Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Mol Sci ; 20(13)2019 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-31269729

RESUMEN

The stimulation of the AMP-activated kinase (AMPK) by 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) has been associated with antihyperalgesia and the inhibition of nociceptive signaling in the spinal cord in models of paw inflammation. The attenuated nociception comes along with a strongly reduced paw edema, indicating that peripheral antiinflammatory mechanisms contribute to antinociception. In this study, we investigated the impact of AICAR on the immune cell composition in inflamed paws, as well as the regulation of inflammatory and resolving markers in macrophages. By using fluorescence-activated cell sorting (FACS) analysis and immunofluorescence, we found a significantly increased fraction of proresolving M2 macrophages and anti-inflammatory interleukin (IL)-10 in inflamed tissue, while M1 macrophages and proinflammatory cytokines such as IL-1 were decreased by AICAR in wild type mice. In AMPKα2 knock-out mice, the M2 polarization of macrophages in the paw was missing. The results were supported by experiments in primary macrophage cultures which also showed a shift to a proresolving phenotype with decreased levels of proinflammatory mediators and increased levels of antiinflammatory mediators. However, in the cell cultures, we did not observe differences between the AMPKα2+/+ and -/- cells, thus indicating that the AICAR-induced effects are at least partially AMPK-independent. In summary, our results indicate that AICAR has potent antiinflammatory and proresolving properties in inflammation which are contributing to a reduction of inflammatory edema and antinociception.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Antiinflamatorios/uso terapéutico , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Ribonucleótidos/uso terapéutico , Aminoimidazol Carboxamida/uso terapéutico , Animales , Células Cultivadas , Edema/complicaciones , Edema/tratamiento farmacológico , Edema/inmunología , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/inmunología , Inflamación/complicaciones , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL
2.
Biochem Biophys Res Commun ; 447(3): 520-5, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24732361

RESUMEN

AMP-activated kinase is a cellular energy sensor which is activated in stages of increased ATP consumption. Its activation has been associated with a number of beneficial effects such as decreasing inflammatory processes and the disease progress of diabetes and obesity, respectively. Furthermore, AMPK activation has been linked with induction of cell cycle arrest and apoptosis in cancer and vascular cells, indicating that it might have a therapeutic impact for the treatment of cancer and atherosclerosis. However, the impact of AMPK on the proliferation of macrophages, which also play a key role in the formation of atherosclerotic plaques and in inflammatory processes, has not been focused so far. We have assessed the influence of AICAR- and metformin-induced AMPK activation on cell viability of macrophages with and without inflammatory stimulation, respectively. In cells without inflammatory stimulation, we found a strong induction of caspase 3-dependent apoptosis associated with decreased mTOR levels and increased expression of p21. Interestingly, these effects could be inhibited by co-stimulation with bacterial lipopolysaccharide (LPS) but not by other proinflammatory cytokines suggesting that AICAR induces apoptosis via AMPK in a TLR4-pathway dependent manner. In conclusion, our results revealed that AMPK activation is not only associated with positive effects but might also contribute to risk factors by disturbing important features of macrophages. The fact that LPS is able to restore AMPK-associated apoptosis might indicate an important role of TLR4 agonists in preventing unfavorable cell death of immune cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Apoptosis/inmunología , Caspasa 3/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Ribonucleótidos/metabolismo , Aminoimidazol Carboxamida/metabolismo , Aminoimidazol Carboxamida/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Metformina/farmacología , Ratones , Ribonucleótidos/farmacología , Serina-Treonina Quinasas TOR/metabolismo
3.
J Immunol ; 187(5): 2617-25, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21813779

RESUMEN

Inhibitor-κB kinase ε (IKKε) was only recently identified as an enzyme with high homology to the classical I-κB kinase subunits, IKKα and IKKß. Despite this similarity, it is mainly discussed as a repressor of viral infections by modulating type I IFNs. However, in vitro studies also showed that IKKε plays a role in the regulation of NF-κB activity, but the distinct mechanisms of IKKε-mediated NF-κB activation are not clear. Given the paramount role of NF-κB in inflammation, we investigated the regulation and function of IKKε in models of inflammatory hyperalgesia in mice. We found that IKKε was abundantly expressed in nociceptive neurons in the spinal cord and in dorsal root ganglia. IKKε mRNA and protein levels rapidly increased in spinal cord and dorsal root ganglia during hind paw inflammation evoked by injection of zymosan or formalin. IKKε knockout mice showed normal nociceptive responses to acute heat or mechanical stimulation. However, in inflammatory pain models, IKKε-deficient mice exhibited a significantly reduced nociceptive behavior in comparison with wild type mice, indicating that IKKε contributed to the development of inflammatory hyperalgesia. Antinociceptive effects were associated with reduced activation of NF-κB and attenuated NF-κB-dependent induction of cyclooxygenase-2, inducible NO synthase, and metalloproteinase-9. In contrast, IRF-3, which is an important IKKε target in viral infections, was not regulated after inflammatory nociceptive stimulation. Therefore, we concluded that IKKε modulates inflammatory nociceptive sensitivity by activation of NF-κB-dependent gene transcription and may be useful as a therapeutic target in the treatment of inflammatory pain.


