce place preference or alter exploratory motor behavior in male mice. The findings support the conclusion that NAAA is a suitable molecular target for the discovery of efficacious analgesic drugs devoid of rewarding potential. SIGNIFICANCE STATEMENT: This study evaluated the pharmacological profile of the orally bioavailable N-acylethanolamine acid amidase (NAAA) inhibitor (2-ethylsulfonylphenyl)-[(2S)-4-(6-fluoro-1,3-benzothiazol-2-yl)-2-methylpiperazin-1-yl]methanone (ARN19702) in mouse and rat models of neurogenic and inflammatory pain. The compound's potential rewarding and sedative effects were also examined. It is concluded that ARN19702 exhibits a broad analgesic profile that can be generalized across rodent species. The findings point to NAAA as a control node in the processing of neuropathic and inflammatory pain and to ARN19702 as a lead to uncover novel pain therapeutics devoid of addictive potential .


Assuntos
Etanolaminas , Amidoidrolases , Animais , Camundongos , Ratos
2.
J Invest Dermatol ; 138(3): 562-569, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29054595

RESUMO

N-acylethanolamine acid amidase (NAAA), a cysteine hydrolase highly expressed in macrophages and B lymphocytes, catalyzes the degradation of palmitoylethanolamide. Palmitoylethanolamide is an agonist of PPAR-α and an important regulator of pain and innate immunity. In this study, we investigated the properties of the NAAA inhibitor, ARN077, in a mouse model of allergic contact dermatitis. Acute topical applications of ARN077 attenuated key signs of DNFB-induced dermatitis in a dose-dependent manner. Moreover, ARN077 increased tissue palmitoylethanolamide content and normalized circulating levels of cytokines and immunoglobulin E. No such effect was seen in PPAR-α-deficient mice. Moreover, mice lacking NAAA failed to develop edema or scratching behavior after challenge with DNFB, confirming that this enzyme plays an important role in dermatitis. Consistent with this conclusion, subchronic applications of ARN077 suppressed DNFB-induced inflammation when administered either before or after the DNFB challenge. The effects of subchronic ARN077 were dose dependent and comparable in size to those produced by the steroids clobetasol and dexamethasone. Unlike the latter, however, ARN077 did not cause skin atrophy. The results identify NAAA as a promising target for the development of effective and safe treatments for atopic dermatitis and other inflammatory disorders of the skin.


Assuntos
Amidoidrolases/antagonistas & inibidores , Carbamatos/uso terapêutico , Dermatite Alérgica de Contato/tratamento farmacológico , Éteres Cíclicos/uso terapêutico , Inflamação/tratamento farmacológico , Prurido/tratamento farmacológico , Amidas , Amidoidrolases/fisiologia , Animais , Dermatite Alérgica de Contato/etiologia , Dinitrofluorbenzeno , Modelos Animais de Doenças , Etanolaminas/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Palmíticos/análise
3.
Proc Natl Acad Sci U S A ; 113(30): E4397-406, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27412859

RESUMO

The intracellular serine amidase, fatty acid amide hydrolase (FAAH), degrades a heterogeneous family of lipid-derived bioactive molecules that include amides of long-chain fatty acids with taurine [N-acyl-taurines (NATs)]. The physiological functions of the NATs are unknown. Here we show that genetic or pharmacological disruption of FAAH activity accelerates skin wound healing in mice and stimulates motogenesis of human keratinocytes and differentiation of human fibroblasts in primary cultures. Using untargeted and targeted lipidomics strategies, we identify two long-chain saturated NATs-N-tetracosanoyl-taurine [NAT(24:0)] and N-eicosanoyl-taurine [NAT(20:0)]-as primary substrates for FAAH in mouse skin, and show that the levels of these substances sharply decrease at the margins of a freshly inflicted wound to increase again as healing begins. Additionally, we demonstrate that local administration of synthetic NATs accelerates wound closure in mice and stimulates repair-associated responses in primary cultures of human keratinocytes and fibroblasts, through a mechanism that involves tyrosine phosphorylation of the epidermal growth factor receptor and an increase in intracellular calcium levels, under the permissive control of transient receptor potential vanilloid-1 receptors. The results point to FAAH-regulated NAT signaling as an unprecedented lipid-based mechanism of wound-healing control in mammalian skin, which might be targeted for chronic wound therapy.


