Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Pharmacol Res ; 151: 104580, 2020 01.
Article in English | MEDLINE | ID: mdl-31786317

ABSTRACT

Neutrophils are peripheral immune cells that represent the first recruited innate immune defense against infections and tissue injury. However, these cells can also induce overzealous responses and cause tissue damage. Although the role of neutrophils activating the immune system is well established, only recently their critical implications in neuro-immune interactions are becoming more relevant. Here, we review several aspects of neutrophils in the bidirectional regulation between the nervous and immune systems. First, the role of neutrophils as a diffuse source of acetylcholine and catecholamines is controversial as well as the effects of these neurotransmitters in neutrophil's functions. Second, neutrophils contribute for the activation and sensitization of sensory neurons, and thereby, in events of nociception and pain. In addition, nociceptor activation promotes an axon reflex triggering a local release of neural mediators and provoking neutrophil activation. Third, the recruitment of neutrophils in inflammatory responses in the nervous system suggests these immune cells as innovative targets in the treatment of central infectious, neurological and neurodegenerative disorders. Multidisciplinary studies involving immunologists and neuroscientists are required to define the role of the neurons-neutrophils communication in the pathophysiology of infectious, inflammatory, and neurological disorders.


Subject(s)
Neuroimmunomodulation , Neutrophils/immunology , Animals , Humans , Immunity, Innate , Inflammation/immunology , Neurotransmitter Agents/immunology , Nociception , Pain/immunology , Sensory Receptor Cells/immunology
2.
Brain Behav Immun ; 81: 444-454, 2019 10.
Article in English | MEDLINE | ID: mdl-31271871

ABSTRACT

The gastrointestinal (GI) tract harbors commensal microorganisms as well as invasive bacteria, toxins and other pathogens and, therefore, plays a pivotal barrier and immunological role against pathogenic agents. The vagus nerve is an important regulator of the GI tract-associated immune system, having profound effects on inflammatory responses. Among GI tract organs, the liver is a key site of immune surveillance, as it has a large population of resident macrophages and receives the blood drained from the guts through the hepatic portal circulation. Although it is widely accepted that the hepatic tissue is a major target for vagus nerve fibers, the role of this neural circuit in liver immune functions is still poorly understood. Herein we used in vivo imaging techniques, including confocal microscopy and scintigraphy, to show that vagus nerve stimulation increases the phagocytosis activity by resident macrophages in the liver, even on the absence of an immune challenge. The activation of this neural circuit in a non-lethal model of sepsis optimized the removal of bacteria in the liver and resulted in the production of anti-inflammatory and pro-regenerative cytokines. Our findings provide new insights into the neural regulation of the immune system in the liver.


Subject(s)
Liver/immunology , Phagocytosis/physiology , Vagus Nerve/physiology , Animals , Cytokines , Female , Gastrointestinal Tract , Liver/pathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phagocytes/metabolism , Sepsis/immunology , Vagus Nerve/pathology , Vagus Nerve Stimulation/methods
3.
Brain Behav Immun ; 64: 330-343, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28392428

ABSTRACT

Articular inflammation is a major clinical burden in multiple inflammatory diseases, especially in rheumatoid arthritis. Biological anti-rheumatic drug therapies are expensive and increase the risk of systemic immunosuppression, infections, and malignancies. Here, we report that vagus nerve stimulation controls arthritic joint inflammation by inducing local regulation of innate immune response. Most of the previous studies of neuromodulation focused on vagal regulation of inflammation via the efferent peripheral pathway toward the viscera. Here, we report that vagal stimulation modulates arthritic joint inflammation through a novel "afferent" pathway mediated by the locus coeruleus (LC) of the central nervous system. Afferent vagal stimulation activates two sympatho-excitatory brain areas: the paraventricular hypothalamic nucleus (PVN) and the LC. The integrity of the LC, but not that of the PVN, is critical for vagal control of arthritic joint inflammation. Afferent vagal stimulation suppresses articular inflammation in the ipsilateral, but not in the contralateral knee to the hemispheric LC lesion. Central stimulation is followed by subsequent activation of joint sympathetic nerve terminals inducing articular norepinephrine release. Selective adrenergic beta-blockers prevent the effects of articular norepinephrine and thereby abrogate vagal control of arthritic joint inflammation. These results reveals a novel neuro-immune brain map with afferent vagal signals controlling side-specific articular inflammation through specific inflammatory-processing brain centers and joint sympathetic innervations.