Asunto(s)
Hiperalgesia/enzimología , Quinasa I-kappa B/metabolismo , Neuronas/metabolismo , Animales , Western Blotting , Citocinas/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Ganglios Espinales/metabolismo , Hiperalgesia/etiología , Hibridación in Situ , Inflamación/complicaciones , Inflamación/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/inmunología , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Médula Espinal/metabolismo
4.
Sports Med ; 45(11): 1497-509, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26186961

RESUMEN

Activation of the adenosine monophosphate (AMP)-activated kinase (AMPK) contributes to beneficial effects such as improvement of the hyperglycemic state in diabetes as well as reduction of obesity and inflammatory processes. Furthermore, stimulation of AMPK activity has been associated with increased exercise capacity. A study published in 2008, directly before the Olympic Games in Beijing, showed that the AMPK activator AICAR (5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide) increased the running capacity of mice without any training and thus, prompted the World Anti-Doping Agency (WADA) to include certain AMPK activators in the list of forbidden drugs. This raises the question as to whether all AMPK activators should be considered for registration or whether the increase in exercise performance is only associated with specific AMPK-activating substances. In this review, we intend to shed light on currently published AMPK-activating drugs, their working mechanisms, and their impact on body fitness.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Tolerancia al Ejercicio/fisiología , Proteínas Quinasas Activadas por AMP/química , Animales , Activación Enzimática/efectos de los fármacos , Humanos , Músculo Esquelético/enzimología , Condicionamiento Físico Animal
5.
Eur J Pharmacol ; 762: 299-305, 2015 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-26049010

RESUMEN

AMP-activated kinase (AMPK) is a cellular energy sensor, which is activated in stages of increased adenosine triphosphate (ATP) consumption. Its activation has been associated with a number of beneficial effects such as decrease of inflammatory processes and inhibition of disease progression of diabetes and obesity. A recent study suggested that salicylate, the active metabolite of the non-steroidal anti-inflammatory drug (NSAID) acetyl-salicylic acid (aspirin), is able to activate AMPK pharmacologically. This observation raised the question whether or not other NSAIDs might also act as AMPK activators and whether this action might contribute to their cyclooxygenase (COX)-independent anti-inflammatory properties. In this study, we investigated mouse and human neuronal cells and liver tissue of mice after treatment with various NSAIDs. Our results showed that the non-selective acidic NSAIDs ibuprofen and diclofenac induced AMPK activation similar to aspirin while the COX-2 selective drug etoricoxib and the non-opioid analgesic paracetamol, both drugs have no acidic structure, failed to activate AMPK. In conclusion, our results revealed that AMPK can be activated by specific non-steroidal anti-inflammatory drugs such as salicylic acid, ibuprofen or diclofenac possibly depending on the acidic structure of the drugs. AMPK might therefore contribute to their antinociceptive and anti-inflammatory properties.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Proteínas Quinasas Activadas por AMP/genética , Animales , Línea Celular Tumoral , Diclofenaco/farmacología , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Ibuprofeno/farmacología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
6.
Pain ; 154(3): 368-376, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23318130

RESUMEN

microRNAs (miRNAs) are small noncoding RNAs that have been linked to a number of disease-related signal transduction pathways. Several studies indicate that they are also involved in nociception. It is not clear, however, which miRNAs are important and which genes are modulated by miRNA-associated mechanisms. This study focuses on the regulation and function of the central nervous system (CNS)-specific miRNA-124a in the spinal cord of mice in a formalin model of inflammatory nociception. miRNA-124a is constitutively expressed in the spinal cord of mice, particularly in neurons of the dorsal horn. Peripheral noxious stimulation with formalin led to significant down-regulation of its expression. Knock-down of miRNA-124a by intravenous administration of a specific miRNA-124a inhibitor further increased the nociceptive behavior associated with an upregulation of the pain-relevant miRNA-124a target MeCP2 and proinflammatory marker genes. In contrast, administration of a miRNA-124a mimic counteracted these effects and decreased nociception by down-regulation of the target gene. In conclusion, our results indicate that miRNA-124a is involved in inflammatory nociception by regulation of relevant target proteins and might therefore constitute a novel target for anti-inflammatory therapy.


Asunto(s)
Inflamación/tratamiento farmacológico , MicroARNs/fisiología , Percepción del Dolor/fisiología , Médula Espinal/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Formaldehído/toxicidad , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Masculino , Proteína 2 de Unión a Metil-CpG/biosíntesis , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , MicroARNs/genética , Terapia Molecular Dirigida , Oligorribonucleótidos/farmacología , Dolor/etiología , Dolor/prevención & control , Percepción del Dolor/efectos de los fármacos , Células del Asta Posterior/metabolismo , ARN Bicatenario/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Método Simple Ciego , Médula Espinal/fisiopatología , Transfección
7.
J Pain ; 14(11): 1330-40, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23916727

RESUMEN

UNLABELLED: The activation of the adenosine monophosphate (AMP)-activated kinase (AMPK) has been associated with beneficial effects such as improvement of hyperglycemic states in diabetes as well as reduction of obesity and inflammatory processes. Recent studies provide evidence for a further role of AMPK in models of acute and neuropathic pain. In this study, we investigated the impact of AMPK on inflammatory nociception. Using 5-amino-1-ß-d-ribofuranosyl-imidazole-4-carboxamide (AICAR) and metformin as AMPK activators, we observed anti-inflammatory and antinociceptive effects in 2 models of inflammatory nociception. The effects were similar to those observed with the standard analgesic ibuprofen. The mechanism appears to be based on regulation of the AMPKα2 subunit of the kinase because AMPKα2 knockout mice showed increased nociceptive responses that could not be reversed by the AMPK activators. On the molecular level, antinociceptive effects are at least partially mediated by reduced activation of different MAP-kinases in the spinal cord and a subsequent decrease in pain-relevant induction of c-fos, which constitutes a reliable marker of elevated activity in spinal cord neurons following peripheral noxious stimulation. In summary, our results indicate that activation of AMPKα2 might represent a novel therapeutic option for the treatment of inflammation-associated pain, providing analgesia with fewer unwanted side effects. PERSPECTIVE: AMPK activation is associated with beneficial effects on diabetes and obesity. In addition, we have shown analgesic properties of pharmacologic AMPK activation in inflammatory nociception, indicating that AMPK might serve as a novel therapeutic target in pain with fewer unwanted side effects.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Hiperalgesia/tratamiento farmacológico , Inflamación/metabolismo , Metformina/farmacología , Nocicepción/efectos de los fármacos , Ribonucleótidos/farmacología , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/farmacología , Animales , Conducta Animal/efectos de los fármacos , Activación Enzimática , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Destreza Motora/efectos de los fármacos , Nocicepción/fisiología , Dimensión del Dolor , Prueba de Desempeño de Rotación con Aceleración Constante
8.
Artículo en Inglés | MEDLINE | ID: mdl-22960082

RESUMEN

Atomoxetine (ATX), a selective norepinephrine reuptake inhibitor, is a non-stimulant approved for the treatment of attention deficit/hyperactivity disorder (ADHD). Little is known about the molecular basis for its therapeutic effect. The objective of this animal study was to determine alterations in gene expression patterns in the prefrontal cortex after long-term administration of atomoxetine. Rats were treated for 21 days during childhood and early adolescent stages of development with a once-daily oral application of 0.05 g/kg atomoxetine, which resulted in plasma levels similar to those described in children. A whole genome RNA-microarray of rat prefrontal cortical gene expression after administration of atomoxetine versus sterile water revealed an mRNA increase in 114 genes (≥2-fold) while 11 genes were down-regulated (≤0.5-fold). By applying quantitative real-time PCR (qRT-PCR) and Western Blot we confirmed a significant increase in the expression of GABA A receptor subunits as well as ubiquinol-cytochrome c reductase complex core protein 2 (Uqcrc2). SNAP-25 (synaptosomal-associated protein of 25 kDa), which is an ADHD candidate gene and an important vesicle protein involved in axonal growth, synaptic plasticity and regulation of neurotransmitter release was also significantly upregulated on RNA- and protein level after atomoxetine treatment. In summary, we could show that long-term treatment with the ADHD drug atomoxetine induces the regulation of several genes in the prefrontal cortex of young rats. Especially the increased expression of SNAP-25 and GABA-A receptor subunits may indicate additional active therapeutic mechanisms for atomoxetine.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Expresión Génica/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Propilaminas/farmacología , Animales , Clorhidrato de Atomoxetina , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Masculino , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA