Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31813624

ABSTRACT

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Nociceptors/physiology , Animals , Epithelium/metabolism , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Nociceptors/metabolism , Peyer's Patches/innervation , Peyer's Patches/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
2.
Cell ; 173(5): 1083-1097.e22, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29754819

ABSTRACT

The nervous system, the immune system, and microbial pathogens interact closely at barrier tissues. Here, we find that a bacterial pathogen, Streptococcus pyogenes, hijacks pain and neuronal regulation of the immune response to promote bacterial survival. Necrotizing fasciitis is a life-threatening soft tissue infection in which "pain is out of proportion" to early physical manifestations. We find that S. pyogenes, the leading cause of necrotizing fasciitis, secretes streptolysin S (SLS) to directly activate nociceptor neurons and produce pain during infection. Nociceptors, in turn, release the neuropeptide calcitonin gene-related peptide (CGRP) into infected tissues, which inhibits the recruitment of neutrophils and opsonophagocytic killing of S. pyogenes. Botulinum neurotoxin A and CGRP antagonism block neuron-mediated suppression of host defense, thereby preventing and treating S. pyogenes necrotizing infection. We conclude that targeting the peripheral nervous system and blocking neuro-immune communication is a promising strategy to treat highly invasive bacterial infections. VIDEO ABSTRACT.


Subject(s)
Neurons/metabolism , Neutrophils/metabolism , Streptococcal Infections/pathology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Botulinum Toxins, Type A/administration & dosage , Calcitonin Gene-Related Peptide/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Diterpenes/pharmacology , Fasciitis, Necrotizing/etiology , Fasciitis, Necrotizing/pathology , Fasciitis, Necrotizing/veterinary , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neutrophils/immunology , Pain/etiology , Signal Transduction , Skin/metabolism , Skin/pathology , Streptococcal Infections/complications , Streptococcal Infections/veterinary , Streptococcus pyogenes/metabolism , Streptolysins/immunology , Streptolysins/metabolism , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
3.
Nature ; 615(7952): 472-481, 2023 03.
Article in English | MEDLINE | ID: mdl-36859544

ABSTRACT

The meninges are densely innervated by nociceptive sensory neurons that mediate pain and headache1,2. Bacterial meningitis causes life-threatening infections of the meninges and central nervous system, affecting more than 2.5 million people a year3-5. How pain and neuroimmune interactions impact meningeal antibacterial host defences are unclear. Here we show that Nav1.8+ nociceptors signal to immune cells in the meninges through the neuropeptide calcitonin gene-related peptide (CGRP) during infection. This neuroimmune axis inhibits host defences and exacerbates bacterial meningitis. Nociceptor neuron ablation reduced meningeal and brain invasion by two bacterial pathogens: Streptococcus pneumoniae and Streptococcus agalactiae. S. pneumoniae activated nociceptors through its pore-forming toxin pneumolysin to release CGRP from nerve terminals. CGRP acted through receptor activity modifying protein 1 (RAMP1) on meningeal macrophages to polarize their transcriptional responses, suppressing macrophage chemokine expression, neutrophil recruitment and dural antimicrobial defences. Macrophage-specific RAMP1 deficiency or pharmacological blockade of RAMP1 enhanced immune responses and bacterial clearance in the meninges and brain. Therefore, bacteria hijack CGRP-RAMP1 signalling in meningeal macrophages to facilitate brain invasion. Targeting this neuroimmune axis in the meninges can enhance host defences and potentially produce treatments for bacterial meningitis.


Subject(s)
Brain , Meninges , Meningitis, Bacterial , Neuroimmunomodulation , Humans , Brain/immunology , Brain/microbiology , Calcitonin Gene-Related Peptide/metabolism , Meninges/immunology , Meninges/microbiology , Meninges/physiopathology , Pain/etiology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Meningitis, Bacterial/complications , Meningitis, Bacterial/immunology , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/pathogenicity , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , Nociceptors/metabolism , Receptor Activity-Modifying Protein 1/metabolism , Macrophages/immunology , Macrophages/metabolism
4.
Trends Immunol ; 45(5): 381-396, 2024 May.
Article in English | MEDLINE | ID: mdl-38697871

ABSTRACT

Recent studies have uncovered a new role for sensory neurons in influencing mammalian host immunity, challenging conventional notions of the nervous and immune systems as separate entities. In this review we delve into this groundbreaking paradigm of neuroimmunology and discuss recent scientific evidence for the impact of sensory neurons on host responses against a wide range of pathogens and diseases, encompassing microbial infections and cancers. These valuable insights enhance our understanding of the interactions between the nervous and immune systems, and also pave the way for developing candidate innovative therapeutic interventions in immune-mediated diseases highlighting the importance of this interdisciplinary research field.


Subject(s)
Sensory Receptor Cells , Animals , Humans , Host-Pathogen Interactions , Immunity , Neoplasms/immunology , Neoplasms/therapy , Neuroimmunomodulation , Sensory Receptor Cells/immunology , Sensory Receptor Cells/physiology
5.
Nature ; 590(7846): 473-479, 2021 02.
Article in English | MEDLINE | ID: mdl-33408417

ABSTRACT

Astrocytes are glial cells that are abundant in the central nervous system (CNS) and that have important homeostatic and disease-promoting functions1. However, little is known about the homeostatic anti-inflammatory activities of astrocytes and their regulation. Here, using high-throughput flow cytometry screening, single-cell RNA sequencing and CRISPR-Cas9-based cell-specific in vivo genetic perturbations in mice, we identify a subset of astrocytes that expresses the lysosomal protein LAMP12 and the death receptor ligand TRAIL3. LAMP1+TRAIL+ astrocytes limit inflammation in the CNS by inducing T cell apoptosis through TRAIL-DR5 signalling. In homeostatic conditions, the expression of TRAIL in astrocytes is driven by interferon-γ (IFNγ) produced by meningeal natural killer (NK) cells, in which IFNγ expression is modulated by the gut microbiome. TRAIL expression in astrocytes is repressed by molecules produced by T cells and microglia in the context of inflammation. Altogether, we show that LAMP1+TRAIL+ astrocytes limit CNS inflammation by inducing T cell apoptosis, and that this astrocyte subset is maintained by meningeal IFNγ+ NK cells that are licensed by the microbiome.


Subject(s)
Astrocytes/immunology , Gastrointestinal Microbiome/immunology , Inflammation/prevention & control , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Lysosomal Membrane Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Astrocytes/metabolism , Biomarkers , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Homeostasis , Humans , Inflammation/immunology , Meninges/cytology , Meninges/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/immunology
6.
Trends Immunol ; 38(1): 5-19, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27793571

ABSTRACT

Nociceptor sensory neurons protect organisms from danger by eliciting pain and driving avoidance. Pain also accompanies many types of inflammation and injury. It is increasingly clear that active crosstalk occurs between nociceptor neurons and the immune system to regulate pain, host defense, and inflammatory diseases. Immune cells at peripheral nerve terminals and within the spinal cord release mediators that modulate mechanical and thermal sensitivity. In turn, nociceptor neurons release neuropeptides and neurotransmitters from nerve terminals that regulate vascular, innate, and adaptive immune cell responses. Therefore, the dialog between nociceptor neurons and the immune system is a fundamental aspect of inflammation, both acute and chronic. A better understanding of these interactions could produce approaches to treat chronic pain and inflammatory diseases.


Subject(s)
Immune System , Inflammation/immunology , Neuroimmunomodulation , Nociceptors/metabolism , Pain/immunology , Sensory Receptor Cells/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism
7.
J Neuroinflammation ; 16(1): 113, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31138231

ABSTRACT

BACKGROUND: The cellular and molecular pathophysiological mecha\nisms of pain processing in neglected parasitic infections such as leishmaniasis remain unknown. The present study evaluated the participation of spinal cord glial cells in the pathophysiology of pain induced by Leishmania amazonensis infection in BALB/c mice. METHODS: Mice received intra-plantar (i.pl.) injection of L. amazonensis (1 × 105) and hyperalgesia, and paw edema were evaluated bilaterally for 40 days. The levels of TNF-α and IL-1ß, MPO activity, and histopathology were assessed on the 40th day. ATF3 mRNA expression was assessed in DRG cells at the 30th day post-infection. Blood TNF-α and IL-1ß levels and systemic parasite burden were evaluated 5-40 days after the infection. At the 30th day post-infection L. amazonensis, the effects of intrathecal (i.t.) treatments with neutralizing antibody anti-CX3CL1, etanercept (soluble TNFR2 receptor), and interleukin-1 receptor antagonist (IL-1ra) on infection-induced hyperalgesia and paw edema were assessed. In another set of experiments, we performed a time course analysis of spinal cord GFAP and Iba-1 (astrocytes and microglia markers, respectively) and used confocal immunofluorescence and Western blot to confirm the expression at the protein level. Selective astrocyte (α-aminoadipate) and microglia (minocycline) inhibitors were injected i.t. to determine the contribution of these cells to hyperalgesia and paw edema. The effects of i.t. treatments with glial and NFκB (PDTC) inhibitors on spinal glial activation, TNF-α, IL-1ß, CX3CR1 and CX3CL1 mRNA expression, and NFκB activation were also evaluated. Finally, the contribution of TNF-α and IL-1ß to CX3CL1 mRNA expression was investigated. RESULTS: L. amazonensis infection induced chronic mechanical and thermal hyperalgesia and paw edema in the infected paw. Mechanical hyperalgesia was also observed in the contralateral paw. TNF-α, IL-1ß, MPO activity, and epidermal/dermal thickness increased in the infected paw, which confirmed the peripheral inflammation at the primary foci of this infection. ATF3 mRNA expression at the ipsilateral DRG of the infected paw was unaltered 30 days post-infection. TNF-α and IL-1ß blood levels were not changed over the time course of disease, and parasitism increased in a time-dependent manner in the ipsilateral draining lymph node. Treatments targeting CX3CL1, TNF-α, and IL-1ß inhibited L. amazonensis-induced ongoing mechanical and thermal hyperalgesia, but not paw edema. A time course of GFAP, Iba-1, and CX3CR1 mRNA expression indicated spinal activation of astrocytes and microglia, which was confirmed at the GFAP and Iba-1 protein level at the peak of mRNA expression (30th day). Selective astrocyte and microglia inhibition diminished infection-induced ipsilateral mechanical hyperalgesia and thermal hyperalgesia, and contralateral mechanical hyperalgesia, but not ipsilateral paw edema. Targeting astrocytes, microglia and NFκB diminished L. amazonensis-induced GFAP, Iba-1, TNF-α, IL-1ß, CX3CR1 and CX3CL1 mRNA expression, and NFκB activation in the spinal cord at the peak of spinal cord glial cells activation. CX3CL1 mRNA expression was also detected in the ipsilateral DRG of infected mice at the 30th day post-infection, and the i.t. injection of TNF-α or IL-1ß in naïve animals induced CX3CL1 mRNA expression in the spinal cord and ipsilateral DRG. CONCLUSIONS: L. amazonensis skin infection produces chronic pain by central mechanisms involving spinal cord astrocytes and microglia-related production of cytokines and chemokines, and NFκB activation contributes to L. amazonensis infection-induced hyperalgesia and neuroinflammation.


Subject(s)
Edema/pathology , Hyperalgesia/pathology , Leishmaniasis/pathology , Neuroglia/pathology , Pain/pathology , Spinal Cord/pathology , Animals , Edema/microbiology , Hyperalgesia/microbiology , Leishmania , Male , Mice , Mice, Inbred BALB C , Neuroglia/microbiology , Pain/microbiology , Spinal Cord/microbiology
8.
Inflammopharmacology ; 27(6): 1229-1242, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30612217

ABSTRACT

BACKGROUND: Naringenin is a biologically active analgesic, anti-inflammatory, and antioxidant flavonoid. Naringenin targets in inflammation-induced articular pain remain poorly explored. METHODS: The present study investigated the cellular and molecular mechanisms involved in the analgesic/anti-inflammatory effects of naringenin in zymosan-induced arthritis. Mice were pre-treated orally with naringenin (16.7-150 mg/kg), followed by intra-articular injection of zymosan. Articular mechanical hyperalgesia and oedema, leucocyte recruitment to synovial cavity, histopathology, expression/production of pro- and anti-inflammatory mediators and NFκB activation, inflammasome component expression, and oxidative stress were evaluated. RESULTS: Naringenin inhibited articular pain and oedema in a dose-dependent manner. The dose of 50 mg/kg inhibited leucocyte recruitment, histopathological alterations, NFκB activation, and NFκB-dependent pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-33), and preproET-1 mRNA expression, but increased anti-inflammatory IL-10. Naringenin also inhibited inflammasome upregulation (reduced Nlrp3, ASC, caspase-1, and pro-IL-1ß mRNA expression) and oxidative stress (reduced gp91phox mRNA expression and superoxide anion production, increased GSH levels, induced Nrf2 protein in CD45+ hematopoietic recruited cells, and induced Nrf2 and HO-1 mRNA expression). CONCLUSIONS: Naringenin presents analgesic and anti-inflammatory effects in zymosan-induced arthritis by targeting its main physiopathological mechanisms. These data highlight this flavonoid as an interesting therapeutic compound to treat joint inflammation, deserving additional pre-clinical and clinical studies.


Subject(s)
Arthritis/drug therapy , Flavanones/therapeutic use , Leukocyte Common Antigens/analysis , NF-E2-Related Factor 2/physiology , Zymosan/pharmacology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Flavanones/pharmacology , Hematopoietic Stem Cells/metabolism , Inflammasomes/drug effects , Knee Joint/pathology , Male , Mice , NF-E2-Related Factor 2/genetics , NF-kappa B/physiology , Oxidative Stress/drug effects , Signal Transduction
9.
Photochem Photobiol Sci ; 16(7): 1162-1173, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28594010

ABSTRACT

trans-Chalcone is a plant flavonoid precursor, which lacks broad investigation on its biological activity in inflammatory processes. In the present study, anti-inflammatory and antioxidant mechanisms of systemic administration with trans-chalcone, a flavonoid precursor, on ultraviolet (UV) irradiation-induced skin inflammation and oxidative stress in hairless mice were investigated by the following parameters: skin edema, myeloperoxidase activity (neutrophil marker), matrix metalloproteinase-9 activity, reduced glutathione levels, catalase activity, lipid peroxidation products, superoxide anion production, gp91phox (NADPH oxidase subunit) mRNA expression by quantitative PCR and cytokine production by ELISA. Systemic treatment with trans-chalcone inhibited skin inflammation by reducing skin edema and neutrophil recruitment, and also inhibited matrix metalloproteinase-9 activity. trans-Chalcone also inhibited oxidative stress, gp91phox mRNA expression, and the production of a wide range of pro-inflammatory cytokines, while it did not affect anti-inflammatory cytokines induced by UV irradiation. However, trans-chalcone did not prevent oxidative stress in vitro, suggesting that its in vivo effect is more related to anti-inflammatory properties rather than a direct antioxidant effect. In conclusion, treatment with trans-chalcone inhibited UV-induced skin inflammation resulting in oxidative stress inhibition in vivo. Therefore, systemic supplementation with this compound may represent an important therapeutic approach in inflammatory skin diseases induced by UV irradiation.


Subject(s)
Chalcone/pharmacology , Cytokines/biosynthesis , Inflammation/prevention & control , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Skin/drug effects , Skin/radiation effects , Ultraviolet Rays , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chalcone/chemistry , Dose-Response Relationship, Drug , Edema/drug therapy , Enzyme-Linked Immunosorbent Assay , Inflammation/metabolism , Male , Mice , Mice, Hairless , Molecular Structure , Oxidative Stress/radiation effects , Skin/metabolism , Skin/pathology , Structure-Activity Relationship
10.
Photochem Photobiol Sci ; 15(4): 554-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27021784

ABSTRACT

Skin exposure to ultraviolet B (UVB) irradiation has increased significantly in recent years due to ozone depletion, and it represents the main cause of many skin diseases. Hesperidin methyl chalcone (HMC) is a compound used to treat vascular diseases that has demonstrated anti-inflammatory activities in pre-clinical studies. Herein, we tested the antioxidant activity of HMC in cell free systems and the in vivo effects of a stable topical formulation containing HMC in a mouse model of skin oxidative stress and inflammation induced by UVB irradiation. HMC presented ferric reducing power, neutralized 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and hydroxyl free radicals, and inhibited lipid peroxidation. In hairless mice, a topical formulation containing HMC inhibited UVB irradiation-induced skin edema, depletion of antioxidant capacity (ferric and ABTS reducing abilities and catalase activity), lipid peroxidation, superoxide anion production and mRNA expression of gp91phox (nicotinamide adenine dinucleotide phosphate [NADPH] oxidase 2 sub-unity). In addition, HMC inhibited UVB irradiation-induced depletion of reduced glutathione levels by maintaining glutathione peroxidase-1 and glutathione reductase mRNA expression, prevented down-regulation of nuclear factor erythroid 2-related factor 2 (Nrf2) mRNA expression and increased heme oxygenase-1 mRNA expression. Finally, we demonstrated that topical application of the formulation containing HMC inhibited cytokine (TNF-α, IL-1ß, IL-6, and IL-10) production induced by UVB irradiation. Therefore, this topical formulation containing HMC is a promising new therapeutic approach to protecting the skin from the deleterious effects of UVB irradiation.


Subject(s)
Chalcones/administration & dosage , Hesperidin/analogs & derivatives , Inflammation/prevention & control , Oxidative Stress/drug effects , Skin/drug effects , Ultraviolet Rays , Administration, Topical , Animals , Chalcones/pharmacology , Cytokines/biosynthesis , Heme Oxygenase-1/genetics , Hesperidin/administration & dosage , Hesperidin/pharmacology , Inflammation/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Hairless , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-E2-Related Factor 2/genetics , RNA, Messenger/genetics , Skin/metabolism , Superoxides/metabolism
11.
J Nat Prod ; 79(5): 1329-38, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27191910

ABSTRACT

Evidence shows beneficial effects of resveratrol (RES) on human health. However, its poor aqueous solubility limits therapeutic effectiveness. Thus, the use of nanostructured delivery systems for RES, such as a liquid-crystalline system (LCS), could be viable. The purpose of this study was to develop, characterize, and determine the in vivo effectiveness of a RES-loaded LCS. We studied an LCS containing silicon glycol copolymer, polyether functional siloxane, and the polymeric dispersion carbomer homopolymer type B (C974) in the ratio 20:55:25 with and without RES. Results obtained using polarized light microscopy, small-angle X-ray scattering, and rheology analysis showed that the RES-loaded LCS system presents a lamellar structure and behaves as a non-Newtonian fluid presenting pseudoplastic (the apparent viscosity decreases as the stress increases) and thixotropic (the apparent viscosity decreases with the duration of stress) behaviors. Cytotoxicity studies showed that the formulation components are noncytotoxic. Topical application of a RES-loaded LCS protected hairless mice from UVB-irradiation-induced skin damage by inhibiting edema, neutrophil recruitment, lipid hydroperoxide and superoxide anion production, gp91phox mRNA expression, and oxidative stress. The RES-loaded LCS maintained 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and ferric reducing abilities, catalase activity, reduced glutathione levels, and mRNA expression of glutathione peroxidase 1 and glutathione reductase. The RES-loaded LCS also up-regulated matrix metalloproteinase-9 activity, IL-10 production, and mRNA expression of transcription factor Nrf2 and heme oxygenase-1. Therefore, a RES-loaded LCS is a promising new therapeutic approach to mitigate skin photodamage.


Subject(s)
Oxidative Stress/radiation effects , Skin/drug effects , Stilbenes/pharmacology , Ultraviolet Rays/adverse effects , Animals , Antioxidants/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Edema , Female , Glutathione/metabolism , Glutathione Peroxidase , Heme Oxygenase-1/metabolism , Humans , Inflammation/metabolism , Interleukin-10/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Hairless , Molecular Structure , Resveratrol , Stilbenes/chemistry , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology , Superoxides/metabolism , Glutathione Peroxidase GPX1
12.
Molecules ; 21(7)2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27367653

ABSTRACT

In this review, we discuss the importance of capsaicin to the current understanding of neuronal modulation of pain and explore the mechanisms of capsaicin-induced pain. We will focus on the analgesic effects of capsaicin and its clinical applicability in treating pain. Furthermore, we will draw attention to the rationale for other clinical therapeutic uses and implications of capsaicin in diseases such as obesity, diabetes, cardiovascular conditions, cancer, airway diseases, itch, gastric, and urological disorders.


Subject(s)
Capsaicin/pharmacology , Capsaicin/therapeutic use , Pain/drug therapy , Analgesics/chemistry , Analgesics/isolation & purification , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Capsaicin/chemistry , Capsaicin/isolation & purification , Capsicum/chemistry , Clinical Studies as Topic , Drug Compounding , Drug Evaluation, Preclinical , Drug Repositioning , Humans , Nociceptors/drug effects , Nociceptors/metabolism , Pain/etiology , Pain/metabolism , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
13.
Inflammopharmacology ; 24(2-3): 97-107, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27160222

ABSTRACT

We evaluated the effect of pyrrolidine dithiocarbamate (PDTC) in superoxide anion-induced inflammatory pain. Male Swiss mice were treated with PDTC and stimulated with an intraplantar or intraperitoneal injection of potassium superoxide, a superoxide anion donor. Subcutaneous PDTC treatment attenuated mechanical hyperalgesia, thermal hyperalgesia, paw oedema and leukocyte recruitment (neutrophils and macrophages). Intraplantar injection of superoxide anion activated NF-κB and increased cytokine production (IL-1ß, TNF-α and IL-10) and oxidative stress (nitrite and lipid peroxidation levels) at the primary inflammatory foci and in the spinal cord (L4-L6). PDTC treatment inhibited superoxide anion-induced NF-κB activation, cytokine production and oxidative stress in the paw and spinal cord. Furthermore, intrathecal administration of PDTC successfully inhibited superoxide anion-induced mechanical hyperalgesia, thermal hyperalgesia and inflammatory response in peripheral foci (paw). These results suggest that peripheral stimulus with superoxide anion activates the local and spinal cord oxidative- and NF-κB-dependent inflammatory nociceptive mechanisms. PDTC targets these events, therefore, inhibiting superoxide anion-induced inflammatory pain in mice.


Subject(s)
Drug Delivery Systems/methods , NF-kappa B/metabolism , Oxidative Stress/drug effects , Pain/metabolism , Pyrrolidines/administration & dosage , Spinal Cord/metabolism , Thiocarbamates/administration & dosage , Animals , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/metabolism , Edema/prevention & control , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , NF-kappa B/antagonists & inhibitors , Oxidative Stress/physiology , Pain/chemically induced , Pain/prevention & control , Spinal Cord/drug effects , Superoxides/toxicity
14.
Exp Physiol ; 100(5): 531-44, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25711612

ABSTRACT

NEW FINDINGS: What is the central question of this study? This study investigated the role of the endogenous anti-inflammatory cytokine interleukin-10 in intense acute swimming-induced muscle mechanical hyperalgesia in mice. What is the main finding and its importance? Endogenous interleukin-10 has a key role in limiting exercise-induced muscle pain in a model presenting similarities to delayed-onset muscle soreness in mice. Interleukin-10 reduced muscle pain by diminishing leucocyte recruitment, hyperalgesic cytokine production, oxidative stress and myocyte damage. Interleukin-10 (IL-10) is an antihyperalgesic cytokine. In this study, IL-10-deficient (IL-10(-/-) ) mice were used to investigate the role of endogenous IL-10 in intense acute swimming-induced muscle mechanical hyperalgesia, which presents similarities with delayed-onset muscle soreness. An intense acute swimming session of 1 or 2 h induced significant muscle mechanical hyperalgesia in a time-dependent manner in wild-type mice compared with the sham group 24 h after the session, which was further increased in IL-10(-/-) mice (P Ë‚ 0.05). Intraperitoneal treatment of wild-type mice with IL-10 (1-10 ng) reduced muscle mechanical hyperalgesia in a dose-dependent manner and reversed the enhanced muscle hyperalgesia in IL-10(-/-) mice (P Ë‚ 0.05). The 2 h swimming session induced increases in tumour necrosis factor-α, interleukin-1ß and IL-10 production in the soleus muscle. However, tumour necrosis factor-α and interleukin-1ß production in the soleus muscle were even higher in IL-10(-/-) mice between 2 and 6 h after the stimulus (P Ë‚ 0.05). There was no statistical difference in the levels of the antihyperalgesic cytokines interleukin-4, interleukin-5, interleukin-13 and transforming growth factor-ß between wild-type and IL-10(-/-) mice (P Ëƒ 0.05). Interleukin-10 deficiency also resulted in increased myeloperoxidase activity, greater depletion of reduced glutathione levels, increased superoxide anion production and the maintenance of high plasma concentrations of creatine kinase (until 24 h after the swimming session) in soleus muscle (P Ë‚ 0.05). These results demonstrate that endogenous IL-10 controls intense acute swimming-induced muscle mechanical hyperalgesia by limiting oxidative stress and cytokine production.


Subject(s)
Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Interleukin-10/metabolism , Muscle, Skeletal/physiopathology , Myalgia/chemically induced , Swimming , Animals , Inflammation/metabolism , Interleukin-10/genetics , Male , Mice, Inbred C57BL , Muscle Cells/cytology , Oxidative Stress/physiology
15.
Inflamm Res ; 64(12): 993-1003, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26456836

ABSTRACT

OBJECTIVE: This study aimed at evaluating the activity of curcumin in superoxide anion-induced pain-like behavior and leukocyte recruitment in mice. TREATMENT: Administration of curcumin 10 mg/kg subcutaneously 1 h before stimulus. METHODS: KO2 was used as superoxide anion donor. Overt pain-like behaviors were determined by the number of abdominal writhings, paw flinches and time spent licking the paw. Mechanical and thermal hyperalgesia were determined using an electronic anesthesiometer and hot plate, respectively. Cytokine concentration and NF-κB activity were determined by ELISA, antioxidant effect by nitrobluetretrazolium assay and ABTS radical scavenging ability. Myeloperoxidase activity was measured by colorimetric assay. The Nrf2, heme oxygenase-1 (HO-1) and gp91phox mRNA expression was determined by quantitative PCR. Data were analyzed by ANOVA followed by Tukey's post hoc and considered significant when p<0.05. RESULTS: Curcumin inhibited superoxide anion-induced overt pain-like behaviors as well as mechanical and thermal hyperalgesia. Curcumin also inhibited superoxide anion-induced leukocyte recruitment in the peritoneal cavity and in the paw skin inhibited myeloperoxidase activity, oxidative stress, IL-1ß and TNF-α production and NF-κB activation as well as enhanced IL-10 production, and HO-1 and Nrf2 mRNA expression. CONCLUSION: Curcumin inhibits superoxide anion-induced inflammatory pain-like behaviors and leukocyte recruitment by targeting inflammatory molecules and oxidative stress; and inducing antioxidant and anti-inflammatory pathways.


Subject(s)
Antioxidants/pharmacology , Curcumin/pharmacology , Leukocytes/drug effects , NF-E2-Related Factor 2/biosynthesis , NF-kappa B/antagonists & inhibitors , Oxidants/toxicity , Pain/psychology , Superoxides/antagonists & inhibitors , Superoxides/toxicity , Animals , Antioxidants/administration & dosage , Curcumin/administration & dosage , Cytokines/biosynthesis , Heme Oxygenase-1/biosynthesis , Hyperalgesia/chemically induced , Hyperalgesia/prevention & control , Injections, Subcutaneous , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , NADPH Oxidases/genetics , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxides/chemistry , Pain/chemically induced , Potassium Compounds/chemistry , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
16.
J Nat Prod ; 78(8): 1799-808, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26192250

ABSTRACT

Vanillic acid (1) is a flavoring agent found in edible plants and fruits. It is an oxidized form of vanillin. Phenolic compounds form a substantial part of plant foods used as antioxidants with beneficial biological activities. These compounds have received considerable attention because of their role in preventing human diseases. Especially, 1 presents antibacterial, antimicrobial, and chemopreventive effects. However, the mechanisms by which 1 exerts its anti-inflammatory effects in vivo are incompletely understood. Thus, the effect of 1 was evaluated in murine models of inflammatory pain. Treatment with 1 inhibited the overt pain-like behavior induced by acetic acid, phenyl-p-benzoquinone, the second phase of the formalin test, and complete Freund's adjuvant (CFA). Treatment with 1 also inhibited carrageenan- and CFA-induced mechanical hyperalgesia, paw edema, myeloperoxidase activity, and N-acetyl-ß-D-glucosaminidase activity. The anti-inflammatory mechanisms of 1 involved the inhibition of oxidative stress, pro-inflammatory cytokine production, and NFκB activation in the carrageenan model. The present study demonstrated 1 presents analgesic and anti-inflammatory effects in a wide range of murine inflammation models, and its mechanisms of action involves antioxidant effects and NFκB-related inhibition of pro-inflammatory cytokine production.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , NF-kappa B/drug effects , Neutrophil Infiltration/drug effects , Oxidative Stress/drug effects , Vanillic Acid/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/pharmacology , Benzaldehydes/chemistry , Benzoquinones/pharmacology , Carrageenan/adverse effects , Cytokines/biosynthesis , Disease Models, Animal , Edema/chemically induced , Freund's Adjuvant/pharmacology , Hyperalgesia/drug therapy , Inflammation/chemically induced , Male , Mice , Molecular Structure , Pain/drug therapy , Signal Transduction/drug effects , Vanillic Acid/chemistry
17.
J Nat Prod ; 78(7): 1647-55, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26154512

ABSTRACT

Ultraviolet B (UVB) irradiation may cause inflammation- and oxidative-stress-dependent skin cancer and premature aging. Naringenin (1) has been reported to have anti-inflammatory and antioxidant properties, but its effects and mechanisms on UVB irradiation-induced inflammation and oxidative stress are still not known. Thus, the present study aimed to investigate the potential of naringenin to mitigate UVB irradiation-induced inflammation and oxidative damage in the skin of hairless mice. Skin edema, myeloperoxidase (neutrophil marker) and matrix metalloproteinase-9 (MMP-9) activity, and cytokine production were measured after UVB irradiation. Oxidative stress was evaluated by 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) radical (ABTS) scavenging ability, ferric reducing antioxidant power (FRAP), reduced glutathione levels, catalase activity, lipid peroxidation products, superoxide anion production, and gp91phox (NADPH oxidase subunit) mRNA expression by quantitative PCR. The intraperitoneal treatment with naringenin reduced skin inflammation by inhibiting skin edema, neutrophil recruitment, MMP-9 activity, and pro-inflammatory (TNF-α, IFN-γ, IL-1ß, IL-4, IL-5, IL-6, IL-12, IL-13, IL-17, IL-22, and IL-23) and anti-inflammatory (TGF-ß and IL-10) cytokines. Naringenin also inhibited oxidative stress by reducing superoxide anion production and the mRNA expression of gp91phox. Therefore, naringenin inhibits UVB irradiation-induced skin damage and may be a promising therapeutic approach to control skin disease.


Subject(s)
Flavanones/pharmacology , Oxidative Stress/drug effects , Skin/drug effects , Ultraviolet Rays , Animals , Antioxidants/pharmacology , Benzothiazoles/pharmacology , Flavanones/chemistry , Glutathione/metabolism , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-12/pharmacology , Interleukin-17 , Interleukin-4 , Interleukin-6/metabolism , Interleukins , Lipid Peroxidation/drug effects , Male , Mice , Mice, Hairless , Molecular Structure , Oxidative Stress/radiation effects , Skin/radiation effects , Sulfonic Acids/pharmacology , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Interleukin-22
18.
Neuron ; 112(9): 1381-1383, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38697021

ABSTRACT

Infections frequently cause behavioral changes, known as sickness behavior. In a recent study,1 Yipp and collaborators discovered a sensory circuit that is activated by a bacterial lipopolysaccharide during lung infection and drives sickness behaviors independent of inflammation. Biofilm-producing bacteria, however, avoid activating this lung-brain circuit, resulting in infection without sickness behavior.


Subject(s)
Illness Behavior , Animals , Humans , Illness Behavior/physiology , Lipopolysaccharides , Brain , Biofilms , Nerve Net/physiology
19.
Int J Exerc Sci ; 17(6): 670-681, 2024.
Article in English | MEDLINE | ID: mdl-38863769

ABSTRACT

Smartpaddle® is a novel wearable device based on inertial measurement units (IMU) for in-field arm-stroke kinetics and kinematics analysis in swimming. However, the lack of data regarding its agreement and reliability, coupled with restricted access to raw data, emphasizes the need to evaluate it against a well-established strain gauge (SG) reference method for assessing swimming forces. Thus, this study aimed to investigate the agreement and reliability between the Smartpaddle® and strain gauge in a 30-s all-out arms-only tethered swimming test. Twelve trained young adult swimmers performed a test-retest 30-s all-out arms-only tethered swimming trial. Peak and mean forces were obtained from IMU (PFIMU and MFIMU) and SG (PFSG and MFSG) simultaneously. Statistical differences and correlations were found in both peak (PFSG = 158.46 ± 48.85 N, PFIMU = 75.47 ± 12.05 N, p < 0.001, r = 0.88) and mean (MFSG = 69.62 ± 16.36 N, MFIMU = 30.06 ± 5.42 N, p < 0.001, r = 0.84) forces between devices, presenting elevated systematic errors for both variables. No differences were found in IMU data between test-retest conditions in both peak (PFIMU = 75.47 ± 12.05 N, PFIMU = 75.45 ± 11.54 N, p = 0.99, ICC = 0.96) and mean (MFIMU = 30.06 ± 5.42 N, MFIMU = 30.21 ± 5.83 N, p = 0.80, ICC = 0.95) forces, with negligible systematic errors. In conclusion, although the Smartpaddle® device is not directly comparable to the strain gauge reference method, it has demonstrated high reliability levels in test-retest trials.

20.
Int J Exerc Sci ; 17(2): 648-659, 2024.
Article in English | MEDLINE | ID: mdl-38864027

ABSTRACT

This study aimed to investigate the effects of chronic ß-alanine (ßA) plus acute sodium bicarbonate (SB) co-supplementation on neuromuscular fatigue during high-intensity intermittent efforts in swimming. Eleven regional and national competitive-level young swimmers performed a neuromuscular fatigue assessment before and immediately after two 20 × 25-m front crawl maximal efforts every 90 s, performed at pre- and post-4-week co-supplementation. Neuromuscular fatigue was evaluated by percutaneous electrical stimuli through the twitch interpolation technique on the triceps brachii and quadriceps femoris. Performance was defined by the mean time of the 20 efforts and blood samples to lactate concentrations were collected every four efforts. Participants supplemented 3.2-6.4 g·day-1 of chronic ßA or placebo (PL) during four weeks, and acute 0.3 g·kg-1 of SB or PL 60 min before the second assessment (allowing ßA+SB and PL+PL groups). No statistical changes were found in neuromuscular fatigue of triceps brachii. In the quadriceps femoris, a main effect of time was found in potentiated twitch delta values in pooled groups, showing a statistical increase of 19.01% after four weeks (Δ = 13.05 [0.35-25.75] N; p = 0.044), without time × group interactions. No statistical difference was found in the swimming performance. Blood lactate increased by 25.06% only in the ßA+SB group (Δ = 6.40 [4.62-8.18] mM; p Bonf < 0.001) after the supplementation period. In conclusion, 4-week ßA and SB co-supplementation were not able to reduce neuromuscular fatigue levels and improve performance in highintensity intermittent efforts, but statistically increased blood lactate levels.

SELECTION OF CITATIONS
SEARCH DETAIL