Assuntos
Pele/metabolismo , Taurina/metabolismo , Cicatrização , Amidoidrolases/genética , Amidoidrolases/metabolismo , Animais , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Recém-Nascido , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pele/efeitos dos fármacos , Pele/patologia , Especificidade por Substrato , Taurina/química , Taurina/farmacologia
4.
Eur J Med Chem ; 109: 216-37, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26774927

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) exert their pharmacological effects by inhibiting cyclooxygenase (COX)-1 and COX-2. Though widely prescribed for pain and inflammation, these agents have limited utility in chronic diseases due to serious mechanism-based adverse events such as gastrointestinal damage. Concomitant blockade of fatty acid amide hydrolase (FAAH) enhances the therapeutic effects of the NSAIDs while attenuating their propensity to cause gastrointestinal injury. This favorable interaction is attributed to the accumulation of protective FAAH substrates, such as the endocannabinoid anandamide, and suggests that agents simultaneously targeting COX and FAAH might provide an innovative strategy to combat pain and inflammation with reduced side effects. Here, we describe the rational design and structure-active relationship (SAR) properties of the first class of potent multitarget FAAH-COX inhibitors. A focused SAR exploration around the prototype 10r (ARN2508) led to the identification of achiral (18b) as well as racemic (29a-c and 29e) analogs. Absolute configurational assignment and pharmacological evaluation of single enantiomers of 10r are also presented. (S)-(+)-10r is the first highly potent and selective chiral inhibitor of FAAH-COX with marked in vivo activity, and represents a promising lead to discover novel analgesics and anti-inflammatory drugs.


Assuntos
Amidoidrolases/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Inibidores de Ciclo-Oxigenase/química , Inibidores de Ciclo-Oxigenase/farmacologia , Desenho de Fármacos , Amidoidrolases/metabolismo , Animais , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Humanos , Camundongos , Estereoisomerismo , Relação Estrutura-Atividade
5.
Crit Rev Biochem Mol Biol ; 51(1): 7-14, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26585314

RESUMO

Macrophages are multi-faceted phagocytic effector cells that derive from circulating monocytes and undergo differentiation in target tissues to regulate key aspects of the inflammatory process. Macrophages produce and degrade a variety of lipid mediators that stimulate or suppress pain and inflammation. Among the analgesic and anti-inflammatory lipids released from these cells are the fatty acid ethanolamides (FAEs), which produce their effects by engaging nuclear peroxisome proliferator activated receptor-α (PPAR-α). Two members of this lipid family, palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), have recently emerged as important intrinsic regulators of nociception and inflammation. These substances are released from the membrane precursor, N-acylphosphatidylethanolamine (NAPE), by the action of a NAPE-specific phospholipase D (NAPE-PLD), and in macrophage are primarily deactivated by the lysosomal cysteine amidase, N-acylethanolamine acid amidase (NAAA). NAPE-PLD and NAAA regulate FAE levels, exerting a tight control over the ability of these lipid mediators to recruit PPAR-α and attenuate the inflammatory response. This review summarizes recent findings on the contribution of the FAE-PPAR-α signaling complex in inflammation, and on NAAA inhibition as a novel mechanistic approach to treat chronic inflammatory disorders.


Assuntos
Inflamação/prevenção & controle , Lipídeos/farmacologia , Macrófagos/metabolismo , PPAR alfa/agonistas , Animais , Humanos , Lipídeos/fisiologia
6.
ChemMedChem ; 11(12): 1242-51, 2016 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-26486424

RESUMO

Pain states that arise from non-resolving inflammation, such as inflammatory bowel disease or arthritis, pose an unusually difficult challenge for therapy because of the complexity and heterogeneity of their underlying mechanisms. It has been suggested that key nodes linking interactive pathogenic pathways of non-resolving inflammation might offer novel targets for the treatment of inflammatory pain. Nonsteroidal anti-inflammatory drugs (NSAIDs), which inhibit the cyclooxygenase (COX)-mediated production of pain- and inflammation-inducing prostanoids, are a common first-line treatment for this condition, but their use is limited by mechanism-based side effects. The endogenous levels of anandamide, an endocannabinoid mediator with analgesic and tissue-protective functions, are regulated by fatty acid amide hydrolase (FAAH). This review outlines the pharmacological and chemical rationale for the simultaneous inhibition of COX and FAAH activities with designed multitarget agents. Preclinical studies indicate that such agents may combine superior anti-inflammatory efficacy with reduced toxicity.


Assuntos
Amidoidrolases/antagonistas & inibidores , Ciclo-Oxigenase 1/química , Ciclo-Oxigenase 2/química , Inibidores Enzimáticos/uso terapêutico , Inflamação/prevenção & controle , Dor/tratamento farmacológico , Amidoidrolases/metabolismo , Compostos de Bifenilo/química , Compostos de Bifenilo/uso terapêutico , Carbazóis/química , Carbazóis/uso terapêutico , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Desenho de Fármacos , Inibidores Enzimáticos/química , Humanos , Fenilpropionatos/química , Fenilpropionatos/uso terapêutico
7.
Pharmacol Res ; 97: 7-15, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25882247