Subject(s)
Arthritis, Experimental/therapy , Locus Coeruleus/physiopathology , Paraventricular Hypothalamic Nucleus/physiopathology , Vagus Nerve Stimulation , Adrenergic beta-Antagonists/administration & dosage , Afferent Pathways/physiopathology , Animals , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/physiopathology , Arthritis, Rheumatoid/therapy , Electric Stimulation , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Rats, Wistar , Sympathetic Nervous System/physiopathology , TRPV Cation Channels/genetics
4.
J Nat Prod ; 79(7): 1828-33, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27367493

ABSTRACT

Chemical compounds belonging to the class of coumarins have promising anti-inflammatory potential. Cinnamoyloxy-mammeisin (CNM) is a 4-phenylcoumarin that can be isolated from Brazilian geopropolis. To our knowledge, its anti-inflammatory activity has never been studied. Therefore, the present study investigated the anti-inflammatory activity of CNM and elucidated its mechanism of action on isolated macrophages. Pretreatment with CNM reduced neutrophil migration into the peritoneal and joint cavity of mice. Likewise, CNM reduced the in vitro and in vivo release of TNF-α and CXCL2/MIP-2. Regarding the possible molecular mechanism of action, CNM reduced the phosphorylation of proteins ERK 1/2, JNK, p38 MAPK, and AP-1 (subunit c-jun) in PG-stimulated macrophages. Pretreatment with CNM also reduced NF-κB activation in RAW 264.7 macrophages stably expressing the NF-κB-luciferase reporter gene. On the other hand, it did not alter IκBα degradation or nuclear translocation of p65. Thus, the results of this study demonstrate promising anti-inflammatory activity of CNM and provide an explanation of its mechanism of action in macrophages via inhibition of MAPK signaling, AP-1, and NF-κB.


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Coumarins/isolation & purification , Coumarins/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Brazil , Coumarins/chemistry , Cyclooxygenase 2/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Molecular Structure , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Signal Transduction/drug effects , Transcription Factor AP-1 , Tumor Necrosis Factor-alpha/pharmacology , eIF-2 Kinase/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Inflamm Res ; 15: 2819-2833, 2022.
Article in English | MEDLINE | ID: mdl-35535053

ABSTRACT

Purpose: Sepsis is the main cause of death in intensive care unit. Maladaptive cytokine storm and T-cell lymphopenia are critical prognosis predictors of sepsis. Electroacupuncture (EA) is expected to be an effective intervention to prevent sepsis. This study aims to determine the potential of EA at ST36 (Zusanli) to prevent experimental septic mice. Methods: Mice were randomly assigned into PBS, LPS, or EA+LPS group. EA (0.1 mA, continuous wave, 10 Hz) was performed stimulating the ST36 for 30 min, once a day for 3 days. After the third day, all mice were challenged with PBS or LPS (4 mg/kg) simultaneously. Mice were evaluated for survival, ear temperature, and other clinical symptoms. Lung and small intestine tissue injuries were analyzed by hematoxylin and eosin staining. Bio-Plex cytokine assay was used to analyze the concentration of cytokines. T lymphocytes were analyzed by flow cytometry and Western blot assays. The role of T cells in preventing sepsis by EA was analyzed by using nude mice lacking T lymphocytes. Results: EA at ST36 improved survival, symptom scores, and ear temperature of endotoxemic mice. EA also improved dramatically pulmonary and intestinal injury by over 50% as compared to untreated mice. EA blunted the inflammatory cytokine storm by inducing a lasting inhibition of the production of major inflammatory factors (TNF-α, IL-1ß, IL-5, IL-6, IL-10, IL-17A, eotaxin, IFN-γ, MIP-1ß and KC). Flow cytometry and Western blot analyses showed EA significantly reduced T-lymphocyte apoptosis and pyroptosis. Furthermore, T lymphocytes were critical for the effects of EA at ST36 stimulation blunted serum TNF-α levels in wild-type but not in nude mice. Conclusion: EA halted systemic inflammation and improved survival in endotoxemic mice. These effects are associated with the protective effect of EA on T lymphocytes, and T cells are required in the anti-inflammatory effects of EA in sepsis.

6.
Pharmacol Rep ; 71(6): 1095-1103, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31629939

ABSTRACT

BACKGROUND: The regulation of the immune system by the sympathetic nervous system is allowing the design of novel treatments for inflammatory disorders such as arthritis. In this study, we have analyzed the effects of α- and ß-adrenoceptor agonists injected subcutaneously, intrathecally, or intra-articularly in zymosan-induced arthritis. METHODS: Murine arthritis was induced by intra-articular (knee joint) injection of zymosan. α1 (phenylephrine), α2 (clonidine), ß1 (dobutamine), or ß2 (salbutamol)-adrenoceptor agonists were injected subcutaneously (sc), intrathecally (it), or intra-articularly (ia) to activate peripheral, spinal, or intra-articular adrenoceptors and to study their effects on articular edema formation and neutrophil migration into the synovial cavity. RESULTS: Treatments with phenylephrine did not affect the edema formation, but it increased neutrophil migration when injected subcutaneously (155.3%) or intra-articularly (187.7%). Treatments with clonidine inhibited neutrophil migration (59.9% sc, 68.7% it, 42.8% ia) regardless of the route of administration, but it inhibited edema formation only when injected intrathecally (66.7%) or intra-articularly (36%) but not subcutaneously. Treatments with dobutamine inhibited both edema (42.0% sc, 69.5% it, 61.6% ia) and neutrophil migration (28.4% sc, 70.3% it, 82.4% ia) in a concentration dependent manner. Likewise, all the treatments with salbutamol also inhibited edema formation (89.9% sc, 62.4% it, 69.8% ia) and neutrophil migration (76.6% sc, 39.1% it, 71.7% ia). CONCLUSION: Whereas the ß-adrenoceptor agonists induced anti-inflammatory effects regardless of their route of administration, α1- and α2-adrenoceptor agonists induced either pro- and anti-inflammatory effects, respectively.


Subject(s)
Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/administration & dosage , Adrenergic beta-Agonists/pharmacology , Arthritis, Experimental/drug therapy , Albuterol/administration & dosage , Albuterol/pharmacology , Animals , Cell Movement/drug effects , Clonidine/administration & dosage , Clonidine/pharmacology , Dobutamine/administration & dosage , Dobutamine/pharmacology , Edema/drug therapy , Injections, Intra-Articular , Injections, Intraperitoneal , Injections, Spinal , Knee Joint , Male , Mice , Neutrophils/drug effects , Phenylephrine/administration & dosage , Phenylephrine/pharmacology , Zymosan
7.
Fundam Clin Pharmacol ; 32(2): 155-162, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29206314

ABSTRACT

The central nervous system controls the innate immunity by modulating efferent neuronal networks. Recently, we have reported that central brain stimulation inhibits inflammatory responses. In the present study, we investigate whether spinal p38 mitogen-activated protein kinase (MAPK) affects joint inflammation in experimental arthritis. Firstly, we observed that intra-articular administration of zymosan in mice induces the phosphorylation of the spinal cord p38 MAPK. In addition, we demonstrated that spinal p38 MAPK inhibition with intrathecal injection of SB203580, a conventional and well-characterized inhibitor, prevents knee joint neutrophil recruitment, edema formation, experimental score and cytokine production. This local anti-inflammatory effect was completely abolished with chemical sympathectomy (guanethidine) and beta-adrenergic receptors blockade (nadolol). In conclusion, our results suggest that pharmacological strategies involving the modulation of spinal p38 MAPK circuit can prevent joint inflammation via sympathetic networks and beta-adrenoceptors activation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/prevention & control , Imidazoles/pharmacology , Joints/drug effects , Neutrophil Infiltration/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Spinal Cord/drug effects , Sympathetic Nervous System/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/administration & dosage , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/physiopathology , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Injections, Spinal , Joints/immunology , Joints/innervation , Male , Mice, Inbred BALB C , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Spinal Cord/enzymology , Spinal Cord/physiopathology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt A): 201-213, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29522782

