Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Periodontal Res ; 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39373294

ABSTRACT

AIM: This study investigated the activity and mechanism of action of the iron tetracarboxyphthalocyanine (FeTcPc) on tumor necrosis factor alpha (TNF-α) production and its impact on experimental periodontitis. METHODS: RAW 264.7 macrophages were treated with FeTcPc, activated with lipopolysaccharide (LPS) at 10 ng/mL, and the TNF-α levels were measured, as well as the nuclear factor kappa B (NF-κB) activation. Subsequently, a mouth gel containing 1% FeTcPc was topically administered to the gingival tissue of mice with periodontitis-induced ligatures. Bone loss and the gene expression of Tnfα, p65 (NF-κB), and receptor-activating nuclear factor kappa B ligand (Rankl) were quantified in gingival tissue. Finally, the systemic toxicity of FeTcPc was estimated in Galleria mellonella larvae. RESULTS: In an activated RAW 264.7 macrophage culture, 100 µM FeTcPc reduced TNF-α release and NF-κB activation. Regarding experimental periodontitis, topical application of mouth gel containing 1% FeTcPc blocked alveolar bone loss. Additionally, 1% FeTcPc reduced the expression of Tnfα, p65 (NF-κB), and Rankl in gingival tissue. Finally, administration FeTcPc at doses ranging from 1 to 1000 mg/kg did not cause acute systemic toxicity in G. mellonella. CONCLUSION: Overall, we demonstrated the potential of mouth gel containing FeTcPc as a therapeutic strategy for managing osteolytic inflammatory disorders, such as periodontitis.

2.
Inflamm Res ; 72(1): 107-114, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36333479

ABSTRACT

OBJECTIVE: In this study, we investigated the modulatory effects of PI3Kγ on IL-17A expression and the progression of experimental periodontitis in vivo. METHODS: Ligature-induced periodontitis was developed around the first molar of mice. Animals were treated with anti-mouse IL-17A or IPI-549 (PI3Kγ inhibitor). In addition, PI3Kγ-deficient mice (PI3Kγ-/-) were used in the study. Alveolar bone loss was measured and real-time PCR of Il17a and Rankl genes was performed. A bioinformatics analysis was carried out using the Gene Set Enrichment Analysis computational tool. RESULTS: Nine days after ligature placement, alveolar bone loss scores were significantly increased, with upregulation of Il17a and Rankl genes in the gingival tissues. Treatment with anti-mouse IL-17A (100 µg/mice) significantly attenuated alveolar bone loss. Mice with ligature-induced periodontitis treated with IPI-549 (3 mg/kg) or PI3Kγ-/- mice showed reduced alveolar bone loss and downregulation of Il17a and Rankl gene expression in the gingival tissues. Consistent with this, the bioinformatics analysis showed upregulation of IL17F, IL17A, IL17D, and STAT3 genes, as well as greater activation of IL-17 and PI3KCI pathways (upregulation of PIK3CG gene) in the gingival tissue of patients with periodontitis. CONCLUSION: PI3Kγ plays an important role in modulating IL-17A expression and alveolar bone loss in vivo and can be considered a promising pathway for the management of periodontal disease and the development of new therapies.


Subject(s)
Alveolar Bone Loss , Periodontitis , Animals , Mice , Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/genetics , Interleukin-17/genetics , Interleukin-17/metabolism , Periodontitis/drug therapy , Periodontitis/genetics , Gingiva/metabolism , Ligation , Disease Models, Animal
3.
Arch Toxicol ; 97(9): 2329-2342, 2023 09.
Article in English | MEDLINE | ID: mdl-37394624

ABSTRACT

Cell culture and invertebrate animal models reflect a significant evolution in scientific research by providing reliable evidence on the physiopathology of diseases, screening for new drugs, and toxicological tests while reducing the need for mammals. In this review, we discuss the progress and promise of alternative animal and non-animal methods in biomedical research, with a special focus on drug toxicity.


Subject(s)
Biomedical Research , Animals , Models, Animal , Mammals
4.
Inflammopharmacology ; 30(3): 981-990, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35303234

ABSTRACT

Epoxyeicosatrienoic acids (EETs) are endogenous molecules that exerts effective antinociceptive and resolutive actions. However, because of their rapid metabolism by the soluble epoxide hydrolase (sEH), EETs are unable to remain bioavailable. Therefore, the aim of this study was to investigate whether local sEH inhibition could prevent inflammatory hyperalgesia in the temporomandibular joint (TMJ) of rats. For that, rats were pre-treated with an intra-TMJ injection of TPPU, followed by the noxious stimulus (1.5% of formalin intra-articular) to evaluate nociceptive behavior. Histological analysis was conducted to explore the inflammatory exudate and mast cell degranulation. Periarticular tissue over the TMJ was used to measure inflammatory lipids and cytokines/chemokine by Enzyme-Linked Immunosorbent Assay (ELISA). We demonstrated that peripheral pretreatment with TPPU prevents formalin-induced inflammatory hyperalgesia in the TMJ, and this effect is strictly local. Moreover, TPPU mitigates the leukocyte exudate in the TMJ, as well as inflammatory lipids mediators. Mast cell number and degranulation were abrogated by TPPU, and the inflammatory cytokine levels were decreased by TPPU. On the other hand, TPPU up-regulated the release of interleukin 10 (IL-10), an anti-inflammatory cytokine. We provide evidence that locally sEH by intra-TMJ injection of TPPU produces an antinociceptive and anti-inflammatory effect on rats' TMJ.


Subject(s)
Epoxide Hydrolases , Hyperalgesia , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Cytokines/metabolism , Epoxide Hydrolases/metabolism , Formaldehyde/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Lipids , Phenylurea Compounds/toxicity , Piperidines/pharmacology , Rats , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology
5.
J Prosthodont ; 30(2): 142-149, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32783328

ABSTRACT

PURPOSE: To investigate the effect of experimental traumatic occlusion (ETO) induced by metal crowns on alveolar bone loss. MATERIALS AND METHODS: Metal crowns were custom-made for the lower first molars with occlusal discrepancy of 0.4 and 0.7 mm from the maximum intercuspation. Thirty-six animals were randomly divided into three groups (n = 12 animals per group): 0.4-mm hyperocclusion group, 0.7-mm hyperocclusion group and the sham group (no metal crown). Twenty-eight days after crown cementation, the animals were euthanized and gingival tissue was collected to assess cytokine levels of IL-17, IL-6, and TNF-α using enzyme-linked immunosorbent assay (ELISA). Mandibles were stained with 1% methylene blue and alveolar bone levels were quantified. Western blotting was used to quantify the expression of receptor activator of nuclear factor κ B (RANK), and its ligand (RANKL), secreted osteoclastogenic factor of activated T cells (SOFAT) and TNF-α-converting enzyme (TACE). Also, mandibles were histologically processed and stained with hematoxylin and eosin, from which the presence of osteoclast-like cells, multinucleated cells containing ≥3 nuclei was counted at 100× magnification. The data were analyzed using one-way ANOVA and Tukey tests. RESULTS: Experimental occlusal trauma for 28 consecutive days significantly increased alveolar bone loss and multinucleated cell counts (p < 0.05). RANK, RANKL, SOFAT, TACE, IL-6, and TNF-α were significantly higher in gingival tissues of ETO groups (p < 0.05). IL-17 titers were unchanged among the groups (p > 0.05). CONCLUSION: Experimental traumatic occlusion activates and sustains bone resorption pathways in the periodontium inducing alveolar bone resorption. As the intensity of occlusal trauma increased, alternative osteoclastic pathways were activated, such as TACE and SOFAT.


Subject(s)
Alveolar Bone Loss , Cementation , Alveolar Bone Loss/etiology , Animals , Crowns , Osteoclasts , Periodontium
6.
J Pharmacol Exp Ther ; 370(3): 834-842, 2019 09.
Article in English | MEDLINE | ID: mdl-30872390

ABSTRACT

Coated microneedles have emerged as a promising drug delivery system for inflammatory pain treatment. We have previously shown that tramadol injection into the rat temporomandibular joint (TMJ) induces an antinociceptive and anti-inflammatory effect. In this study, microneedles coated with tramadol were investigated as a platform to treat TMJ pain. Male Wistar rats were administered tramadol using an intra-TMJ injection or with microneedles coated with tramadol, followed by 1.5% formalin nociceptive challenge administered 15 minutes later. The nociceptive behavior of rats was evaluated, and their periarticular tissues were removed after euthanasia for analysis. The duration of antinociceptive effect was determined by performing the formalin challenge at different time points extending up to 6 days post tramadol administration. Microneedles coated with tramadol produced an antinociceptive effect similar to injection of tramadol into the rat TMJ. Surprisingly, tramadol delivery using coated microneedles produced a more durable antinociceptive effect lasting as much as 2 days post tramadol delivery as compared with an antinociceptive effect lasting under 2 hours from intra-TMJ injection of tramadol. The proinflammatory cytokines tumor necrosis factor-α and interleukin-1ß (IL-1ß) were found to be reduced, whereas the anti-inflammatory cytokine IL-10 was found to be elevated in tramadol-treated groups. In conclusion, microneedles coated with tramadol can offer a therapeutic option for pain control of inflammatory disorders in the TMJ.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Hyperalgesia/drug therapy , Needles , Temporomandibular Joint Dysfunction Syndrome/drug therapy , Tramadol/administration & dosage , Tramadol/therapeutic use , Animals , Cytokines/blood , Drug Delivery Systems , Formaldehyde , Hyperalgesia/chemically induced , Hyperalgesia/psychology , Injections, Intra-Articular , Injections, Intralesional , Male , Rats , Rats, Wistar , Temporomandibular Joint , Temporomandibular Joint Dysfunction Syndrome/chemically induced , Temporomandibular Joint Dysfunction Syndrome/psychology
7.
Clin Oral Investig ; 23(4): 1905-1912, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30232624

ABSTRACT

OBJECTIVES: The goal of this study is to propose a standard protocol of experimental occlusal trauma to evaluate the inflammatory hyperalgesia induced by metallic crowns on orofacial tissues of rats. MATERIALS AND METHODS: Thirty animals were randomly divided into six groups (n = 5 per group). Detailed methodology on the manufacturing of metallic crowns is described. The inflammatory hyperalgesia induced by occlusal interference was evaluated by intra-articular injection of a low dose of 0.5% formalin (30 µl) or vehicle (saline) into temporomandibular joint, 21 or 28 days after metallic crown cementation. Posteriorly, pro-inflammatory cytokines were evaluated by enzyme-linked immunosorbent assay to assess the effect of occlusal interference on periodontium. RESULTS: The cementation of metallic crowns with dental anatomy on the lower molar of rats does not show signs of stress and lack of feeding. Metallic crown-induced occlusal trauma results in a temporomandibular joint inflammatory hyperalgesia (P < 0.05: ANOVA, Tukey's test). Otherwise, it was observed that occlusal trauma results in the increase of protein level of pro-inflammatory cytokines TNF-alpha and IL-1beta in the gingival tissues (P < 0.05). CONCLUSION: This study demonstrates in detail a methodology of occlusal trauma resulting from the cementation of metallic crowns in the lower molars of rats, mimicking occlusal interferences commonly evaluated in the dental clinic. This methodology makes new studies to better understand the mechanisms involved in the occlusal trauma of orofacial tissues possible. CLINICAL RELEVANCE: The standardization of an experimental occlusal interference model will allow us to understand the deleterious effect and mechanisms that affect the orofacial tissues.


Subject(s)
Crowns , Dental Occlusion, Traumatic , Inflammation , Periodontium/physiopathology , Temporomandibular Joint/physiopathology , Animals , Cytokines , Hyperalgesia , Male , Random Allocation , Rats , Rats, Wistar
9.
Article in English | MEDLINE | ID: mdl-37984607

ABSTRACT

Rheumatoid arthritis is a common systemic inflammatory autoimmune disease characterized by damage to joints, inflammation and pain. It is driven by an increase of inflammatory cytokines and lipids mediators such as prostaglandins. Epoxides of polyunsaturated fatty acids (PUFAs) are lipid chemical mediators in a group of regulatory compounds termed eicosanoids. These epoxy fatty acids (EpFA) have resolutive functions but are rapidly metabolized by the soluble epoxide hydrolase enzyme (sEH) into the corresponding diols. The pharmacological inhibition of sEH stabilizes EpFA from hydrolysis, improving their half-lives and biological effects. These anti-inflammatory EpFA, are analgesic in neuropathic and inflammatory pain conditions. Nonetheless, inhibition of sEH on arthritis and the resulting effects on eicosanoids profiles are little explored despite the physiological importance. In this study, we investigated the effect of sEH inhibition on collagen-induced arthritis (CIA) and its impact on the plasma eicosanoid profile. We measured the eicosanoid metabolites by LC-MS/MS-based lipidomic analysis. The treatment with a sEH inhibitor significantly modulated 11 out of 69 eicosanoids, including increased epoxides 12(13)-EpODE, 12(13)-EpOME, 13-oxo-ODE, 15-HEPE, 20-COOH-LTB4 and decreases several diols 15,6-DiHODE, 12,13-DiHOME, 14,15-DiHETrE, 5,6-DiHETrE and 16,17-DiHDPE. Overall the inhibition of sEH in the rheumatoid arthritis model enhanced epoxides generally considered anti-inflammatory or resolutive mediators and decreased several diols with inflammatory features. These findings support the hypothesis that inhibiting the sEH increases systemic EpFA levels, advancing the understanding of the impact of these lipid mediators as therapeutical targets.