RESUMO

We need better medicines to control acute and chronic pain. Fatty acid amide hydrolase (FAAH) and soluble epoxide hydrolase (sEH) catalyze the deactivating hydrolysis of two classes of bioactive lipid mediators--fatty acid ethanolamides (FAEs) and epoxidized fatty acids (EpFAs), respectively--which are biogenetically distinct but share the ability to attenuate pain responses and inflammation. In these experiments, we evaluated the antihyperalgesic activity of small-molecule inhibitors of FAAH and sEH, administered alone or in combination, in two pain models: carrageenan-induced hyperalgesia in mice and streptozocin-induced allodynia in rats. When administered separately, the sEH inhibitor 1-trifluoromethoxyphenyl-3-(1-propionylpiperidine-4-yl)urea (TPPU) and the peripherally restricted FAAH inhibitor URB937 were highly active in the two models. The combination TPPU plus URB937 was markedly synergistic, as assessed using isobolographic analyses. The results of these experiments reveal the existence of a possible functional crosstalk between FAEs and EpFAs in regulating pain responses. Additionally, the results suggest that combinations of sEH and FAAH inhibitors might be exploited therapeutically to achieve greater analgesic efficacy.


Assuntos
Amidoidrolases/antagonistas & inibidores , Analgésicos/farmacologia , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Animais , Canabinoides/uso terapêutico , Carragenina , Neuropatias Diabéticas/complicações , Neuropatias Diabéticas/tratamento farmacológico , Sinergismo Farmacológico , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Medição da Dor/efeitos dos fármacos , Compostos de Fenilureia/uso terapêutico , Piperidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Bibliotecas de Moléculas Pequenas , Estreptozocina
8.
FASEB J ; 29(6): 2616-27, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25757568

RESUMO

The ability of nonsteroidal anti-inflammatory drugs (NSAIDs) to inhibit cyclooxygenase (Cox)-1 and Cox-2 underlies the therapeutic efficacy of these drugs, as well as their propensity to damage the gastrointestinal (GI) epithelium. This toxic action greatly limits the use of NSAIDs in inflammatory bowel disease (IBD) and other chronic pathologies. Fatty acid amide hydrolase (FAAH) degrades the endocannabinoid anandamide, which attenuates inflammation and promotes GI healing. Here, we describe the first class of systemically active agents that simultaneously inhibit FAAH, Cox-1, and Cox-2 with high potency and selectivity. The class prototype 4: (ARN2508) is potent at inhibiting FAAH, Cox-1, and Cox-2 (median inhibitory concentration: FAAH, 0.031 ± 0.002 µM; Cox-1, 0.012 ± 0.002 µM; and Cox-2, 0.43 ± 0.025 µM) but does not significantly interact with a panel of >100 off targets. After oral administration in mice, ARN2508 engages its intended targets and exerts profound therapeutic effects in models of intestinal inflammation. Unlike NSAIDs, ARN2508 causes no gastric damage and indeed protects the GI from NSAID-induced damage through a mechanism that requires FAAH inhibition. Multitarget FAAH/Cox blockade may provide a transformative approach to IBD and other pathologies in which FAAH and Cox are overactive.


Assuntos
Amidoidrolases/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Trato Gastrointestinal/enzimologia , Inflamação/enzimologia , Amidoidrolases/antagonistas & inibidores , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Carragenina , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/enzimologia , Gastroenteropatias/prevenção & controle , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/patologia , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Intestinos/efeitos dos fármacos , Intestinos/enzimologia , Intestinos/patologia , Camundongos , Estrutura Molecular , Fenilcarbamatos/química , Fenilcarbamatos/farmacocinética , Fenilcarbamatos/farmacologia , Fenilpropionatos/química , Fenilpropionatos/farmacocinética , Fenilpropionatos/farmacologia , Fatores de Tempo , Resultado do Tratamento
9.
Nat Neurosci ; 17(2): 164-74, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24473264

RESUMO

Primary sensory afferents and their neighboring host-defense cells are a rich source of lipid-derived mediators that contribute to the sensation of pain caused by tissue damage and inflammation. But an increasing number of lipid molecules have been shown to act in an opposite way, to suppress the inflammatory process, restore homeostasis in damaged tissues and attenuate pain sensitivity by regulating neural pathways that transmit nociceptive signals from the periphery of the body to the CNS. Here we review the molecular and cellular mechanisms that contribute to the modulatory actions of lipid mediators in peripheral nociceptive signaling.


Assuntos
Metabolismo dos Lipídeos , Neurotransmissores/metabolismo , Nociceptores/fisiologia , Dor/patologia , Nervos Periféricos/fisiopatologia , Vias Aferentes/fisiologia , Animais , Humanos , Inflamação/complicações , Neurotransmissores/uso terapêutico , Dor/etiologia , Dor/metabolismo , Nervos Periféricos/patologia
10.
ACS Chem Biol ; 9(2): 398-404, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24191663

RESUMO

Among the growing family of ribosomally synthesized, post-translationally modified peptides, particularly intriguing are class III lanthipeptides containing the triamino acid labionin. In the course of a screening program aimed at finding bacterial cell wall inhibitors, we discovered a new lanthipeptide produced by an Actinoplanes sp. The molecule, designated NAI-112, consists of 22 amino acids and contains an N-terminal labionin and a C-terminal methyl-labionin. Unique among lanthipeptides, it carries a 6-deoxyhexose moiety N-linked to a tryptophan residue. Consistently, the corresponding gene cluster encodes, in addition to the LanKC enzyme characteristic of this lanthipeptide class, a glycosyl transferase. Despite possessing weak antibacterial activity, NAI-112 is effective in experimental models of nociceptive pain, reducing pain symptoms in mice in both the formalin and the chronic constriction injury tests. Thus, NAI-112 represents, after the labyrinthopeptins, the second example of a lanthipeptide effective against nociceptive pain.


Assuntos
Analgésicos/química , Antibacterianos/química , Bacteriocinas/química , Micromonosporaceae/química , Peptídeos/química , Sequência de Aminoácidos , Analgésicos/metabolismo , Analgésicos/uso terapêutico , Animais , Antibacterianos/metabolismo , Antibacterianos/uso terapêutico , Bacteriocinas/genética , Bacteriocinas/uso terapêutico , Genes Bacterianos , Glicosilação , Masculino , Camundongos , Micromonosporaceae/genética , Dados de Sequência Molecular , Família Multigênica , Dor/tratamento farmacológico , Peptídeos/genética , Peptídeos/uso terapêutico
11.
Pain ; 154(3): 350-360, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23218523

RESUMO

Fatty acid ethanolamides (FAEs), which include palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), are endogenous agonists of peroxisome proliferator-activated receptor-α (PPAR-α) and important regulators of the inflammatory response. They are degraded in macrophages by the lysosomal cysteine amidase, N-acylethanolamine acid amidase (NAAA). Previous studies have shown that pharmacological inhibition of NAAA activity suppresses macrophage activation in vitro and causes marked anti-inflammatory effects in vivo, which is suggestive of a role for NAAA in the control of inflammation. It is still unknown, however, whether NAAA-mediated FAE deactivation might regulate pain signaling. The present study examined the effects of ARN077, a potent and selective NAAA inhibitor recently disclosed by our group, in rodent models of hyperalgesia and allodynia caused by inflammation or nerve damage. Topical administration of ARN077 attenuated, in a dose-dependent manner, heat hyperalgesia and mechanical allodynia elicited in mice by carrageenan injection or sciatic nerve ligation. The antinociceptive effects of ARN077 were prevented by the selective PPAR-α antagonist GW6471 and did not occur in PPAR-α-deficient mice. Furthermore, topical ARN077 reversed the allodynia caused by ultraviolet B radiation in rats, and this effect was blocked by pretreatment with GW6471. Sciatic nerve ligation or application of the proinflammatory phorbol ester 12-O-tetradecanoylphorbol 13-acetate decreased FAE levels in sciatic nerve and skin tissue, respectively. ARN077 reversed these biochemical effects. The results identify ARN077 as a potent inhibitor of intracellular NAAA activity, which is active in vivo by topical administration. The findings further suggest that NAAA regulates peripheral pain initiation by interrupting endogenous FAE signaling at PPAR-α.


Assuntos
Amidoidrolases/antagonistas & inibidores , Analgésicos/uso terapêutico , Carbamatos/uso terapêutico , Endocanabinoides/fisiologia , Inibidores Enzimáticos/uso terapêutico , Éteres Cíclicos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Ácidos Oleicos/fisiologia , PPAR alfa/fisiologia , Percepção da Dor/efeitos dos fármacos , Amidas , Amidoidrolases/genética , Amidoidrolases/fisiologia , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Animais , Queimaduras/tratamento farmacológico , Queimaduras/etiologia , Carbamatos/administração & dosagem , Carbamatos/farmacologia , Carragenina/toxicidade , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Etanolaminas , Éteres Cíclicos/administração & dosagem , Éteres Cíclicos/farmacologia , Células HEK293 , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Lisossomos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR alfa/agonistas , PPAR alfa/deficiência , Percepção da Dor/fisiologia , Ácidos Palmíticos , Lesões por Radiação/tratamento farmacológico , Lesões por Radiação/etiologia , Ratos , Proteínas Recombinantes de Fusão/fisiologia , Nervo Isquiático/lesões , Acetato de Tetradecanoilforbol/toxicidade , Raios Ultravioleta/efeitos adversos
12.
Pharmacol Res ; 65(5): 553-63, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22420940