ABSTRACT

The neuronal control of the immune system is fundamental to the development of new therapeutic strategies for inflammatory disorders. Recent studies reported that afferent vagal stimulation attenuates peripheral inflammation by activating specific sympathetic central and peripheral networks, but only few subcortical brain areas were investigated. In the present study, we report that afferent vagal stimulation also activates specific cortical areas, as the parietal and cingulate cortex. Since these cortical structures innervate sympathetic-related areas, we investigate whether electrical stimulation of parietal cortex can attenuate knee joint inflammation in non-anesthetized rats. Our results show that cortical stimulation in rats increased sympathetic activity and improved joint inflammatory parameters, such as local neutrophil infiltration and pro-inflammatory cytokine levels, without causing behavioral disturbance, brain epileptiform activity or neural damage. In addition, we superposed the areas activated by afferent vagal or cortical stimulation to map common central structures to depict a brain immunological homunculus that can allow novel therapeutic approaches against inflammatory joint diseases, such as rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/physiopathology , Arthritis, Experimental/therapy , Cerebral Cortex/physiopathology , Deep Brain Stimulation , Animals , Arthritis, Experimental/pathology , Cerebral Cortex/pathology , Implantable Neurostimulators , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats, Wistar , Vagus Nerve/physiopathology , Vagus Nerve Stimulation , Zymosan
9.
Behav Brain Res ; 182(2): 301-7, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17208313

ABSTRACT

Housing conditions change the emotional state of the animals. Ultrasound vocalizations (USVs) termed as 22 kHz are the usual components of the defensive responses of rats exposed to threatening conditions such as isolation. The amount of emission of 22 kHz USVs depends on the intensity of the aversive stimuli. While short periods of isolation caused an anxiolytic-sensitive enhancement of the defensive responses, long-term isolation tended to reduce the defensive performance of the animals to aversive stimuli. The dorsal periaqueductal gray (dPAG) is an important vocal center and a crucial structure for the expression of defensive response. While it has been shown that Substance P (SP) at this midbrain level is involved in the modulation of the defensive response, its role in the emission of ultrasound vocalizations has not been evaluated. In this study we examined whether the resocialization and local injections of SP into the dPAG have an influence on the isolation-induced 22 kHz USVs recorded within the frequency range of 18-26 kHz. Rats isolated for 1 day showed a significant increase in the number and duration of USVs, which were reversed by resocialization. On the other hand, 2-week isolation reduced the number and duration of 22 kHz USVs, which could not be reversed by resocialization. SP injections into the dPAG (35 pmol/0.2 microL) caused a reduction in the 22 kHz USVs. Pretreatment with the NK-1 receptor antagonist spantide (100 pmol/0.2 microL) blocked these effects but exhibited no effect when given alone. These findings suggest that 1-day and 2-week isolation recruit distinct brain defensive systems. Also, in agreement with the notion that intense fear is associated with the neural substrates of fear of the dPAG, activation of NK-1 receptors of this midbrain structure reduces the 22 kHz USVs.


Subject(s)
Periaqueductal Gray/drug effects , Social Isolation , Substance P/pharmacology , Ultrasonics , Vocalization, Animal/drug effects , Analgesics/pharmacology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Drug Interactions , Male , Microinjections/methods , Rats , Rats, Wistar , Substance P/analogs & derivatives , Time Factors , Ultrasonics/classification , Vocalization, Animal/classification , Vocalization, Animal/physiology
10.
Pharmacol Biochem Behav ; 79(2): 367-76, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15501314

ABSTRACT

The blockade of GABA-mediated Cl(-) influx with pentylenetetrazol (PTZ) was used in the present work to induce seizures in animals. The neurotransmission in the postictal period has been the focus of many studies, and there is evidence suggesting antinociceptive mechanisms following tonic-clonic seizures in both animals and men. The aim of this work was to study the involvement of acetylcholine in the antinociception induced by convulsions elicited by peripheral administration of PTZ (64 mg/kg). Analgesia was measured by the tail-flick test in eight albino Wistar rats per group. Convulsions were followed by significant increases in tail-flick latencies (TFLs) at least for 120 min of the postictal period. Peripheral administration of atropine (0.25, 1 and 4 mg/kg) caused a significant dose-dependent decrease in the TFL in seizing animals, as compared to controls. These data were corroborated by peripheral administration of mecamylamine, a nicotinic cholinergic receptor blocker, at the same doses (0.25, 1 and 4 mg/kg) used for the muscarinic cholinergic receptor antagonist. The recruitment of the muscarinic receptor was made 10 min postconvulsions and in subsequent periods of postictal analgesia, whereas the involvement of the nicotinic cholinergic receptor was implicated only after 30 min postseizures. The cholinergic antagonists caused a minimal reduction in body temperature, but did not impair baseline TFL, spontaneous exploration or motor coordination in the rotarod test at the maximal dose of 4 mg/kg. These results indicate that acetylcholine may be involved as a neurotransmitter in postictal analgesia.


Subject(s)
Epilepsy/physiopathology , Nociceptors/physiopathology , Receptors, Muscarinic/physiology , Receptors, Nicotinic/physiology , Animals , Atropine/pharmacology , Chlorides/metabolism , Convulsants/toxicity , Epilepsy/chemically induced , GABA Antagonists/toxicity , Male , Mecamylamine/pharmacology , Muscarinic Antagonists/pharmacology , Nicotinic Antagonists/pharmacology , Pain Measurement , Pentylenetetrazole/toxicity , Rats , Rats, Wistar , Receptors, GABA-A/physiology
11.
Neurosci Lett ; 569: 121-5, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24708929

ABSTRACT

The amygdala, together with the dorsal periaqueductal gray (dPAG), medial hypothalamus, and deep layers of the superior and inferior colliculi, constitutes the encephalic aversion system, which has been considered the main neural substrate for the organization of fear and anxiety. The basolateral nucleus of the amygdala (BLA) acts as a filter for aversive stimuli to higher structures while the central (CeA) and the medial (MeA) nuclei constitute the output for the autonomic and somatic components of the emotional reaction through major projections to the limbic and brainstem regions. Although some findings point to the distinct participation of the substance P (SP) and the NK1 receptors system in the different nuclei of the amygdala on the expression of emotional behaviors, it is not clear if this system modulates anxiety-like responses in the distinct nuclei of the amygdala as well as the dPAG. Thus, it was investigated if the injection of SP into the BLA, CeA, or MeA affects the expression of anxiety-like responses of rats submitted to the elevated plus-maze (EPM) test and, if the effects are mediated by NK1 receptors. The results showed that SP and Sar-Met-SP (NK1 receptor selective agonist) injected into the CeA and MeA, but not into the BLA, caused anxiogenic-like effects in the EPM. Altogether, the data indicates that the SP may mimic the effects of anxiogenic stimuli via NK1 receptor activation only in the CeA and MeA (amygdala's nuclei output) and may activate the neural mechanisms involved in the defensive reaction genesis. The SP/NK1 receptors system activation may be phasically involved in very specific aspects of anxiety behaviors.


Subject(s)
Anxiety/psychology , Central Amygdaloid Nucleus/drug effects , Corticomedial Nuclear Complex/drug effects , Receptors, Neurokinin-1/agonists , Substance P/analogs & derivatives , Substance P/pharmacology , Animals , Male , Maze Learning/drug effects , Microinjections , Rats, Wistar
12.
Exp Neurol ; 213(2): 410-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18671968

ABSTRACT

The post-ictal immobility syndrome is followed by a significant increase in the nociceptive thresholds in animals and humans. The aim of this study was to assess the involvement of the dorsal raphe nucleus (DRN) in the post-ictal antinociception. The second aim was to study the role of serotonergic intrinsic mechanisms of the DRN in this hypo-algesic phenomenon. Pentylenetetrazole (PTZ), an ionophore GABA-mediated Cl(-) influx antagonist, was peripherally used to induce tonic-clonic seizures in Wistar rats. The nociceptive threshold was measured by the tail-flick test. Neurochemical lesions of the DRN, performed with microinjection of ibotenic acid (1.0 microg/0.2 microL), caused a significant decrease of tonic-clonic seizure-induced antinociception, suggesting the involvement of this nucleus in this antinociceptive process. Microinjections of methysergide (1.0 and 5.0 microg/0.2 microL), a non-selective serotonergic receptor antagonist, into DRN caused a significant decrease in the post-ictal antinociception in seizing animals, compared to controls, in all post-ictal periods presently studied. These findings were corroborated by microinjections of ketanserin (at 1.0 and 5.0 microg/0.2 microL) into DRN. Ketanserin is an antagonist with large affinity for 5-HT(2A/2C) serotonergic receptors, which, in this case, caused a significant decrease in the tail-flick latencies in seizing animals, compared to controls after the first 20 min following tonic-clonic convulsive reactions. These results indicate that serotonergic neurotransmission of the DRN neuronal clusters is involved in the organization of the post-ictal hypo-algesia. The 5-HT(2A/2C) receptors of DRN neurons seem to be critically involved in the increase of nociceptive thresholds following tonic-clonic seizures.


Subject(s)
Pain Measurement/methods , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT2A/physiology , Receptor, Serotonin, 5-HT2C/physiology , Serotonin/physiology , Synaptic Transmission/physiology , Animals , Male , Pain Measurement/drug effects , Raphe Nuclei/drug effects , Rats , Rats, Wistar , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL