Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Biol Chem ; 297(3): 101085, 2021 09.
Article in English | MEDLINE | ID: mdl-34411562

ABSTRACT

The complement cascade is a key component of the innate immune system that is rapidly recruited through a cascade of enzymatic reactions to enable the recognition and clearance of pathogens and promote tissue repair. Despite its well-understood role in immunology, recent studies have highlighted new and unexpected roles of the complement cascade in neuroimmune interaction and in the regulation of neuronal processes during development, aging, and in disease states. Complement signaling is particularly important in directing neuronal responses to tissue injury, neurotrauma, and nerve lesions. Under physiological conditions, complement-dependent changes in neuronal excitability, synaptic strength, and neurite remodeling promote nerve regeneration, tissue repair, and healing. However, in a variety of pathologies, dysregulation of the complement cascade leads to chronic inflammation, persistent pain, and neural dysfunction. This review describes recent advances in our understanding of the multifaceted cross-communication that takes place between the complement system and neurons. In particular, we focus on the molecular and cellular mechanisms through which complement signaling regulates neuronal excitability and synaptic plasticity in the nociceptive pathways involved in pain processing in both health and disease. Finally, we discuss the future of this rapidly growing field and what we believe to be the significant knowledge gaps that need to be addressed.


Subject(s)
Complement Pathway, Classical/immunology , Neuroimmunomodulation/physiology , Nociceptive Pain/physiopathology , Animals , Complement Activation/immunology , Complement System Proteins/immunology , Humans , Immunity, Innate/physiology , Neuroimmunomodulation/immunology , Neuronal Plasticity/physiology , Neurons , Nociception , Nociceptive Pain/immunology , Pain/immunology , Pain/physiopathology , Signal Transduction
2.
Am J Physiol Renal Physiol ; 317(1): F90-F98, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31091120

ABSTRACT

Altered Toll-like receptor (TLR)4 activation has been identified in several chronic pain conditions but has not been well studied in interstitial cystitis/bladder pain syndrome (IC/BPS). Our previously published human studies indicated that patients with IC/BPS present altered systemic TLR4-mediated inflammatory responses, which were significantly correlated with reported pain severity. In the present study, we sought to determine whether altered TLR4 activation plays a role in pelvic/bladder pain seen in patients with IC/BPS using our validated IC/BPS-like transgenic autoimmune cystitis model (URO-OVA). URO-OVA mice developed responses consistent with pelvic and bladder pain after cystitis induction, which was associated with increased splenocyte production of TLR4-mediated proinflammatory cytokines IL-1ß, IL-6, and TNF-α. Increased spinal expression of mRNAs for proinflammatory cytokines IL-6 and TNF-α, glial activation markers CD11b and glial fibrillary acidic protein, and endogenous TLR4 ligand high mobility group box 1 was also observed after cystitis induction. Compared with URO-OVA mice, TLR4-deficient URO-OVA mice developed significantly reduced nociceptive responses, although similar bladder inflammation and voiding dysfunction, after cystitis induction. Intravenous administration of TAK-242 (a TLR4-selective antagonist) significantly attenuated nociceptive responses in cystitis-induced URO-OVA mice, which was associated with reduced splenocyte production of TLR4-mediated IL-1ß, IL-6, and TNF-α as well as reduced spinal expression of mRNAs for IL-6, TNF-α, CD11b, glial fibrillary acidic protein, and high mobility group box 1. Our results indicate that altered TLR4 activation plays a critical role in bladder nociception independent of inflammation and voiding dysfunction in the URO-OVA model, providing a potential mechanistic insight and therapeutic target for IC/BPS pain.


Subject(s)
Autoimmune Diseases/metabolism , Cystitis, Interstitial/metabolism , Nociceptive Pain/metabolism , Pain Threshold , Toll-Like Receptor 4/metabolism , Urinary Bladder/metabolism , Analgesics/pharmacology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Cells, Cultured , Cystitis, Interstitial/genetics , Cystitis, Interstitial/immunology , Cystitis, Interstitial/physiopathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nociceptive Pain/genetics , Nociceptive Pain/immunology , Nociceptive Pain/physiopathology , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Pain Threshold/drug effects , Signal Transduction , Spine/immunology , Spine/metabolism , Spleen/immunology , Spleen/metabolism , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Urinary Bladder/drug effects , Urinary Bladder/immunology , Urinary Bladder/physiopathology , Urodynamics
3.
Biochim Biophys Acta ; 1822(1): 74-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21496484

ABSTRACT

Cross-talk between the immune- and nervous-system is considered an important biological process in health and disease. Because mast cells are often strategically placed between nerves and surrounding (immune)-cells they may function as important intermediate cells. This review summarizes the current knowledge on bidirectional interaction between mast cells and nerves and its possible relevance in (inflammation-induced) increased nociception. Our main focus is on mast cell mediators involved in sensitization of TRP channels, thereby contributing to nociception, as well as neuron-released neuropeptides and their effects on mast cell activation. Furthermore we discuss mechanisms involved in physical mast cell-nerve interactions. This article is part of a Special Issue entitled: Mast cells in inflammation.


Subject(s)
Cell Communication , Intestines/physiopathology , Mast Cells/metabolism , Neuroimmunomodulation , Nociception , Animals , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/physiopathology , Intestines/innervation , Intestines/pathology , Neurons, Afferent/metabolism , Nociceptive Pain/immunology , Nociceptive Pain/pathology , Nociceptive Pain/physiopathology , Signal Transduction , Stress, Physiological
4.
Exp Parasitol ; 133(2): 193-200, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23206953

ABSTRACT

Several reports have shown that cutaneous leishmaniasis lesions are painless, suggesting that Leishmania infection interferes with pain perception. Comparisons of inflammation-induced hyperalgesia between BALB/c and C57BL/6 mice have been little explored in the literature, and comparative data regarding nociception in leishmaniasis are non-existent. In susceptible BALB/c mice and resistant C57BL/6 mice that were intradermally inoculated with a low dose of Leishmania major in the ear, we investigated the variation in nociception over a 12-wk period post-infection and this variation's association with the structure of nerve fibres and the presence of endogenous cytokines that are classically considered hyper- or hypo-nociceptive. Infected BALB/c mice presented susceptibility and severe lesions. Infected C57BL/6 mice exhibited resistance and healing lesions. The immune response involved pro- and anti-inflammatory cytokine secretion, respectively. The infection-induced hypoalgesia in BALB/c mice after wks 9 was accompanied by decreased levels of IL-6 and IL-10 in ear tissue with intact nerves. C57BL/6 mice showed short-lived hyperalgesia in wks 2, which was related to increased local levels of IL-6, KC/CXCL-1, TNF-α and IL-10 and a decrease in nerve density. The increase in pro-inflammatory cytokine IL-6, KC/CXCL-1 and TNF-α levels during hyperalgesia suggested a role for these mediators in afferent nerve sensitisation, which was secondary to the inflammatory damage of nerve fibres stained by PGP 9.5. In contrast, the mechanisms of hypoalgesia may include the downregulation of cytokines, the preservation of the structure of nerve endings, and as yet uninvestigated unidentified differences in neurotransmitter release or a direct role of the parasites in the context of the progressive and permissive inflammatory response of BALB/c mice.


Subject(s)
Cytokines/analysis , Ear, External/parasitology , Leishmania major/immunology , Leishmaniasis, Cutaneous/physiopathology , Nociception/physiology , Animals , Cytokines/metabolism , Down-Regulation , Ear, External/immunology , Ear, External/innervation , Female , Immunohistochemistry , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/complications , Leishmaniasis, Cutaneous/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nociceptive Pain/etiology , Nociceptive Pain/immunology , Nociceptive Pain/physiopathology , Pain Threshold/physiology , Ubiquitin Thiolesterase/analysis
5.
Molecules ; 18(9): 10953-72, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-24013409

ABSTRACT

The P2X7 receptor (P2X7R) is a nonselective cation channel that is activated by extracellular ATP and triggers the secretion of several proinflammatory substances, such as IL-1ß, IL-18, TNF-α, and nitric oxide. Recently, several preclinical studies have demonstrated that this receptor participates in inflammation and pain mechanisms. Taken together, these results indicate that P2X7R is a promising pharmacological target, and compounds that modulate the function of this receptor show potential as new anti-inflammatory medicines. In this review, we discuss aspects of P2X7R pharmacology and the participation of this protein in inflammation and pain and provide an overview of some promising compounds that have been tested as antagonists of P2X7R, with clinical applicability.


Subject(s)
Nociception/physiology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Neuralgia/drug therapy , Neuralgia/immunology , Neuralgia/metabolism , Nociceptive Pain/drug therapy , Nociceptive Pain/immunology , Nociceptive Pain/metabolism , Purinergic P2X Receptor Antagonists/therapeutic use
6.
Pharm Biol ; 51(9): 1144-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23795810

ABSTRACT

CONTEXT: Citronellal is a monoterpene present in the oil of many species, including Cymbopogon winterianus Jowitt (Poaceae). OBJECTIVE: The present study investigated the effect of citronellal on inflammatory nociception induced by different stimuli and examined the involvement of the NO-cGMP-ATP-sensitive K⁺ channel pathway. MATERIALS AND METHODS: We used male Swiss mice (n = 6 per group) that were treated intraperitoneally with citronellal (25, 50 or 100 mg/kg) 0.5 h after the subplantar injection of 20 µl of carrageenan (CG; 300 µg/paw), tumor necrosis factor-α (TNF-α; 100 pg/paw), prostaglandin E2 (PGE2; 100 ng/paw) or dopamine (DA; 30 µg/paw). The mechanical nociception was evaluated at 0.5, 1, 2 and 3 h after the injection of the agents, using a digital analgesimeter (von Frey). The effects of citronellal were also evaluated in the presence of L-NAME (30 mg/kg) or glibenclamide (5 mg/kg). RESULTS: At all times, citronellal in all doses inhibited the development of mechanical nociception induced by CG (p < 0.001 and p < 0.01) and TNF-α (p < 0.001, p < 0.01, and p < 0.05). The citronellal was able to increase the pain threshold in the DA test (p < 0.001, p < 0.01, and p < 0.05) and in the PGE2 test at all times (p < 0.001 and p < 0.05). L-NAME and glibenclamide reversed the antinociceptive effects of the citronellal at higher doses in the PGE2 test. DISCUSSION AND CONCLUSION: These data suggest that citronellal attenuated mechanical nociception, mediated in part by the NO-cGMP-ATP-sensitive K⁺ channel pathway.


Subject(s)
Aldehydes/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclic GMP/metabolism , Disease Models, Animal , KATP Channels/metabolism , Monoterpenes/therapeutic use , Nitric Oxide/metabolism , Nociceptive Pain/prevention & control , Acyclic Monoterpenes , Aldehydes/administration & dosage , Aldehydes/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cyclic GMP/antagonists & inhibitors , Cymbopogon/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glyburide/pharmacology , Indonesia , KATP Channels/antagonists & inhibitors , Male , Mice , Monoterpenes/administration & dosage , Monoterpenes/antagonists & inhibitors , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nociceptive Pain/immunology , Nociceptive Pain/metabolism , Oils, Volatile/chemistry , Pain Threshold/drug effects , Plant Oils/chemistry , Potassium Channel Blockers/pharmacology , Signal Transduction/drug effects
7.
Bull Exp Biol Med ; 154(6): 711-3, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23658904

ABSTRACT

Microinjections of LPS into the specific nuclei of rat thalamus (ventrobasal thalamic nuclei VPL and VPM) slightly increased perceptual component and significantly decreased emotional component of systemic nociceptive response.


Subject(s)
Lipopolysaccharides/pharmacology , Nociception , Nociceptive Pain/immunology , Thalamus/immunology , Animals , Emotions , Male , Microinjections , Pain/immunology , Pain/psychology , Rats , Rats, Wistar , Thalamus/physiology , Vocalization, Animal
8.
Int Immunopharmacol ; 122: 110674, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37481846

ABSTRACT

We previously demonstrated that experimental traumatic occlusion (ETO) induces a long-lasting nociceptive response. These findings were associated with altered neuronal patterns and suggestive satellite glial cell activation. This study aimed to elucidate the activation of satellite glial cells following ETO in the trigeminal ganglion. Moreover, we explored the involvement of resident and infiltrating cells in trigeminal ganglion in ETO. Finally, we investigated the overexpression of purinergic signaling and the CX3CL1/CX3CR1 axis. RT-qPCR and electrophoresis showed overexpression of GFAP in the trigeminal ganglion (TG), and immunohistochemistry corroborated these findings, demonstrating SGCs activation. ELISA reveals enhanced levels of TNF-α and IL-1ß in TG after 28 d of ETO. In trigeminal ganglia, ETO groups improved the release of CX3CL1, and immunohistochemistry showed higher CX3CR1+ -immunoreactive cells in ETO groups. Immunohistochemistry and electrophoresis of the P2X7 receptor were found in ETO groups. The mRNA levels of IBA1 are upregulated in the 0.7-mm ETO group, while immunohistochemistry showed higher IBA1+ -immunoreactive cells in both ETO groups. The expression of CD68 by electrophoresis and immunohistochemistry was observed in the ETO groups. For last, ELISA revealed increased levels of IL-6, IL-12, and CCL2 in the TG of ETO groups. Furthermore, the mRNA expression revealed augmented transcription factors and cytokines associated with lymphocyte activation, such as RORγt, IL-17, Tbet, IFNγ, FOXP3, and IL-10. The findings of this study suggested that ETO activates SGCs in TG, and purinergic signaling and CX3CL1/CX3CR1 axis were upregulated. We uncovered the involvement of a distinct subtype of macrophages, named sensory neuron-associated macrophage activation (sNMAs), and detected an expanded number of infiltrated macrophages onto TG. These findings indicate that ETO induces chronic/persistent immune response.


Subject(s)
Lymphocyte Activation , Macrophage Activation , Nociceptive Pain , Oligodendroglia , Trigeminal Ganglion , Trigeminal Ganglion/injuries , Nociceptive Pain/immunology , CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Animals , Rats , Glial Fibrillary Acidic Protein/metabolism , Male , Rats, Wistar , Oligodendroglia/immunology , Receptors, Purinergic P2X/metabolism
9.
J Pain ; 22(3): 322-343, 2021 03.
Article in English | MEDLINE | ID: mdl-33227508

ABSTRACT

Pain is a common but potentially debilitating symptom, often requiring complex management strategies. To understand the molecular dynamics of peripheral inflammation and nociceptive pain, we investigated longitudinal changes in behavior, tissue structure, and transcriptomic profiles in the rat carrageenan-induced peripheral inflammation model. Sequential changes in the number of differentially expressed genes are consistent with temporal recruitment of key leukocyte populations, mainly neutrophils and macrophages with each wave being preceded by upregulation of the cell-specific chemoattractants, Cxcl1 and Cxcl2, and Ccl2 and Ccl7, respectively. We defined 12 temporal gene clusters based on expression pattern. Within the patterns we extracted genes comprising the inflammatory secretome and others related to nociceptive tissue remodeling and to sensory perception of pain. Structural tissue changes, involving upregulation of multiple collagens occurred as soon as 1-hour postinjection, consistent with inflammatory tissue remodeling. Inflammatory expression profiling revealed a broad-spectrum, temporally orchestrated molecular and cellular recruitment process. The results provide numerous potential targets for modulation of pain and inflammation. PERSPECTIVE: This study investigates the highly orchestrated biological response during tissue inflammation with precise assessment of molecular dynamics at the transcriptional level. The results identify transcriptional changes that define an evolving inflammatory state in rats. This study provides foundational data for identifying markers of, and potential treatments for, inflammation and pain in patients.


Subject(s)
Gene Expression Profiling , Hyperalgesia/immunology , Immunity, Innate/immunology , Inflammation/immunology , Nociceptive Pain/immunology , Secretome/immunology , Animals , Carrageenan/pharmacology , Disease Models, Animal , Foot , Hyperalgesia/chemically induced , Inflammation/chemically induced , Male , Nociceptive Pain/chemically induced , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA
10.
Nat Commun ; 12(1): 2936, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006861

ABSTRACT

Host protection against cutaneous herpes simplex virus 1 (HSV-1) infection relies on the induction of a robust adaptive immune response. Here, we show that Nav1.8+ sensory neurons, which are involved in pain perception, control the magnitude of CD8 T cell priming and expansion in HSV-1-infected mice. The ablation of Nav1.8-expressing sensory neurons is associated with extensive skin lesions characterized by enhanced inflammatory cytokine and chemokine production. Mechanistically, Nav1.8+ sensory neurons are required for the downregulation of neutrophil infiltration in the skin after viral clearance to limit the severity of tissue damage and restore skin homeostasis, as well as for eliciting robust CD8 T cell priming in skin-draining lymph nodes by controlling dendritic cell responses. Collectively, our data reveal an important role for the sensory nervous system in regulating both innate and adaptive immune responses to viral infection, thereby opening up possibilities for new therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Nociceptive Pain/immunology , Sensory Receptor Cells/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cytokines/immunology , Cytokines/metabolism , Female , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/immunology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neutrophil Infiltration/immunology , Nociceptive Pain/genetics , Nociceptive Pain/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/virology , Skin/immunology , Skin/metabolism , Skin/virology
11.
Gene ; 742: 144583, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32184167

ABSTRACT

BACKGROUND: Studies showed that increased let-7b-5p microRNA during repeated electroacupuncture (EA) treatment was associated the formation of EA tolerance, which manifested as gradually decreased nociceptive threshold. Proenkephalin (PENK) is the precursor of enkephalin which is a pivot neuropeptide responsible for the decreased nociceptive threshold in EA. The aim of this study was to evaluate the relationship between let-7b-5p and PENK in EA tolerance. METHODS: The target gene of let-7b-5p microRNA was determined through the dual-luciferase reporter assay in cortical neurons. Seventy-two Sprague Dawley rats received a combination of EA and intracerebroventricular injection of microRNA (let-7b-5p agomir, antagomir or their controls). The nociceptive thresholds were assessed with radiant heat tail-flick method. PENK and let-7b-5p were measured with Western Blot and qPCR, respectively, after administration of let-7b-5p agomir, antagomir, and their controls at day 1, 4 and 7. RESULTS: Let-7b-5p targeted the 3' untranslated region of Penk1. The nociceptive thresholds in Let-7b-5p agomir + EA group were decreased (p < 0.05) compared with those in Let-7b-5p antagomir + EA group at day 1 to 7. Compared with Let-7b-5p agomir + EA group, the expression level of PENK in Let-7b-5p antagomir + EA group was increased at days 1, 4, and 7 (p < 0.05) CONCLUSION: Let-7b-5p may be a new potential target for decreasing the EA tolerance effect and facilitating the application of EA in treating chronic nociception of patients.


Subject(s)
Electroacupuncture , Enkephalins/genetics , MicroRNAs/metabolism , Nociceptive Pain/therapy , Protein Precursors/genetics , Animals , Antagomirs/administration & dosage , Disease Models, Animal , Down-Regulation , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Humans , Injections, Intraventricular , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Nociception/drug effects , Nociceptive Pain/diagnosis , Nociceptive Pain/genetics , Nociceptive Pain/immunology , Pain Threshold/drug effects , Rats
12.
Behav Brain Res ; 390: 112683, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32442548

ABSTRACT

Inflammatory pain is commonly associated with cognitive impairment. However, its molecular mechanisms are poorly understood. Thus, this study was conducted to investigate the molecular mechanisms of behavioral changes associated with inflammatory pain. Briefly, 36 Wistar rats were randomly divided into two main groups: CFA group treated with 100 µL of Complete Freunds' Adjuvant (CFA) and CFA + Minocycline group treated with 100 µL of CFA+40 mg/kg/day of minocycline). After that, each group was divided into three subgroups based on different time points of the study. The pain was induced using CFA and subsequent behavioral changes (i.e., hyperalgesia and learning and spatial memory) were analyzed by the Morris Water Maze (MWM) task and Radiant Heat. Then, the cellular and molecular changes were assessed using Western Blotting, Immunohistochemistry, and Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) techniques. Results of the study indicated that CFA-induced pain impaired spatial learning and memory functions. Studying the cellular changes showed that persistent inflammatory pain increased the microglial activity in CA1 and Dentate Gyrus (DG) regions. Furthermore, an increase was observed in the percentage of TUNEL-positive cells. Also, pro-Brain-Derived Neurotrophic Factor (BDNF)/BDNF ratio, Caspase3, and Receptor-Interacting Protein kinase 3 (RIP3) levels increased in the rats' hippocampus following induction of persistent inflammatory pain. These changes were reversed following the cessation of pain as well as the injection of minocycline. Taking together, the results of the current study for the first time revealed that an increase in the microglia dependent proBDNF/BDNF ratio following persistent inflammatory pain leads to cell death of the CA1 and DG neurons that subsequently causes a cognitive deficit in the learning and spatial memory functions.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal , Cognitive Dysfunction/etiology , Dentate Gyrus , Inflammation/complications , Microglia , Nociceptive Pain/complications , Spatial Memory , Animals , Anti-Bacterial Agents/administration & dosage , Behavior, Animal/physiology , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/physiopathology , Cell Death/physiology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Dentate Gyrus/immunology , Dentate Gyrus/physiopathology , Freund's Adjuvant/administration & dosage , Inflammation/immunology , Inflammation/metabolism , Male , Microglia/immunology , Microglia/metabolism , Minocycline/administration & dosage , Nociceptive Pain/immunology , Nociceptive Pain/metabolism , Rats , Rats, Wistar , Spatial Memory/physiology
13.
Eur J Pharm Sci ; 111: 38-45, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28943444

ABSTRACT

The current study investigated the effect of organoselenium compound p,p'-methoxyl-diphenyl diselenide [(OMePhSe)2], free or incorporated into nanocapsules, on behavioral, biochemical and molecular alterations in an inflammatory pain model induced by complete Freund's adjuvant (CFA). Male Swiss mice received an intraplantar injection of CFA in the hindpaw and 24 h later they were treated via the intragastric route with a single (OMePhSe)2 administration, in its free form (dissolved in canola oil) or (OMePhSe)2 NC. The anti-hypernociceptive time- and dose-response curves were carried out using the von Frey hair test. Biochemical and histological parameters were determined in samples of injected paws and those of cerebral contralateral cortex were collected to determine immuno content of inflammatory proteins. Both (OMePhSe)2 forms reduced the hypernociception induced by CFA as well as attenuated the altered parameters of the inflammatory process in the paw (paw edema, myeloperoxidase and histological). However, the (OMePhSe)2 NC had a more prolonged anti-hypernociceptive action (7h) at a lower dose (10mg/kg) and superior effects on the paw alterations than the free compound form (4h and 25mg/kg). Furthermore, independent of the (OMePhSe)2 form, its administration decreased the MAPKs pathway activation (JNK;ERK1,2; p38) as well as iNOS, COX-2, Nf-κB and IL-1ß protein contents in the cerebral contralateral cortex that were increased by paw CFA injection. Therefore, (OMePhSe)2 NC had superior anti-inflammatory action, which possibly occurs by the inflammatory protein content modulation and also attenuates paw biochemical and histological inflammatory alterations induced by CFA injection.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Behavior, Animal/drug effects , Drug Carriers/chemistry , Nanocapsules/chemistry , Nociceptive Pain/drug therapy , Organoselenium Compounds/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Inflammation , Male , Mice , Nociceptive Pain/enzymology , Nociceptive Pain/immunology , Organoselenium Compounds/administration & dosage , Pain Measurement , Peroxidase/metabolism , Time Factors
14.
Neuron ; 97(4): 806-822.e10, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29429934

ABSTRACT

Human autoantibodies to contactin-associated protein-like 2 (CASPR2) are often associated with neuropathic pain, and CASPR2 mutations have been linked to autism spectrum disorders, in which sensory dysfunction is increasingly recognized. Human CASPR2 autoantibodies, when injected into mice, were peripherally restricted and resulted in mechanical pain-related hypersensitivity in the absence of neural injury. We therefore investigated the mechanism by which CASPR2 modulates nociceptive function. Mice lacking CASPR2 (Cntnap2-/-) demonstrated enhanced pain-related hypersensitivity to noxious mechanical stimuli, heat, and algogens. Both primary afferent excitability and subsequent nociceptive transmission within the dorsal horn were increased in Cntnap2-/- mice. Either immune or genetic-mediated ablation of CASPR2 enhanced the excitability of DRG neurons in a cell-autonomous fashion through regulation of Kv1 channel expression at the soma membrane. This is the first example of passive transfer of an autoimmune peripheral neuropathic pain disorder and demonstrates that CASPR2 has a key role in regulating cell-intrinsic dorsal root ganglion (DRG) neuron excitability.


Subject(s)
Ganglia, Spinal/physiopathology , Immunoglobulin G/administration & dosage , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Nociceptive Pain/immunology , Nociceptive Pain/physiopathology , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Female , Humans , Immunization, Passive , Male , Mechanotransduction, Cellular , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Posterior Horn Cells/physiology , Shaker Superfamily of Potassium Channels/physiology
15.
Peptides ; 76: 73-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26774587

ABSTRACT

ARA 290 is an erythropoietin-derived polypeptide that possesses analgesic and tissue protective effect in many diseases such as diabetes and cancer. The analgesic effect of ARA 290 is mediated by its anti-inflammatory and immunomodulatory functions, or more specifically, by targeting the innate repair receptor (IRR) to down-regulate inflammation to alleviate neuropathic pain. However, whether other mechanisms or pathways are involved in ARA 290-mediated analgesic effect remains elusive. In this study, we are particularly interested in whether ARA 290 could directly target peripheral nociceptors by blocking or influencing receptors in pain sensation. Using calcium imaging, cell culture and behavioral tests, we demonstrated that ARA 290 was able to specifically inhibit TRPV1 channel activity, and relieve the mechanical hypersensitivity induced by capsaicin. Our study suggested that ARA 290 could potentially function as a novel antagonist for TRPV1 channel. This finding would not only contribute to the development of new pain treatment using ARA 290, but also help to improve our understanding of the integration between the immune system and the peripheral nervous system.


Subject(s)
Analgesics/pharmacology , Nociceptive Pain/drug therapy , Oligopeptides/pharmacology , TRPV Cation Channels/physiology , Animals , Calcium Signaling , Capsaicin/pharmacology , Drug Evaluation, Preclinical , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Nociception , Nociceptive Pain/immunology
16.
Arthritis Rheumatol ; 67(6): 1657-67, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25733371

ABSTRACT

OBJECTIVE: A hallmark of rheumatoid arthritis (RA) is the chronic pain that accompanies inflammation and joint deformation. Patients with RA rate pain relief as the highest priority; however, few studies have addressed the efficacy and safety of therapies directed specifically toward pain pathways. The ω-conotoxin MVIIA (ziconotide) is used in humans to alleviate persistent pain syndromes, because it specifically blocks the voltage-gated calcium 2.2 (CaV 2.2) channel, which mediates the release of neurotransmitters and proinflammatory mediators from peripheral nociceptor nerve terminals. The aims of this study were to investigate whether blockade of CaV 2.2 can suppress arthritis pain, and to examine the progression of induced arthritis during persistent CaV 2.2 blockade. METHODS: Transgenic mice expressing a membrane-tethered form of MVIIA under the control of a nociceptor-specific gene (MVIIA-transgenic mice) were used in the experiments. The mice were subjected to unilateral induction of joint inflammation using a combination of antigen and collagen. RESULTS: CaV 2.2 blockade mediated by tethered MVIIA effectively suppressed arthritis-induced pain; however, in contrast to their wild-type littermates, which ultimately regained use of their injured joint as inflammation subsided, MVIIA-transgenic mice showed continued inflammation, with up-regulation of the osteoclast activator RANKL and concomitant joint and bone destruction. CONCLUSION: Taken together, our results indicate that alleviation of peripheral pain by blockade of CaV 2.2- mediated calcium influx and signaling in nociceptor sensory neurons impairs recovery from induced arthritis and point to the potentially devastating effects of using CaV 2.2 channel blockers as analgesics during inflammation.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Calcium Channels, N-Type/metabolism , Nociceptive Pain/metabolism , Nociceptors/metabolism , RANK Ligand/metabolism , Stifle/metabolism , omega-Conotoxins/genetics , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/immunology , Disease Models, Animal , Mice , Mice, Transgenic , Nociceptive Pain/drug therapy , Nociceptive Pain/immunology , Stifle/pathology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , omega-Conotoxins/therapeutic use
17.
Inflammation ; 38(6): 2269-78, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26178479

ABSTRACT

The present study aimed to investigate the potential anti-inflammatory and anti-nociceptive activities of glycyrrhizin (GL) in mice and to explore the possible related mechanisms. Xylene-induced ear edema, carrageenan-induced paw edema and acetic acid-induced vascular permeability test were used to investigate the anti-inflammatory activities of GL in mice. Anti-nociceptive effects of GL were assessed by using acetic acid-induced writhing, hot plate test and formalin test, as well as evaluation of spontaneous locomotor activity and motor performance. The mRNA expression of pro-inflammatory cytokines (such as TNF-α, IL-6 and iNOS) and the protein expression of cyclooxygenase-2 (COX-2) were explored by using real-time fluorogenic PCR and Western blot, respectively. The results showed that GL significantly reduced xylene-induced ear edema, carrageenan-induced paw edema, and acetic acid-induced vascular permeation. Additionally, GL significantly inhibited the nociceptions induced by acetic acid and formalin. However, the nociceptions could not be decreased by GL in the hot plate test, and GL did not affect spontaneous locomotor activity and motor performance. The expression levels of TNF-α, IL-6, iNOS and COX-2 were significantly downregulated by GL. In conclusion, GL exerts significant anti-inflammatory and analgesic activities by attenuating the expression levels of TNF-α, IL-6, iNOS and COX-2.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Edema/prevention & control , Glycyrrhizic Acid/pharmacology , Inflammation Mediators/metabolism , Inflammation/prevention & control , Nociceptive Pain/prevention & control , Acetic Acid , Animals , Capillary Permeability/drug effects , Carrageenan , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/genetics , Edema/immunology , Edema/metabolism , Formaldehyde , Indomethacin/pharmacology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/immunology , Male , Mice, Inbred ICR , Motor Activity/drug effects , Nitric Oxide Synthase Type II/metabolism , Nociceptive Pain/chemically induced , Nociceptive Pain/genetics , Nociceptive Pain/immunology , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Pain Threshold/drug effects , Psychomotor Performance/drug effects , Time Factors , Xylenes
18.
Expert Opin Investig Drugs ; 22(10): 1295-303, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23902423

ABSTRACT

INTRODUCTION: Managing burn injury-associated pain and wounds is a major unresolved clinical problem. Opioids, nonsteroidal antiinflammatory drugs (NSAIDs), antidepressants and anticonvulsants remain the most common forms of analgesic therapy to treat burn patients. However, prolonged treatment with these drugs leads to dose escalation and serious side effects. Additionally, severe burn wounds cause scarring and are susceptible to infection. Recent encouraging findings demonstrate that curcumin, a major bioactive component found in turmeric, is a natural pharmacotherapeutic for controlling both severe burn pain and for improved wound healing. AREAS COVERED: This article covers current pr-clinical and clinical studies on the analgesic and wound healing effects. Particular emphasis has been placed on studies aimed at developing improved curcumin delivery vehicles that increase its bioavailability. Based on the available evidence, a hypothesis is proposed that the dual beneficial effects of curcumin, analgesia and enhanced wound healing are mediated through common anti-inflammatory mechanisms. EXPERT OPINION: Emerging studies have demonstrated that curcumin is a promising investigational drug to treat both pain and wounds. The adequate control of severe burn pain, particularly over the long courses required for healing, as well improvements in burn wound healing are unmet clinical needs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Burns/drug therapy , Curcumin/therapeutic use , Nociceptive Pain/drug therapy , Wound Healing/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Burns/immunology , Clinical Trials as Topic , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Drug Delivery Systems , Drug Evaluation, Preclinical , Nociceptive Pain/immunology , Wound Healing/immunology
SELECTION OF CITATIONS
SEARCH DETAIL