RESUMO

Fatty-acid amide hydrolase (FAAH) catalyzes the intracellular hydrolysis of the endocannabinoid anandamide and other bioactive lipid amides. In the present study, we conducted a comparative characterization of the effects of the newly identified brain-impermeant FAAH inhibitor, URB937 ([3-(3-carbamoylphenyl)-4-hydroxy-phenyl] N-cyclohexylcarbamate), in various rodent models of acute and persistent pain. When administered by the oral route in mice, URB937 was highly active (median effective dose, ED(50), to inhibit liver FAAH activity: 0.3mgkg(-1)) and had a bioavailability of 5.3%. The antinociceptive effects of oral URB937 were investigated in mouse models of acute inflammation (carrageenan), peripheral nerve injury (chronic sciatic nerve ligation) and arthritis (complete Freund's adjuvant). In all models, URB937 was as effective or more effective than standard analgesic and anti-inflammatory drugs (indomethacin, gabapentin, dexamethasone) and reversed pain-related responses (mechanical hyperalgesia, thermal hyperalgesia, and mechanical allodynia) in a dose-dependent manner. ED(50) values ranged from 0.2 to 10mgkg(-1), depending on model and readout. Importantly, URB937 was significantly more effective than two global FAAH inhibitors, URB597 and PF-04457845, in the complete Freund's adjuvant model. The effects of a combination of URB937 with the non-steroidal anti-inflammatory agent, indomethacin, were examined in the carrageenan and chronic sciatic nerve ligation models. Isobolographic analyses showed that the two compounds interacted synergistically to attenuate pain-related behaviors. Furthermore, URB937 reduced the number and severity of gastric lesions produced by indomethacin, while exerting no ulcerogenic effect when administered alone. The results indicate that the peripheral FAAH inhibitor URB937 is more effective than globally active FAAH inhibitors at inhibiting inflammatory pain. Our findings further suggest that FAAH and cyclooxygenase inhibitors interact functionally in peripheral tissues, to either enhance or hinder each other's actions.


Assuntos
Amidoidrolases/antagonistas & inibidores , Úlcera Gástrica/prevenção & controle , Analgésicos/administração & dosagem , Analgésicos/farmacocinética , Analgésicos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/toxicidade , Artrite Experimental/tratamento farmacológico , Artrite Experimental/fisiopatologia , Benzamidas/farmacologia , Canabinoides/administração & dosagem , Canabinoides/farmacocinética , Canabinoides/farmacologia , Carbamatos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Indometacina/administração & dosagem , Indometacina/toxicidade , Masculino , Camundongos , Dor/tratamento farmacológico , Dor/fisiopatologia , Piridazinas/farmacologia , Nervo Isquiático/lesões , Nervo Isquiático/fisiopatologia , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/fisiopatologia , Ureia/análogos & derivados , Ureia/farmacologia
13.
Pain ; 153(1): 33-41, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21890273

RESUMO

We investigated the involvement of de novo neurosteroid synthesis in the mechanisms underlying the analgesic and antihyperalgesic effects of N-palmitoylethanolamine (PEA) in two models of acute and persistent pain, the formalin test and carrageenan-induced paw edema. The pivotal role of peroxisome proliferator-activated receptor (PPAR)-α in the antinocifensive effect of PEA was confirmed by the lack of this effect in PPAR-α-null mice. PEA antinociceptive activity was partially reduced when the animals were treated with aminoglutethimide or finasteride, implying that de novo neurosteroid synthesis is involved in the effect of PEA. Accordingly, in the spinal cord, the allopregnanolone (ALLO) levels were increased by PEA treatment both in formalin- and carrageenan-exposed mice, as revealed by gas chromatography-mass spectrometry. In agreement with those data, in both pain models, PEA administration in challenged mice specifically restored the expression of two proteins involved in neurosteroidogenensis, the steroidogenic acute regulatory protein (StAR) and cytochrome P450 side-chain cleavage (P450scc) in the ipsilateral horns of spinal cord, without affecting their expression in the contralateral side. These results provide new information about the involvement of de novo neurosteroid synthesis in the modulation of pain behavior by PEA.


Assuntos
Analgésicos/uso terapêutico , Dor/tratamento farmacológico , Ácidos Palmíticos/uso terapêutico , Pregnanolona/biossíntese , Medula Espinal/metabolismo , Amidas , Analgésicos/farmacologia , Animais , Endocanabinoides , Etanolaminas , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Camundongos , Dor/metabolismo , Medição da Dor , Ácidos Palmíticos/farmacologia , Medula Espinal/efeitos dos fármacos
14.
Nat Neurosci ; 15(1): 64-9, 2011 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-22101642

RESUMO

The endocannabinoid anandamide is removed from the synaptic space by a selective transport system, expressed in neurons and astrocytes, that remains molecularly uncharacterized. Here we describe a partly cytosolic variant of the intracellular anandamide-degrading enzyme fatty acid amide hydrolase-1 (FAAH-1), termed FAAH-like anandamide transporter (FLAT), that lacked amidase activity but bound anandamide with low micromolar affinity and facilitated its translocation into cells. Known anandamide transport inhibitors, such as AM404 and OMDM-1, blocked these effects. We also identified a competitive antagonist of the interaction of anandamide with FLAT, the phthalazine derivative ARN272, that prevented anandamide internalization in vitro, interrupted anandamide deactivation in vivo and exerted profound analgesic effects in rodent models of nociceptive and inflammatory pain, which were mediated by CB(1) cannabinoid receptors. The results identify FLAT as a critical molecular component of anandamide transport in neural cells and a potential target for therapeutic drugs.


Assuntos
Amidoidrolases/metabolismo , Ácidos Araquidônicos/metabolismo , Neurônios/metabolismo , Alcamidas Poli-Insaturadas/metabolismo , Amidoidrolases/genética , Animais , Ácidos Araquidônicos/farmacologia , Compostos de Benzil/farmacologia , Endocanabinoides , Neurônios/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos
15.
Glia ; 58(14): 1710-26, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20629190

RESUMO

Microglia respond rapidly to injury, increasing their synthesis and release of inflammatory mediators, many of which contribute to the maintenance of persistent pain following CNS or PNS injury. We have recently shown that the lysosomal cysteine protease Cathepsin S (CatS) expressed by spinal microglia is vital for the full expression of neuropathic pain. Here we evaluated the mechanisms by which CatS release occurs from primary microglia in culture. Stimulation of microglia with lipopolysaccharide (LPS) or adenosine tri-phosphate (ATP) alone was insufficient to induce release of enzymatically active CatS in extracellular media. However, following priming with LPS, ATP at 1 mM but not 50 µM resulted in significant release of CatS in the media and maturation of CatS protein in cell extracts. The enzymatic activity measured in media at neutral pH was specific for CatS as it was completely prevented by the CatS inhibitor LHVS. ATP-induced release of CatS required potassium efflux and both extracellular calcium influx and mobilization of intracellular calcium. Pharmacological modulation of ATP-induced release of CatS enzymatic activity revealed that this was dependent on activation of the P2X7 receptor and intracellular phospholipase C and phospholipase A(2). In addition, ATP-induced CatS release involved p38 mitogen activated protein kinase (MAPK) phosphorylation, but not ERK and PI3K signalling pathways. Thus, as high concentration of extracellular ATP promotes release of active CatS from microglia via P2X7 receptor activation, we suggest that the inhibition of CatS release is one of the mechanisms responsible for P2X7 antagonist efficacy in neuropathic pain.


Assuntos
Catepsinas/metabolismo , Microglia/enzimologia , Doenças do Sistema Nervoso Periférico/enzimologia , Doenças do Sistema Nervoso Periférico/etiologia , Receptores Purinérgicos P2X7/fisiologia , Animais , Animais Recém-Nascidos , Catepsinas/antagonistas & inibidores , Células Cultivadas , Técnicas de Cocultura , Microglia/efeitos dos fármacos , Neuralgia/enzimologia , Neuralgia/etiologia , Ratos , Ratos Wistar , Receptores Purinérgicos P2X7/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
16.
J Neurochem ; 114(4): 1143-57, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20524966

RESUMO

The chemokine fractalkine (FKN) is a critical mediator of spinal neuronal-microglial communication in chronic pain. Mature FKN is enzymatically cleaved from neuronal membranes and activation of its receptor, CX3CR1, which is expressed by microglia, induces phosphorylation of p38 MAPK. We used CX3CR1 knockout (KO) mice to examine pain behaviour in the absence of FKN signalling. Naive CX3CR1 KO mice had normal responses to acute noxious stimuli. However, KO mice showed deficits in inflammatory and neuropathic nociceptive responses. After intraplantar zymosan, KO mice did not display thermal hyperalgesia, whereas mechanical allodynia developed fully. In the partial sciatic nerve ligation model of neuropathic pain, both mechanical allodynia and thermal hyperalgesia were less severe in KO mice than in wild-types (WT). Dorsal horn Iba1 immunostaining and phosphorylation of p38 MAPK increased after injury in WT controls but not in KO animals. In WT mice, inflammation and nerve injury increased spinal cord CX3CR1 and FKN expression. FKN protein was also increased in KO mice following inflammation but not after neuropathy, suggesting the FKN/CX3CR1 system is differently affected in the two pain models. Loss of FKN/CX3CR1 neuroimmune communication attenuates hyperalgesia and allodynia in a modality-dependent fashion highlighting the complex nature of microglial response in pathological pain models.