Subject(s)
Arthritis, Rheumatoid , Epoxide Hydrolases , Humans , Chromatography, Liquid , Tandem Mass Spectrometry , Fatty Acids/metabolism , Pain , Eicosanoids , Arthritis, Rheumatoid/drug therapy , Anti-Inflammatory Agents , Epoxy Compounds/pharmacology
10.
Arch Oral Biol ; 155: 105778, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37572522

ABSTRACT

OBJECTIVE: Semaphorin 4D (Sema4D) is a coupling factor expressed on osteoclasts that may hinder osteoblast differentiation. Since the leukocyte platelet-rich fibrin (L-PRF) membrane promotes growth factor concentration, this study aims to quantify the amount of Sema4D in L-PRF membranes, and analyze the impact of Sema4D on osteoblast cell function in vitro. DESIGN: Enzyme-linked immunosorbent assay (ELISA) was used to quantify the levels of Sema4D in both L-PRF and whole blood (serum). To analyze the impairment of Sema4D on osteoblasts, MC3T3-E1 cells were induced to osteogenic differentiation and exposed to Sema4D ranging from 10 to 500 ng/ml concentrations. The following parameters were assayed: 1) cell viability by MTT assay after 24, 48, and 72 h; 2) matrix mineralization by Alizarin Red staining after 14 days, 3) Runt-related transcription factor 2 (RUNX-2), osteocalcin (OCN), osteonectin (ONC), bone sialoprotein (BSP) and alkaline phosphatase (ALP) gene expression by qPCR. For all data, the significance level was set at 5%. RESULTS: The amount of Sema4D in the whole blood (serum) was higher than in L-PRF. Osteoblasts exposed to Sema4D at all tested concentrations exhibited a decrease in matrix mineralization formation as well in RUNX-2, OCN, ONC, BSP, and ALP gene expression (p < 0.05). CONCLUSION: The presence of Sema4D, a molecule known for suppressing osteoblast activity, diminishes within L-PRF, enhancing its ability to facilitate bone regeneration.


Subject(s)
Platelet-Rich Fibrin , Semaphorins , Cell Differentiation/genetics , Leukocytes/metabolism , Osteoblasts , Osteocalcin/metabolism , Osteogenesis/genetics , Platelet-Rich Fibrin/metabolism , Semaphorins/pharmacology , Semaphorins/metabolism , Animals , Mice
11.
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
12.
Expert Rev Clin Pharmacol ; 15(3): 295-303, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35481412

ABSTRACT

INTRODUCTION: Pain is a multidimensional experience involving the biological, psychological, and social dimensions of each individual. Particularly, the biological aspects of pain conditions are a response of the neuroimmunology system and the control of painful conditions is a worldwide challenge for researchers. Although years of investigation on pain experience and treatment exist, the high prevalence of chronic pain is still a fact. AREAS COVERED: Peroxisome proliferator-activated receptor-gamma (PPARγ) is a ligand-activated transcription factor belonging to the nuclear hormone receptor superfamily. It regulates several metabolic pathways, including lipid biosynthesis and glucose metabolism, when activated. However, PPARγ activation also has a critical immunomodulatory and neuroprotective effect. EXPERT OPINION: This review summarizes the evidence of synthetic or natural PPARγ ligands such as 15d-PGJ2, epoxyeicosatrienoic acids, thiazolidinediones, and specialized pro-resolving mediators, representing an interesting therapeutic tool for pain control.


Subject(s)
Immunomodulation , PPAR gamma , Humans , Immunomodulation/drug effects , Immunomodulation/physiology , Ligands , PPAR gamma/metabolism , Pain , Prostaglandin D2/metabolism , Thiazolidinediones/therapeutic use
13.
J Photochem Photobiol B ; 222: 112281, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34388640

ABSTRACT

Photobiomodulation (PBM) has been applied as a non-invasive technique for treating temporomandibular joint symptoms, especially on painful condition's relief, however the anti-inflammatory mechanism underlying the effect of PBM remains uncertain. This study aims to evaluate the mechanisms of action of PBM (808 nm) in a carrageenan-induced inflammation on temporomandibular joint (TMJ) of rats. In this study male Wistar rats were pre-treated with irradiation of a low-power diode laser for 15 s on TMJ (infra-red 808 nm, 100 mW, 50 J/cm2 and 1.5 J) 15 min prior an injection in the temporomandibular joint of carrageenan (100 µg/TMJ). 1 h after the TMJ treatments, the rats were terminally anesthetized for joint cavity wash and periarticular tissues collect. Samples analysis demonstrated that PBM inhibit leukocytes chemotaxis in the TMJ and significantly reduces amounts of TNF-α, IL-1ß and CINC-1. In addition, Western blotting analysis demonstrated that PBM significantly decreased the protein levels of P2X3 and P2X7 receptors in the periarticular tissues. On the other hand, PBM was able to increase protein level of IL-10 (anti-inflammatory cytokine). In summary, it is possible to suggest that PBM inhibit inflammatory chemotaxis, modulation the balance of the pro- and anti-inflammatory characteristics of inflammatory cells.


Subject(s)
Inflammation/therapy , Lasers, Semiconductor/therapeutic use , Low-Level Light Therapy , Temporomandibular Joint/radiation effects , Animals , Carrageenan/toxicity , Cell Movement/radiation effects , Down-Regulation/radiation effects , Enzyme-Linked Immunospot Assay , Inflammation/chemically induced , Interleukin-10/analysis , Leukocytes/cytology , Leukocytes/metabolism , Male , Rats , Rats, Wistar , Receptors, Purinergic P2X3/metabolism , Receptors, Purinergic P2X7/metabolism , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Tumor Necrosis Factor-alpha/analysis
14.
Int Immunopharmacol ; 97: 107680, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33932698

ABSTRACT

Peripheral tramadol's delivery in the temporomandibular joint (TMJ) leads to significant analgesic outcomes and inflammatory process's resolvent actions. Mechanistically, these properties are apart from the opioid system. Nevertheless, the molecular mechanisms behind these effects are still unclear. Therefore, the present study investigated the hypothesis that adenosine A1 receptors are involved in the tramadol-induced analgesic and anti-inflammatory effects in the TMJ. Animals were pretreated with an intra-TMJ injection of DPCPX (antagonist of A1 receptor) or tramadol and subsequent nociceptive challenge with an intra-TMJ injection of 1.5% formalin. For over 45 min, the nociceptive behavior was quantitated, and by the end of this assessment, the animals were euthanized, and the periarticular tissue was collected. Lastly, an in vitro assay of BMDM (Bone Marrow-Derived Macrophages) was performed to investigate tramadol activity in macrophages. The intra-TMJ injection of tramadol ameliorates formalin-induced hypernociception along with inhibiting leukocyte migration. The tramadol's peripheral anti-inflammatory effect was mediated by the adenosine A1 receptor and was associated with increased protein expression of α2a-adrenoceptor in the periarticular tissues (p < 0.05: ANOVA, Tukey's test). Also, tramadol inhibits formalin-induced leukocyte migration and protein expression of P2X7 receptors in the periarticular tissue (p < 0.05); however, DPCPX did not alter this effect (p > 0.05). Moreover, DPCPX significantly reduced the protein expression of the M2 macrophage marker, MRC1. In BMDM, tramadol significantly reduces inflammatory cytokines release, and DPCPX abrogated this effect (p < 0.05). We identify tramadol's peripheral effect is mediated by adenosine A1 receptor, possibly expressed in macrophages in the TMJ tissue. We also determined an important discovery related to the activation of A1R/α2a receptors in the tramadol action.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Arthralgia/drug therapy , Receptor, Adenosine A1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Tramadol/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Arthralgia/chemically induced , Arthralgia/immunology , Arthralgia/pathology , Disease Models, Animal , Formaldehyde/administration & dosage , Formaldehyde/toxicity , Humans , Injections, Intra-Articular , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Nociception/drug effects , Rats , Temporomandibular Joint/drug effects , Temporomandibular Joint/immunology , Temporomandibular Joint/pathology , Xanthines/administration & dosage , Xanthines/toxicity
15.
Toxicon ; 187: 116-121, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32882256

ABSTRACT

Analgesic mechanism of Botulinum toxin type A (BoNT/A) involves retrograde axonal transport to central nervous system, where it may interact with sensory neurons. Though, some authors suggested that BoNT/A antinociceptive action may also be associated with the inhibition intracellular factors and neuromodulators expressed by immune cells, especially by microglia. Antigen-induced arthritis in the temporomandibular joint (TMJ) of rats is signal by P2X7 receptor/Cathepsin S (CatS)/Fractalkine (FKN) microglia-activated pathway. Thus, we aimed to evaluate the possible modulatory effect of an intra-TMJ injection of BoNT/A on the P2X7/CatS/FKN microglia-activated pathway in the trigeminal subnucleus caudalis of rats with antigen-induced arthritis of the TMJ. A model of antigen-induced arthritis was used on Wistar rats (n = 40) by systemic injections of an emulsion containing complete Freund's adjuvant and methylated bovine serum albumin (mBSA) diluted in PBS. The arthritic condition was stablished by an intra-TMJ injection of mBSA (10 µg/TMJ/week) for 3 weeks. Then, animals were treated with an intra-TMJ injection of BoNT/A (onabotulinumtoxinA, Allergan®; 7U/kg) or vehicle saline. Animals were euthanized 24 h, 7 or 14 days after BoNT/A treatment and their trigeminal nucleus caudalis was harvested to evaluate the protein level of microglial purinergic P2X7 receptor and CX3 chemokine receptor 1 (CX3CR1) by Western blot, and to measure the protein level of microglial modulators CatS, FKN, and the pro-inflammatory cytokines tumor necrosis alfa (TNF-α) and interleukin 1ß (IL-1ß) by enzyme-linked immunosorbent assay (ELISA). The antigen-induced arthritis in the TMJ significantly increased the protein levels of P2X7, CatS, FKN, TNF-α and IL-1ß in the trigeminal subnucleus caudalis (P < 0.05). The intra-TMJ injection of BoNT/A reduced the protein levels of P2X7 in all time points tested. Additionally, BoNT/A significantly reduced the protein levels of CatS, FKN, and TNF-α 14 days after treatment. However, IL-1ß was significantly reduced just 24 h after the BoNT/A intra-TMJ treatment. Based on our results, we can suggest that the intra-TMJ injection of BoNT/A may promote a central effect by reducing the P2X7/CatS/FKN microglia-activated pathway in the trigeminal subnucleus caudalis.


Subject(s)
Botulinum Toxins, Type A/therapeutic use , Temporomandibular Joint , Animals , Arthritis, Experimental/drug therapy , Chemokine CX3CL1/metabolism , Freund's Adjuvant , Injections, Intra-Articular , Male , Microglia/metabolism , Rats , Rats, Wistar
16.
Int Immunopharmacol ; 87: 106841, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32736189

ABSTRACT

Rheumatoid arthritis (RA) is characterized by chronic inflammation of the synovial tissue, joint dysfunction, and damage. Epoxyeicosatrienoic acids (EETs) are endogenous anti-inflammatory compounds, which are quickly converted by the soluble epoxide hydrolase (sEH) enzyme into a less active form with decreased biological effects. The inhibition of the sEH enzyme has been used as a strategy to lower nociception and inflammation. The goal of this study was to investigate whether the peripheral treatment with the sEH enzyme inhibitor 1- trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) could prevent the hypernociception and inflammation in the albumin-induced arthritis model in rats' temporomandibular joint (TMJ). After the induction of experimental arthritis, animals were assessed for nociceptive behavior test, leukocyte infiltration counts and histologic analysis, ELISA to quantify several cytokines and Western blotting. The peripheral pretreatment with TPPU inhibited the arthritis-induced TMJ hypernociception and leukocyte migration. Moreover, the local concentrations of proinflammatory cytokines were diminished by TPPU, while the anti-inflammatory cytokine interleukin-10 was up-regulated in the TMJ tissue. Finally, TPPU significantly decreased protein expression of iNOS, while did not alter the expression of MRC1. This study provides evidence that the peripheral administration of TPPU reduces hypernociception and inflammation in TMJ experimental arthritis.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Epoxide Hydrolases/antagonists & inhibitors , Phenylurea Compounds/therapeutic use , Piperidines/therapeutic use , Temporomandibular Joint/drug effects , Albumins , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cytokines/immunology , Male , Nitric Oxide Synthase Type II/immunology , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Rats, Wistar , Temporomandibular Joint/immunology , Temporomandibular Joint/pathology
17.
Int Immunopharmacol ; 84: 106565, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32388213

ABSTRACT

Natural or synthetic ligands for peroxisome proliferator-activated receptor gamma (PPAR-γ) represent an interesting tool for pharmacological interventions to treat inflammatory conditions. In particular, PPAR-γ activation prevents pain and inflammation in the temporomandibular joint (TMJ) by decreasing cytokine release and stimulating the synthesis of endogenous opioids. The goal of this study was to clarify whether PPAR-γ activation induces macrophage polarization, inhibiting inflammatory cytokine release and leukocyte recruitment. In addition, we investigated the involvement of heme oxygenase 1 (HO-1) in downstream events after PPAR-γ activation. Our results demonstrate that PPAR-γ activation ablates cytokine release by Bone Marrow-Derived Macrophages (BMDM) in vitro. 15d-PGJ2 induces the PPAR-γ heterodimer activation from rat macrophages, with macrophage polarization from M1-like cells toward M2-like cells. This response is mediated through HO-1. PPAR-γ activation diminished neutrophil migration induced by carrageenan, which was also HO-1 dependent. Ca2+/calmodulin expression did not change after PPAR-γ activation indicating that is not required for the activation of the intracellular L-arginine/NO/cGMP/K+ATP channel pathway. In summary, the anti-inflammatory actions induced by PPAR-γ activation involve macrophage polarization. HO-1 expression is increased and HO-1 activity is required for the suppression of neutrophil migration.


Subject(s)
Heme Oxygenase-1/immunology , Macrophages/immunology , Neutrophils/physiology , PPAR gamma/immunology , Anilides/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/immunology , Carrageenan/pharmacology , Cell Movement/drug effects , Cells, Cultured , Cytokines/immunology , Humans , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Nitric Oxide/immunology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Rats, Wistar , Temporomandibular Joint/drug effects , Temporomandibular Joint/immunology
18.
Front Immunol ; 11: 1442, 2020.
Article in English | MEDLINE | ID: mdl-32849501

ABSTRACT

Rheumatoid arthritis (RA) has an inflammatory milieu in the synovial compartment, which is regulated by a complex cytokine and chemokine network that induces continuously degenerative and inflammatory reactions. The secreted osteoclastogenic factor of activated T cells (SOFAT) is a unique cytokine and represents an alternative pathway for osteoclast activation. In this study, we examined whether SOFAT is able to induce joint pain and investigated the presence of SOFAT in a Collagen-induced Arthritis (CIA) model and in human subjects. Here, we found that an intra-articular stimulation with SOFAT (1, 10, 100, or 1,000 ng/10 µl) in the knee joint significantly decreases the mechanical threshold in the hind paw of mice (p < 0.05). Moreover, after a second injection of SOFAT, the mechanical threshold decrease was sustained for up to 8 days (p < 0.05). In the CIA model, the immunohistochemical assay of knee joint showed positivity stained for SOFAT, and the mRNA and protein expression of SOFAT were significantly higher in the affected-group (p < 0.05). Besides, the mRNA of RANKL, IL-1ß, IL-6, and IL-15 were significantly higher in the affected-group (p < 0.05). Finally, SOFAT was detected in the synovial fluid of RA patients, but not in OA patients (p < 0.05). In conclusion, SOFAT is up regulated in inflammatory milieu such as RA but not in non-inflammatory OA. SOFAT may be a novel molecule in the complex inflammatory phenotype of RA.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Carbon-Oxygen Lyases/metabolism , Cytokines/metabolism , Joints/physiology , Pain/metabolism , Animals , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred DBA , Osteoclasts/physiology , Osteogenesis , Signal Transduction , Up-Regulation
19.
Curr Drug Deliv ; 17(7): 588-598, 2020.
Article in English | MEDLINE | ID: mdl-32394838

ABSTRACT

BACKGROUND: Antiproliferative and cytotoxic effects of lidocaine have been reported in tumor cells. However, the use of these drugs is restricted due to their short action with rapid dispersion from the injected site. The complexation of local anesthetics in 2-hydroxypropyl-ß-cyclodextrin (HP-ß-CD) is able to improve pharmacological features. OBJECTIVE: This study evaluated the antitumor effects of lidocaine and the complex HP-ß-CD-lidocaine (HP-ß-CD-lido). METHODS: In vitro;, human adenocarcinoma (HeLa) and keratinocytes (HaCaT) were exposed to lidocaine formulations and cell viability, proliferation and apoptosis induction were measured. In vivo;, Walker 256 carcinoma cells were subcutaneously injected into the plantar region of the rat right hind paw. The animals were treated with a local application of 5% lidocaine or 5% HP-ß-CD-lido. Doxorubicin (3 mg/Kg/day, intraperitoneal) was used as a positive control. Edema sizes were measured daily and the release of cytokines (TNF-α, IL-1α and CXCL-1) and prostaglandin E2 was evaluated. Histological analysis was also performed. RESULTS: HaCaT IG50 values were 846 µM and 2253 µM for lido and HP-ß-CD-lido, respectively. In HeLa cells, the IG50 was 1765 µM for lido and 2044 µM for HP-ß-CD-lido. Lidocaine formulations significantly reduced the paw edema on day 6 after Walker 256 cells inoculation. However, there were no differences in the release of inflammatory mediators in comparison to the control group. CONCLUSION: Lidocaine formulations were able to reduce the edema in vivo;, without affecting the tumor- induced inflammatory response. The antiproliferative effects of lidocaine formulations may have contributed to tumor reduction.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/chemistry , Drug Carriers/chemistry , Edema/drug therapy , Lidocaine/administration & dosage , Neoplasms/drug therapy , Animals , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Chemokine CXCL1/metabolism , Disease Models, Animal , Drug Compounding/methods , Drug Screening Assays, Antitumor , Edema/diagnosis , Edema/immunology , Edema/pathology , HaCaT Cells , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Interleukin-1alpha/metabolism , Lidocaine/pharmacokinetics , Male , Neoplasms/complications , Neoplasms/immunology , Neoplasms/pathology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Neuroscience ; 391: 120-130, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30248434

ABSTRACT

Temporomandibular joint (TMJ) is frequently involved with rheumatoid arthritis with a high prevalence that could result in a chronic pain state. Once the disease is established in the joint, the antigen-specific immune reaction initiates a neuro-immune cascade of events that causes sensitization of the central nervous system. This study establishes animal experimental models that evaluate the chronicity of albumin-induced arthritis hypernociception in the TMJ. Antigen-induced arthritis was generated in rats with methylated bovine serum albumin (mBSA) diluted in complete Freund's. Intra-articular injection of mBSA (10 µg/TMJ/week) during 3 weeks resulted in a persistent inflammatory hypernociception which was characterized by an inflammatory episode characterized by the increased of lymphocytes, macrophages and pro-inflammatory interleukins IL-12 and IL-18. The persistent model of inflammatory hypernociception induced by arthritis in the TMJ elicited protein levels of P2X7 receptors, cathepsin S and fractalkine in the trigeminal subnucleus caudalis. Overall, the results of the present work suggest that a persistent inflammatory hypernociception of albumin-induced arthritis in the TMJ leads to the activation of the central nervous system signaling by P2X7/cathepsin S/fractalkine pathway.


Subject(s)
Arthritis/metabolism , Cathepsins/metabolism , Chemokine CX3CL1/metabolism , Nociception , Receptors, Purinergic P2X7/metabolism , Temporomandibular Joint Disorders/metabolism , Trigeminal Nuclei/metabolism , Animals , Arthritis/complications , Arthritis/immunology , Arthritis, Experimental/chemically induced , Disease Models, Animal , Male , Rats, Wistar , Serum Albumin, Bovine/administration & dosage , Signal Transduction , Temporomandibular Joint Disorders/complications , Temporomandibular Joint Disorders/immunology , Trigeminal Nuclei/immunology
SELECTION OF CITATIONS
SEARCH DETAIL