Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Inflamm Res ; 72(8): 1649-1664, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37498393

RESUMEN

BACKGROUND, OBJECTIVES AND DESIGN: Arachidonic acid 15-lipoxygenase (ALOX15) has been implicated in the pathogenesis of inflammatory diseases but since pro- and anti-inflammatory roles have been suggested, the precise function of this enzyme is still a matter of discussion. To contribute to this discussion, we created transgenic mice, which express human ALOX15 under the control of the activating protein 2 promoter (aP2-ALOX15 mice) and compared the sensitivity of these gain-of-function animals in two independent mouse inflammation models with Alox15-deficient mice (loss-of-function animals) and wildtype control animals. MATERIALS AND METHODS: Transgenic aP2-ALOX15 mice were tested in comparison with Alox15 knockout mice (Alox15-/-) and corresponding wildtype control animals (C57BL/6J) in the complete Freund's adjuvant induced hind-paw edema model and in the dextran sulfate sodium induced colitis (DSS-colitis) model. In the paw edema model, the degree of paw swelling and the sensitivity of the inflamed hind-paw for mechanic (von Frey test) and thermal (Hargreaves test) stimulation were quantified as clinical readout parameters. In the dextran sodium sulfate induced colitis model the loss of body weight, the colon lengths and the disease activity index were determined. RESULTS: In the hind-paw edema model, systemic inactivation of the endogenous Alox15 gene intensified the inflammatory symptoms, whereas overexpression of human ALOX15 reduced the degree of hind-paw inflammation. These data suggest anti-inflammatory roles for endogenous and transgenic ALOX15 in this particular inflammation model. As mechanistic reason for the protective effect downregulation of the pro-inflammatory ALOX5 pathways was suggested. However, in the dextran sodium sulfate colitis model, in which systemic inactivation of the Alox15 gene protected female mice from DSS-induced colitis, transgenic overexpression of human ALOX15 did hardly impact the intensity of the inflammatory symptoms. CONCLUSION: The biological role of ALOX15 in the pathogenesis of inflammation is variable and depends on the kind of the animal inflammation model.


Asunto(s)
Araquidonato 15-Lipooxigenasa , Colitis , Humanos , Ratones , Femenino , Animales , Ratones Transgénicos , Adyuvante de Freund , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/uso terapéutico , Dextranos/efectos adversos , Dextranos/metabolismo , Ratones Endogámicos C57BL , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/tratamiento farmacológico , Colitis/metabolismo , Colon/metabolismo , Antiinflamatorios/farmacología , Ratones Noqueados , Edema/inducido químicamente , Edema/genética , Edema/metabolismo , Sulfato de Dextran/efectos adversos , Sulfato de Dextran/metabolismo , Modelos Animales de Enfermedad
2.
Int J Mol Sci ; 24(13)2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37446212

RESUMEN

Mammalian arachidonic acid lipoxygenases (ALOXs) have been implicated in the pathogenesis of inflammatory diseases, and its pro- and anti-inflammatory effects have been reported for different ALOX-isoforms. Human ALOX15B oxygenates arachidonic acid to its 15-hydroperoxy derivative, whereas the corresponding 8-hydroperoxide is formed by mouse Alox15b (Alox8). This functional difference impacts the biosynthetic capacity of the two enzymes for creating pro- and anti-inflammatory eicosanoids. To explore the functional consequences of the humanization of the reaction specificity of mouse Alox15b in vivo, we tested Alox15b knock-in mice that express the arachidonic acid 15-lipoxygenating Tyr603Asp and His604Val double mutant of Alox15b, instead of the arachidonic acid 8-lipoxygenating wildtype enzyme, in two different animal inflammation models. In the dextran sodium sulfate-induced colitis model, female Alox15b-KI mice lost significantly more bodyweight during the acute phase of inflammation and recovered less rapidly during the resolution phase. Although we observed significant differences in the colonic levels of selected pro- and anti-inflammatory eicosanoids during the time-course of inflammation, there were no differences between the two genotypes at any time-point of the disease. In Freund's complete adjuvant-induced paw edema model, Alox15b-KI mice were less susceptible than outbred wildtype controls, though we did not observe significant differences in pain perception (Hargreaves-test, von Frey-test) when the two genotypes were compared. our data indicate that humanization of the reaction specificity of mouse Alox15b (Alox8) sensitizes mice for dextran sodium sulfate-induced experimental colitis, but partly protects the animals in the complete Freund's adjuvant-induced paw edema model.


Asunto(s)
Colitis , Dextranos , Humanos , Ratones , Femenino , Animales , Ácido Araquidónico , Inflamación/genética , Mamíferos , Antiinflamatorios , Edema/inducido químicamente , Edema/genética , Modelos Animales de Enfermedad
3.
J Neurosci ; 41(13): 2870-2882, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33593854

RESUMEN

Interleukin-4 (IL-4) is an anti-inflammatory cytokine, which can be protective in inflammatory and neurologic disorders, and can alleviate pain. Classically, IL-4 diminishes pain by blocking the production of proinflammatory cytokines. Here, we uncovered that IL-4 induces acute antinociception by IL-4 receptor α (IL-4Rα)-dependent release of opioid peptides from M1 macrophages at injured nerves. As a model of pathologic pain, we used a chronic constriction injury (CCI) of the sciatic nerve in male mice. A single application of IL-4 at the injured nerves (14 d following CCI) attenuated mechanical hypersensitivity evaluated by von Frey filaments, which was reversed by co-injected antibody to IL-4Rα, antibodies to opioid peptides such as Met-enkephalin (ENK), ß-endorphin and dynorphin A 1-17, and selective antagonists of δ-opioid, µ-opioid, and κ-opioid receptors. Injured nerves were predominately infiltrated by proinflammatory M1 macrophages and IL-4 did not change their numbers or the phenotype, assessed by flow cytometry and qRT-PCR, respectively. Macrophages isolated from damaged nerves by immunomagnetic separation (IMS) and stimulated with IL-4 dose dependently secreted all three opioid peptides measured by immunoassays. The IL-4-induced release of ENK was diminished by IL-4Rα antibody, intracellular Ca2+ chelator, and inhibitors of protein kinase A (PKA), phosphoinositide 3-kinase (PI3K), and ryanodine receptors. Together, we identified a new opioid mechanism underlying the IL-4-induced antinociception that involves PKA-mediated, PI3K-mediated, ryanodine receptor-mediated, and intracellular Ca2+-mediated release from M1 macrophages of opioid peptides, which activate peripheral opioid receptors in injured tissue.SIGNIFICANCE STATEMENT Interleukin-4 (IL-4) is an anti-inflammatory cytokine, which can ameliorate pain. The IL-4-mediated effects are considered to mostly result from the inhibition of the production of proinflammatory mediators (e.g., IL-1ß, tumor necrosis factor, prostaglandin E2). Here, we found that IL-4 injected at the injured nerves attenuates pain by releasing opioid peptides from the infiltrating macrophages in mice. The opioids were secreted by IL-4 in the intracellular Ca2+-dependent manner and activated local peripheral opioid receptors. These actions represent a novel mode of IL-4 action, since its releasing properties have not been so far reported. Importantly, our findings suggest that the IL-4-opioid system should be targeted in the peripheral damaged tissue, since this can be devoid of central and systemic side effects.


Asunto(s)
Interleucina-4/farmacología , Macrófagos/metabolismo , Neuralgia/metabolismo , Neuralgia/patología , Péptidos Opioides/metabolismo , Animales , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Antagonistas de Narcóticos/farmacología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología
4.
J Neuroinflammation ; 13(1): 262, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27717401

RESUMEN

BACKGROUND: During the inflammation which occurs following nerve damage, macrophages are recruited to the site of injury. Phenotypic diversity is a hallmark of the macrophage lineage and includes pro-inflammatory M1 and anti-inflammatory M2 populations. Our aim in this study was to investigate the ability of polarized M0, M1, and M2 macrophages to secrete opioid peptides and to examine their relative contribution to the modulation of neuropathic pain. METHODS: Mouse bone marrow-derived cells were cultured as unstimulated M0 macrophages or were stimulated into an M1 phenotype using lipopolysaccharide and interferon-γ or into an M2 phenotype using interleukin-4. The macrophage phenotypes were verified using flow cytometry for surface marker analysis and cytokine bead array for cytokine profile assessment. Opioid peptide levels were measured by radioimmunoassay and enzyme immunoassay. As a model of neuropathic pain, a chronic constriction injury (CCI) of the sciatic nerve was employed. Polarized M0, M1, and M2 macrophages (5 × 105 cells) were injected perineurally twice, on days 14 and 15 following CCI or sham surgery. Mechanical and heat sensitivity were measured using the von Frey and Hargreaves tests, respectively. To track the injected macrophages, we also transferred fluorescently stained polarized cells and analyzed the surface marker profile of endogenous and injected cells in the nerves ex vivo. RESULTS: Compared to M0 and M1 cells, M2 macrophages contained and released higher amounts of opioid peptides, including Met-enkephalin, dynorphin A (1-17), and ß-endorphin. M2 cells transferred perineurally at the nerve injury site reduced mechanical, but not heat hypersensitivity following the second injection. The analgesic effect was reversed by the perineurally applied opioid receptor antagonist naloxone methiodide. M2 cells did not affect sensitivity following sham surgery. Neither M0 nor M1 cells altered mechanical and heat sensitivity in CCI or sham-operated animals. Tracing the fluorescently labeled M0, M1, and M2 cells ex vivo showed that they remained in the nerve and preserved their phenotype. CONCLUSIONS: Perineural transplantation of M2 macrophages resulted in opioid-mediated amelioration of neuropathy-induced mechanical hypersensitivity, while M1 macrophages did not exacerbate pain. Therefore, rather than focusing on macrophage-induced pain generation, promoting opioid-mediated M2 actions may be more relevant for pain control.