Assuntos
Hiperalgesia/genética , Hiperalgesia/metabolismo , Mediadores da Inflamação/fisiologia , Microglia/metabolismo , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Receptores de Quimiocinas/metabolismo , Medula Espinal/metabolismo , Animais , Receptor 1 de Quimiocina CX3C , Proteínas de Ligação ao Cálcio/metabolismo , Quimiocina CX3CL1/metabolismo , Modelos Animais de Doenças , Feminino , Hiperalgesia/fisiopatologia , Mediadores da Inflamação/antagonistas & inibidores , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos , Microglia/patologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Células do Corno Posterior/metabolismo , Células do Corno Posterior/fisiopatologia , Receptores de Quimiocinas/genética , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Neuropatia Ciática/fisiopatologia , Medula Espinal/fisiopatologia , Regulação para Cima/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Eur Neuropsychopharmacol ; 20(3): 195-206, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19864116

RESUMO

Palmitoylethanolamde (PEA) is an endogenous lipid neuromodulator that mediates a broad spectrum of pharmacological effects by activation of peroxisome proliferator-activated receptor alpha (PPAR-alpha). Detectable or high levels of PEA in the CNS have been found, but the specific function of this lipid remains to be clarified. Here we report evidence that PEA, activating PPAR-alpha receptor and involving neurosteroids de novo synthesis, modulates pentobarbital-evoked hypnotic effect. A single i.c.v. administration of PEA (1-5microg) increases pentobarbital induced loss of righting reflex (LORR) duration in mice. This effect is mimicked by GW7647 (3microg), a synthetic PPAR-alpha agonist, and disappears in PPAR-alpha knockout mice. Antagonism experiments strongly support the engaging of neurosteroidogenic pathway in the increase of LORR duration induced by PEA. This effect disappeared using two inhibitors blocking the key steps of neurosteroids synthesis, aminogluthetimide and finasteride. Moreover, we demonstrated that in brainstem PEA increased the expression of steroidogenic acute regulatory protein (StAR) and cytochrome P450 side-chain cleavage (P450scc), both involved in neurosteroidogenesis. Accordingly, allopregnanolone (ALLO) levels were in turn higher in brainstem of PEA and pentobarbital treated mice vs pentobarbital alone, as revealed by quantitative analysis using gas chromatography-mass spectrometry. A Our results demonstrate that exogenous administration of PEA, through a PPAR-alpha-dependent mechanism, modulates neurosteroids formation increasing ALLO levels and leading to a positive modulation of GABA(A) receptor. These data further strengthen our previous data on the role of PPAR-alpha in PEA's actions and could provide a new framework to understand its role in the CNS.


Assuntos
Hipnóticos e Sedativos/farmacologia , Ácidos Palmíticos/farmacologia , Pentobarbital/farmacologia , Pregnanolona/biossíntese , Amidas , Animais , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Sinergismo Farmacológico , Endocanabinoides , Etanolaminas , Masculino , Camundongos , PPAR alfa/agonistas , PPAR alfa/fisiologia
18.
J Med Chem ; 52(12): 3794-800, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19459639

RESUMO

Although ketorolac is one of the most potent anti-inflammatory and analgesic drugs, its use has been strongly limited owing to the high incidence of adverse effects reported, particularly in the gastrointestinal tract. Using the prodrug approach, which allows the reduction of toxicological features of the parent drug without altering its pharmacological properties, we synthesized an orally administrable prodrug of ketorolac by means of its reversible conjugation to D-galactose (ketogal). In a single dose study, its pharmacokinetic profile was compared with that of ketorolac. Moreover, we found that this prodrug was able to maintain the anti-inflammatory and the analgesic activity of the drug without giving rise to gastric ulcer formation. Thus, these results indicate that ketogal is a highly effective and valid therapeutic alternative to ketorolac itself.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/farmacocinética , Galactose/química , Cetorolaco/farmacologia , Cetorolaco/farmacocinética , Pró-Fármacos/farmacologia , Pró-Fármacos/farmacocinética , Analgésicos/síntese química , Analgésicos/química , Analgésicos/farmacocinética , Analgésicos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/síntese química , Anti-Inflamatórios não Esteroides/química , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Concentração de Íons de Hidrogênio , Cetorolaco/efeitos adversos , Cetorolaco/química , Camundongos , Conformação Molecular , Dor/induzido quimicamente , Pró-Fármacos/síntese química , Pró-Fármacos/química , Úlcera Gástrica/induzido quimicamente , Fatores de Tempo
19.
Eur J Pharmacol ; 613(1-3): 54-9, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19386271

RESUMO

Despite the clear roles played by peroxisome proliferators-activated receptor alpha (PPAR-alpha) in lipid metabolism, inflammation and feeding, the effects of its activation in the central nervous system (CNS) are largely unknown. Palmitoylethanolamide (PEA), a member of the fatty-acid ethanolamide family, acts peripherally as an endogenous PPAR-alpha agonist, exerting analgesic and anti-inflammatory effects. Both PPAR-alpha and PEA are present in the CNS, but the specific functions of this lipid and its receptor remain to be clarified. Using the carrageenan-induced paw model of hyperalgesia in mice, we report here that intracerebroventricular administration of PEA (0.1-1 microg) 30 min before carrageenan injection markedly reduced mechanical hyperalgesia up to 24 h following inflammatory insult. This effect was mimicked by GW7647 (1 microg), a synthetic PPAR-alpha agonist. The obligatory role of PPAR-alpha in mediating PEA's actions was confirmed by the lack of anti-hyperalgesic effects in mutant mice lacking PPAR-alpha. PEA significantly reduced the expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in sciatic nerves and restored carrageenan-induced reductions of PPAR-alpha in the L4-L6 dorsal root ganglia (DRG). To investigate the mechanism by which PEA attenuated hyperalgesia, we evaluated inhibitory kB-alpha (IkB-alpha) degradation and p65 nuclear factor kB (NF-kappaB) activation in DRG. PEA prevented IkB-alpha degradation and p65 NF-kappaB nuclear translocation, confirming the involvement of this transcriptional factor in the control of peripheral hyperalgesia. These results add further support to the broad-spectrum of biological and pharmacological effects induced by PPAR-alpha agonists, suggesting a centrally mediated component for these drugs in controlling inflammatory pain.


Assuntos
Núcleo Celular/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/patologia , NF-kappa B/metabolismo , Ácidos Palmíticos/administração & dosagem , Ácidos Palmíticos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Amidas , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Butiratos/administração & dosagem , Butiratos/farmacologia , Carragenina/toxicidade , Núcleo Celular/metabolismo , Sistema Nervoso Central/metabolismo , Ciclo-Oxigenase 2/biossíntese , Endocanabinoides , Indução Enzimática/efeitos dos fármacos , Etanolaminas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Camundongos , Óxido Nítrico Sintase Tipo II/biossíntese , PPAR alfa/agonistas , PPAR alfa/metabolismo , Ácidos Palmíticos/uso terapêutico , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/farmacologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo
20.
J Pharmacol Exp Ther ; 322(3): 1137-43, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17565008

RESUMO

Peroxisome proliferator-activated receptor (PPAR)-alpha is a nuclear transcription factor. Although the presence of this receptor in different areas of central nervous system (CNS) has been reported, its role remains unclear. Palmitoylethanolamide (PEA), a member of the fatty-acid ethanolamide family, acts peripherally as an endogenous PPAR-alpha ligand, exerting analgesic and anti-inflammatory effects. High levels of PEA in the CNS have been found, but the specific function of this lipid remains to be clarified. Using carrageenan-induced paw edema in mice, we show that i.c.v. administration of PEA may control peripheral inflammation through central PPAR-alpha activation. A single i.c.v. administration of 0.01 to 1 microg of PEA, 30 min before carrageenan injection, reduced edema formation in the mouse carrageenan test. This effect was mimicked by 0.01 to 1 microg of GW7647 [2-[[4-[2-[[(cyclohexylamino)carbonyl](4-cyclohexylbutyl)amino]ethyl]phenyl]thio]-2-methylpropanoic acid], a synthetic PPAR-alpha agonist. Moreover, central PEA administration significantly reduced the expression of the proinflammatory enzymes cyclooxygenase-2 and inducible nitric-oxide synthase, and it significantly restored carrageenan-induced PPAR-alpha reduction in the spinal cord. To investigate the mechanism by which i.c.v. PEA attenuated the development of carrageenan-induced paw edema, we evaluated inhibitor kappaB-alpha (I kappa B-alpha) degradation and nuclear factor-kappaB (NF-kappaB) p65 activation in the cytosolic or nuclear extracts from spinal cord tissue. PEA prevented IkB-alpha degradation and NF-kappaB nuclear translocation, confirming the involvement of this transcriptional factor in the control of peripheral inflammation. The obligatory role of PPAR-alpha in mediating the effects of PEA was confirmed by the lack of the compounds anti-inflammatory effects in mutant mice lacking PPAR-alpha. In conclusion, our data show for the first time that PPAR-alpha activation in the CNS can control peripheral inflammation.


Assuntos
Edema/tratamento farmacológico , PPAR alfa/agonistas , Ácidos Palmíticos/administração & dosagem , Amidas , Animais , Carragenina , Sistema Nervoso Central , Vias de Administração de Medicamentos , Edema/induzido quimicamente , Endocanabinoides , Etanolaminas , Inflamação/tratamento farmacológico , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...