Asunto(s)
Traslado Adoptivo/métodos , Citocinas/metabolismo , Macrófagos/fisiología , Neuralgia/inmunología , Neuralgia/patología , Péptidos Opioides/metabolismo , Aciltransferasas/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Dinorfinas/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Lipopolisacáridos/farmacología , Macrófagos/clasificación , Macrófagos/efectos de los fármacos , Macrófagos/trasplante , Masculino , Ratones , Ratones Endogámicos C57BL , Umbral del Dolor/fisiología , Estimulación Física , betaendorfina/metabolismo
5.
Brain Behav Immun ; 57: 227-242, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27139929

RESUMEN

Opioids are the most powerful analgesics. As pain is driven by sensory transmission and opioid receptors couple to inhibitory G proteins, according to the classical concept, opioids alleviate pain by activating receptors on neurons and blocking the release of excitatory mediators (e.g., substance P). Here we show that analgesia can be mediated by opioid receptors in immune cells. We propose that activation of leukocyte opioid receptors leads to the secretion of opioid peptides Met-enkephalin, ß-endorphin and dynorphin A (1-17), which subsequently act at local neuronal receptors, to relieve pain. In a mouse model of neuropathic pain induced by a chronic constriction injury of the sciatic nerve, exogenous agonists of δ-, µ- and κ-opioid receptors injected at the damaged nerve infiltrated by opioid peptide- and receptor-expressing leukocytes, produced analgesia, as assessed with von Frey filaments. The analgesia was attenuated by pharmacological or genetic inactivation of opioid peptides, and by leukocyte depletion. This decrease in analgesia was restored by the transfer of wild-type, but not opioid receptor-lacking leukocytes. Ex vivo, exogenous opioids triggered secretion of opioid peptides from wild-type immune cells isolated from damaged nerves, which was diminished by blockade of Gαi/o or Gßγ (but not Gαs) proteins, by chelator of intracellular (but not extracellular) Ca(2+), by blockers of phospholipase C (PLC) and inositol 1,4,5-trisphosphate (IP3) receptors, and was partially attenuated by protein kinase C inhibitor. Similarly, the leukocyte depletion-induced decrease in exogenous opioid analgesia was re-established by transfer of immune cells ex vivo pretreated with extracellular Ca(2+) chelator, but was unaltered by leukocytes pretreated with intracellular Ca(2+) chelator or blockers of Gαi/o and Gßγ proteins. Thus, both ex vivo opioid peptide release and in vivo analgesia were mediated by leukocyte opioid receptors coupled to the Gαi/o-Gßγ protein-PLC-IP3 receptors-intracellular Ca(2+) pathway. Our findings suggest that opioid receptors in immune cells are important targets for the control of pathological pain.


Asunto(s)
Analgesia , Calcio/metabolismo , Leucocitos/metabolismo , Neuralgia/metabolismo , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Ratones , Neuralgia/tratamiento farmacológico , Receptores Opioides/agonistas , Método Simple Ciego
6.
J Pharmacol Exp Ther ; 346(3): 535-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23820126

RESUMEN

Activation of opioid receptors on peripheral sensory neurons has the potential for safe pain control, as it lacks centrally mediated side effects. While this approach often only partially suppressed neuropathic pain in animal models, opioids were mostly applied to animal paws although neuropathy was induced at the nerve trunk. Here we aimed to identify the most relevant peripheral site of opioid action for efficient antinociception in neuropathy. On days 2 and 14 following a chronic constriction injury (CCI) of the sciatic nerve in mice, we evaluated dose and time relationships of the effects of µ-, δ-, and κ-opioid receptor agonists injected either at the CCI site or intraplantarly (i.pl.) into the lesioned nerve-innervated paw, on spontaneous paw lifting and heat and mechanical hypersensitivity (using Hargreaves and von Frey tests, respectively). We found that neither agonist diminished spontaneous paw lifting, despite the application site. Heat hypersensitivity was partially attenuated by i.pl. µ-receptor agonist only, while it was improved by all three agonists applied at the CCI site. Mechanical hypersensitivity was slightly diminished by all agonists administered i.pl., whereas it was completely blocked by all opioids injected at the CCI site. These antinociceptive effects were opioid receptor type-selective and site-specific. Thus, opioids might not be effective against spontaneous pain, but they improve heat and mechanical hypersensitivity in neuropathy. Importantly, efficient alleviation of hypersensitivity is governed by peripheral opioid receptors at the injured nerve trunk rather than at its peripheral terminals. Identifying the primary action site of analgesics is important for the development of adequate pain therapies.


Asunto(s)
Analgésicos Opioides/farmacología , Terminaciones Nerviosas/efectos de los fármacos , Neuralgia/tratamiento farmacológico , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Nervios Periféricos/patología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos Opioides/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Constricción Patológica/patología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina D-Penicilamina (2,5)/farmacología , Pie/inervación , Pie/patología , Calor , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/patología , Dimensión del Dolor/efectos de los fármacos , Traumatismos de los Nervios Periféricos/patología , Nervios Periféricos/efectos de los fármacos , Estimulación Física , Receptores Opioides/agonistas
7.
FASEB J ; 26(12): 5161-71, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22923332

RESUMEN

Inflammatory pain can be controlled by endogenous opioid peptides. Here we blocked the degradation of opioids in peripheral injured tissue to locally augment this physiological system. In rats with hindpaw inflammation, inhibitors of aminopeptidase N (APN; bestatin) or neutral endopeptidase (NEP; thiorphan), and a dual inhibitor, NH(2)-CH-Ph-P(O)(OH)CH(2)-CH-CH(2)Ph(p-Ph)-CONH-CH-CH(3)-COOH (P8B), were applied to injured paws. Combined bestatin (1.25-5 mg)/thiorphan (0.2-0.8 mg) or P8B (0.0625-1 mg) alone elevated mechanical nociceptive thresholds to 307 and 227% of vehicle-treated controls, respectively. This analgesia was abolished by antibodies to methionine-enkephalin, leucine-enkephalin, and dynorphin A 1-17, by peripherally restricted and by selective µ-, δ-, and κ-opioid receptor antagonists. Flow cytometry and photospectrometry revealed expression and metabolic activity of APN and NEP on macrophages, granulocytes, and sciatic nerves from inflamed tissue. Radioimmunoassays showed that inhibition of leukocytic APN and NEP by bestatin (5-500 µM)/thiorphan (1-100 µM) combinations or by P8B (1-100 µM) prevented the degradation of enkephalins. Blockade of neuronal peptidases by bestatin (0.5-10 mM)/thiorphan (0.1-5 mM) or by P8B (0.1-10 mM) additionally hindered dynorphin A 1-17 catabolism. Thus, leukocytes and peripheral nerves are important sources of APN and NEP in inflamed tissue, and their blockade promotes peripheral opioid analgesia.


Asunto(s)
Antígenos CD13/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Inflamación/prevención & control , Neprilisina/antagonistas & inhibidores , Dolor/prevención & control , Alanina/análogos & derivados , Alanina/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Antígenos CD13/metabolismo , Relación Dosis-Respuesta a Droga , Dinorfinas/inmunología , Dinorfinas/metabolismo , Dinorfinas/farmacología , Encefalina Leucina/inmunología , Encefalina Leucina/metabolismo , Encefalina Leucina/farmacología , Encefalina Metionina/inmunología , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacología , Citometría de Flujo , Miembro Posterior/efectos de los fármacos , Miembro Posterior/inervación , Miembro Posterior/fisiopatología , Inflamación/complicaciones , Inflamación/enzimología , Leucina/análogos & derivados , Leucina/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/enzimología , Masculino , Antagonistas de Narcóticos , Neprilisina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Péptidos Opioides/inmunología , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Dolor/complicaciones , Dolor/enzimología , Umbral del Dolor/efectos de los fármacos , Ácidos Fosfínicos/farmacología , Ratas , Ratas Wistar , Receptores Opioides/metabolismo , Tiorfan/farmacología
8.
Nature ; 445(7124): 206-9, 2007 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-17167420

RESUMEN

Touch and mechanical pain are first detected at our largest sensory surface, the skin. The cell bodies of sensory neurons that detect such stimuli are located in the dorsal root ganglia, and subtypes of these neurons are specialized to detect specific modalities of mechanical stimuli. Molecules have been identified that are necessary for mechanosensation in invertebrates but so far not in mammals. In Caenorhabditis elegans, mec-2 is one of several genes identified in a screen for touch insensitivity and encodes an integral membrane protein with a stomatin homology domain. Here we show that about 35% of skin mechanoreceptors do not respond to mechanical stimuli in mice with a mutation in stomatin-like protein 3 (SLP3, also called Stoml3), a mammalian mec-2 homologue that is expressed in sensory neurons. In addition, mechanosensitive ion channels found in many sensory neurons do not function without SLP3. Tactile-driven behaviours are also impaired in SLP3 mutant mice, including touch-evoked pain caused by neuropathic injury. SLP3 is therefore indispensable for the function of a subset of cutaneous mechanoreceptors, and our data support the idea that this protein is an essential subunit of a mammalian mechanotransducer.


Asunto(s)
Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Tacto/fisiología , Canales Iónicos Sensibles al Ácido , Vías Aferentes , Animales , Conductividad Eléctrica , Femenino , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Masculino , Mecanorreceptores/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Mutación/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Estructura Terciaria de Proteína , Ratas , Canales de Sodio/metabolismo
9.
Mol Pain ; 8: 81, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23116256

RESUMEN

BACKGROUND: Peripheral nerve injuries often trigger a hypersensitivity to tactile stimulation. Behavioural studies demonstrated efficient and side effect-free analgesia mediated by opioid receptors on peripheral sensory neurons. However, mechanistic approaches addressing such opioid properties in painful neuropathies are lacking. Here we investigated whether opioids can directly inhibit primary afferent neuron transmission of mechanical stimuli in neuropathy. We analysed the mechanical thresholds, the firing rates and response latencies of sensory fibres to mechanical stimulation of their cutaneous receptive fields. RESULTS: Two weeks following a chronic constriction injury of the saphenous nerve, mice developed a profound mechanical hypersensitivity in the paw innervated by the damaged nerve. Using an in vitro skin-nerve preparation we found no changes in the mechanical thresholds and latencies of sensory fibres from injured nerves. The firing rates to mechanical stimulation were unchanged or reduced following injury. Importantly, µ-opioid receptor agonist [D-Ala2,N-Me-Phe4,Gly5]-ol-enkephalin (DAMGO) significantly elevated the mechanical thresholds of nociceptive Aδ and C fibres. Furthermore, DAMGO substantially diminished the mechanically evoked discharges of C nociceptors in injured nerves. These effects were blocked by DAMGO washout and pre-treatment with the selective µ-opioid receptor antagonist Cys2-Tyr3-Orn5-Pen7-amide. DAMGO did not alter the responses of sensory fibres in uninjured nerves. CONCLUSIONS: Our findings suggest that behaviourally manifested neuropathy-induced mechanosensitivity does not require a sensitised state of cutaneous nociceptors in damaged nerves. Yet, nerve injury renders nociceptors sensitive to opioids. Prevention of action potential generation or propagation in nociceptors might represent a cellular mechanism underlying peripheral opioid-mediated alleviation of mechanical hypersensitivity in neuropathy.


Asunto(s)
Neuralgia/metabolismo , Nociceptores/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Receptores Opioides mu/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/genética , Neuronas Aferentes/metabolismo , Traumatismos de los Nervios Periféricos/genética , Receptores Opioides mu/genética
10.
Mol Pain ; 8: 83, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23146666

RESUMEN

BACKGROUND: Proopiomelanocortin (POMC)-derived beta-endorphin1-31 from immune cells can inhibit inflammatory pain. Here we investigated cytokine signaling pathways regulating POMC gene expression and beta-endorphin production in lymphocytes to augment such analgesic effects. RESULTS: Interleukin-4 dose-dependently elevated POMC mRNA expression in naïve lymph node-derived cells in vitro, as determined by real-time PCR. This effect was neutralized by janus kinase (JAK) inhibitors. Transfection of Signal Transducer and Activator of Transcription (STAT) 1/3 but not of STAT6 decoy oligonucleotides abolished interleukin-4 induced POMC gene expression. STAT3 was phosphorylated in in vitro interleukin-4 stimulated lymphocytes and in lymph nodes draining inflamed paws in vivo. Cellular beta-endorphin increased after combined stimulation with interleukin-4 and concanavalin A. Consistently, in vivo reduction of inflammatory pain by passively transferred T cells improved significantly when donor cells were pretreated with interleukin-4 plus concanavalin A. This effect was blocked by naloxone-methiodide. CONCLUSION: Interleukin-4 can amplify endogenous opioid peptide expression mediated by JAK-STAT1/3 activation in mitogen-activated lymphocytes. Transfer of these cells leads to inhibition of inflammatory pain via activation of peripheral opioid receptors.


Asunto(s)
Inflamación/metabolismo , Janus Quinasa 3/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Dolor/metabolismo , Proopiomelanocortina/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Células Cultivadas , Concanavalina A/farmacología , Inflamación/tratamiento farmacológico , Interleucina-4/farmacología , Interleucina-4/uso terapéutico , Janus Quinasa 3/genética , Masculino , Naloxona/análogos & derivados , Naloxona/farmacología , Dolor/tratamiento farmacológico , Compuestos de Amonio Cuaternario/farmacología , ARN Mensajero , Ratas , Ratas Wistar , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética
11.
J Clin Invest ; 119(2): 278-86, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19139563

RESUMEN

The analgesic effects of leukocyte-derived opioids have been exclusively demonstrated for somatic inflammatory pain, for example, the pain associated with surgery and arthritis. Neuropathic pain results from injury to nerves, is often resistant to current treatments, and can seriously impair a patient's quality of life. Although it has been recognized that neuronal damage can involve inflammation, it is generally assumed that immune cells act predominately as generators of neuropathic pain. However, in this study we have demonstrated that leukocytes containing opioids are essential regulators of pain in a mouse model of neuropathy. About 30%-40% of immune cells that accumulated at injured nerves expressed opioid peptides such as beta-endorphin, Met-enkephalin, and dynorphin A. Selective stimulation of these cells by local application of corticotropin-releasing factor led to opioid peptide-mediated activation of opioid receptors in damaged nerves. This ultimately abolished tactile allodynia, a highly debilitating heightened response to normally innocuous mechanical stimuli, which is symptomatic of neuropathy. Our findings suggest that selective targeting of opioid-containing immune cells promotes endogenous pain control and offers novel opportunities for management of painful neuropathies.


Asunto(s)
Leucocitos/metabolismo , Péptidos Opioides/fisiología , Dolor/prevención & control , Enfermedades del Sistema Nervioso Periférico/prevención & control , Animales , Hormona Liberadora de Corticotropina , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Opioides/fisiología
12.
Anesthesiology ; 116(2): 448-57, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22273860

RESUMEN

BACKGROUND: Kinins (e.g., bradykinin) acting through the constitutively expressed B2 and the injury-induced B1 receptors are involved in pain and hyperalgesia, as previously shown by use of receptor-selective antagonists and single-receptor knockout models. Because the overall contribution of kinins to painful processes remains unclear, the aim of this study was to analyze pain-related behaviors of mice unable to respond to kinins because of a lack of both B1 and B2 receptors. METHODS: In knockout mice lacking both B1 and B2 receptors and in wild-type mice (n = 8-21 per group) the authors assessed nociceptive thresholds to mechanical and heat stimuli (von Frey and Hargreaves tests, respectively) in healthy animals and after induction of inflammatory and neuropathic pain, acid-induced visceral nociception, and modulation of nociceptive responses by peripherally administered opioid agonists. RESULTS: In knockout mice lacking both B1 and B2 receptors baseline nociceptive responses to heat were unaltered, nocifensive responses to bradykinin were abolished, acute acetic acid-induced visceral nociception was reduced by approximately 70% (mean difference: 19.5 writhes/30 min) and heat hypersensitivity in carrageenan-induced paw inflammation was decreased 48 h after injection (mean difference 2.88 s), hypersensitivities in chronic complete Freund's adjuvant-induced paw inflammation or after chronic constriction injury of the sciatic nerve were unchanged, and peripheral µ- and δ-opioid-induced analgesia after chronic constriction injury was reduced by 30-35% (mean differences: µ-agonist: 0.495 g, δ-agonist: 0.555 g). CONCLUSIONS: These data suggest that kinins are important for nociception associated with acute short-lasting inflammation but are less essential in chronic stages of pain. The results also highlight a new protective function of kinins via interactions with the opioid system.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Nocicepción/fisiología , Receptor de Bradiquinina B1/deficiencia , Receptor de Bradiquinina B2/deficiencia , Neuropatía Ciática/metabolismo , Analgésicos Opioides/antagonistas & inhibidores , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nocicepción/efectos de los fármacos , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B2/agonistas , Neuropatía Ciática/tratamiento farmacológico , Neuropatía Ciática/genética
13.
PLoS Pathog ; 5(4): e1000362, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19343210

RESUMEN

In inflammation, pain is regulated by a balance of pro- and analgesic mediators. Analgesic mediators include opioid peptides which are secreted by neutrophils at the site of inflammation, leading to activation of opioid receptors on peripheral sensory neurons. In humans, local opioids and opioid peptides significantly downregulate postoperative as well as arthritic pain. In rats, inflammatory pain is induced by intraplantar injection of heat inactivated Mycobacterium butyricum, a component of complete Freund's adjuvant. We hypothesized that mycobacterially derived formyl peptide receptor (FPR) and/or toll like receptor (TLR) agonists could activate neutrophils, leading to opioid peptide release and inhibition of inflammatory pain. In complete Freund's adjuvant-induced inflammation, thermal and mechanical nociceptive thresholds of the paw were quantified (Hargreaves and Randall-Selitto methods, respectively). Withdrawal time to heat was decreased following systemic neutrophil depletion as well as local injection of opioid receptor antagonists or anti-opioid peptide (i.e. Met-enkephalin, beta-endorphin) antibodies indicating an increase in pain. In vitro, opioid peptide release from human and rat neutrophils was measured by radioimmunoassay. Met-enkephalin release was triggered by Mycobacterium butyricum and formyl peptides but not by TLR-2 or TLR-4 agonists. Mycobacterium butyricum induced a rise in intracellular calcium as determined by FURA loading and calcium imaging. Opioid peptide release was blocked by intracellular calcium chelation as well as phosphoinositol-3-kinase inhibition. The FPR antagonists Boc-FLFLF and cyclosporine H reduced opioid peptide release in vitro and increased inflammatory pain in vivo while TLR 2/4 did not appear to be involved. In summary, mycobacteria activate FPR on neutrophils, resulting in tonic secretion of opioid peptides from neutrophils and in a decrease in inflammatory pain. Future therapeutic strategies may aim at selective FPR agonists to boost endogenous analgesia.


Asunto(s)
Mycobacterium/inmunología , Neutrófilos/metabolismo , Nociceptores/metabolismo , Péptidos Opioides/metabolismo , Receptores de Formil Péptido/metabolismo , Análisis de Varianza , Animales , Calcio/metabolismo , Ciclosporina/metabolismo , Encefalina Metionina/metabolismo , Adyuvante de Freund/metabolismo , Humanos , Masculino , Monocitos/metabolismo , Antagonistas de Narcóticos , Neurotransmisores/metabolismo , Dolor/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Wistar , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores Toll-Like/agonistas
14.
Methods Mol Biol ; 2201: 83-95, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32975791

RESUMEN

Real-time quantitative reverse transcription-PCR (qRT-PCR ) is a highly sensitive molecular biology method based on the amplification of the cDNA of mRNA to detect and quantify the levels of mRNA of interest. In this chapter, we describe real-time qRT-PCR to detect and quantify mRNA of opioid receptors in immune cells. Specifically, we analyze mouse immune cells isolated from the blood and sciatic nerves exposed to a chronic constriction injury, which represents a model of neuropathic pain. We describe in detail the requirements and techniques to induce the chronic constriction injury, to isolate immune cells from the blood and injured nerves, to isolate the total RNA from immune cells, to perform a cDNA reverse transcription from the total RNA, and to perform real-time qRT-PCR for µ-, δ-, and κ-opioid receptor mRNAs.


Asunto(s)
Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Receptores Opioides mu/genética , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/genética , Receptores Opioides/inmunología , Receptores Opioides/metabolismo , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/inmunología , Receptores Opioides mu/metabolismo , Transcripción Reversa , Nervio Ciático/lesiones
15.
Methods Mol Biol ; 2201: 127-137, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32975795

RESUMEN

Patch clamp is an electrophysiological technique that allows to analyze the activity of ion channels in neurons. In this chapter, we provide a detailed description of patch clamp protocol to measure the effect of a µ-opioid receptor agonist on the activity of G protein-coupled inwardly rectifying potassium (GIRK or Kir3) channels. This is performed in peripheral sensory neurons isolated from dorsal root ganglia (DRG) of mice without or with a chronic constriction injury (CCI) of the sciatic nerve, which models neuropathic pain. We describe the induction of the CCI , isolation and culture of DRG neurons, performance of the patch clamp recordings, and identification of opioid-responding neurons.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/análisis , Técnicas de Placa-Clamp/métodos , Células Receptoras Sensoriales/fisiología , Animales , Modelos Animales de Enfermedad , Electrofisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Ganglios Espinales/lesiones , Ganglios Espinales/metabolismo , Hiperalgesia , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neuralgia , Traumatismos de los Nervios Periféricos/metabolismo , Nervio Ciático/lesiones
16.
Metabolites ; 11(10)2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34677413

RESUMEN

Arachidonic acid 5-lipoxygenase (ALOX5) is the key enzyme in the biosynthesis of pro-inflammatory leukotrienes. We recently created knock-in mice (Alox5-KI) which express an arachidonic acid 15-lipoxygenating Alox5 mutant instead of the 5-lipoxygenating wildtype enzyme. These mice were leukotriene deficient but exhibited an elevated linoleic acid oxygenase activity. Here we characterized the polyenoic fatty acid metabolism of these mice in more detail and tested the animals in three different experimental inflammation models. In experimental autoimmune encephalomyelitis (EAE), Alox5-KI mice displayed an earlier disease onset and a significantly higher cumulative incidence rate than wildtype controls but the clinical score kinetics were not significantly different. In dextran sodium sulfate-induced colitis (DSS) and in the chronic constriction nerve injury model (CCI), Alox5-KI mice performed like wildtype controls with similar genetic background. These results were somewhat surprising since in previous loss-of-function studies targeting leukotriene biosynthesis (Alox5-/- mice, inhibitor studies), more severe inflammatory symptoms were observed in the EAE model but the degree of inflammation in DSS colitis was attenuated. Taken together, our data indicate that these mutant Alox5-KI mice respond differently in two models of experimental inflammation than Alox5-/- animals tested previously in similar experimental setups.

17.
Brain Behav Immun ; 24(7): 1045-53, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20385224

RESUMEN

Neuropathic pain is a debilitating consequence of nerve injuries and is frequently resistant to classical therapies. T lymphocytes mediate adaptive immune responses and have been suggested to generate neuropathic pain. In contrast, in this study we investigated T cells as a source of opioidergic analgesic ß-endorphin for the control of augmented tactile sensitivity following neuropathy. We employed in vivo nociceptive (von Frey) testing, flow cytometry and immunofluorescence in wild-type and mice with severe combined immunodeficiency (SCID) subjected to a chronic constriction injury of the sciatic nerve. In wild-type mice, T lymphocytes constituted approximately 11% of all immune cells infiltrating the injury site, and they expressed ß-endorphin and receptors for corticotropin-releasing factor (CRF), an agent releasing opioids from leukocytes. CRF applied at the nerve injury site fully reversed neuropathy-induced mechanical hypersensitivity in wild-type animals. In SCID mice, T cells expressing ß-endorphin and CRF receptors were absent at the damaged nerve. Consequently, these animals had substantially reduced CRF-mediated antinociception. Importantly, the decreased antinociception was fully restored by transfer of wild-type mice-derived T lymphocytes in SCID mice. The re-established CRF antinociception could be reversed by co-injection of an antibody against ß-endorphin or an opioid receptor antagonist with limited access to the central nervous system. We propose that, in response to CRF stimulation, T lymphocytes accumulating at the injured nerves utilize ß-endorphin for activation of local neuronal opioid receptors to reduce neuropathy-induced mechanical hypersensitivity. Our findings reveal ß-endorphin-containing T cells as a crucial component of beneficial adaptive immune responses associated with painful peripheral nerve injuries.


Asunto(s)
Neuralgia/inmunología , Neuralgia/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Nervio Ciático/lesiones , Linfocitos T/metabolismo , betaendorfina/metabolismo , Analgésicos/metabolismo , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Antagonistas de Narcóticos , Dimensión del Dolor/métodos , Umbral del Dolor/psicología , Linfocitos T/inmunología , betaendorfina/antagonistas & inhibidores
18.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32102987

RESUMEN

IL-4 is a pleiotropic antiinflammatory cytokine, which can be neuroprotective after nervous system injury. The beneficial actions of IL-4 are thought to result from the blunting of action of inflammatory mediators, such as proinflammatory cytokines. Here, we demonstrate that IL-4 induces M2 macrophages to continuously produce opioid peptides and ameliorate pain. IL-4 application at injured nerves in mice shifted F4/80+ macrophages from the proinflammatory M1 to the antiinflammatory M2 phenotype, which synthesized opioid peptides (Met-enkephalin, ß-endorphin, and dynorphin A 1-17). These effects were accompanied by a long-lasting attenuation of neuropathy-induced mechanical hypersensitivity, beyond the IL-4 treatment. This IL-4-induced analgesia was decreased by opioid peptide antibodies and opioid receptor (δ, µ, κ) antagonists applied at injured nerves, which confirms the involvement of the local opioid system. The participation of M2 macrophages was supported by analgesia in recipient mice injected at injured nerves with F4/80+ macrophages from IL-4-treated donors. Together, IL-4-induced M2 macrophages at injured nerves produced opioid peptides, which activated peripheral opioid receptors to diminish pain. Fostering the opioid-mediated actions of intrinsic M2 macrophages may be a strategy to tackle pathological pain.


Asunto(s)
Analgesia , Interleucina-4/farmacología , Macrófagos/efectos de los fármacos , Péptidos Opioides/biosíntesis , Animales , Calor , Interleucina-4/uso terapéutico , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/tratamiento farmacológico , Péptidos Opioides/fisiología , Tiempo de Reacción/efectos de los fármacos , Receptores de Interleucina-4/antagonistas & inhibidores , Receptores de Interleucina-4/fisiología
19.
Sci Rep ; 10(1): 4366, 2020 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-32127599

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

20.
Mol Pain ; 5: 72, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-20003437

RESUMEN

BACKGROUND: Leukocytes infiltrating inflamed tissue produce and release opioid peptides such as beta-endorphin, which activate opioid receptors on peripheral terminals of sensory nerves resulting in analgesia. Gene therapy is an attractive strategy to enhance continuous production of endogenous opioids. However, classical viral and plasmid vectors for gene delivery are hampered by immunogenicity, recombination, oncogene activation, anti-bacterial antibody production or changes in physiological gene expression. Non-viral, non-plasmid minimalistic, immunologically defined gene expression (MIDGE) vectors may overcome these problems as they carry only elements needed for gene transfer. Here, we investigated the effects of a nuclear localization sequence (NLS)-coupled MIDGE encoding the beta-endorphin precursor proopiomelanocortin (POMC) on complete Freund's adjuvant-induced inflammatory pain in rats. RESULTS: POMC-MIDGE-NLS injected into inflamed paws appeared to be taken up by leukocytes resulting in higher concentrations of beta-endorphin in these cells. POMC-MIDGE-NLS treatment reversed enhanced mechanical sensitivity compared with control MIDGE-NLS. However, both effects were moderate, not always statistically significant or directly correlated with each other. Also, the anti-hyperalgesic actions could not be increased by enhancing beta-endorphin secretion or by modifying POMC-MIDGE-NLS to code for multiple copies of beta-endorphin. CONCLUSION: Although MIDGE vectors circumvent side-effects associated with classical viral and plasmid vectors, the current POMC-MIDGE-NLS did not result in reliable analgesic effectiveness in our pain model. This was possibly associated with insufficient and variable efficacy in transfection and/or beta-endorphin production. Our data point at the importance of the reproducibility of gene therapy strategies for the control of chronic pain.


Asunto(s)
Manejo del Dolor , betaendorfina/metabolismo , Animales , Citometría de Flujo , Vectores Genéticos , Inmunohistoquímica , Inflamación/terapia , Masculino , Ratones , Modelos Biológicos , Proopiomelanocortina/genética , Radioinmunoensayo , Ratas , Ratas Wistar , betaendorfina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA