Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 16(3)2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36986443

ABSTRACT

Kaurenoic acid (KA) is a diterpene extracted from Sphagneticola trilobata (L.) Pruski. KA presents analgesic properties. However, the analgesic activity and mechanisms of action of KA in neuropathic pain have not been investigated so far; thus, we addressed these points in the present study. A mouse model of neuropathic pain was induced by chronic constriction injury (CCI) of the sciatic nerve. Acute (at the 7th-day post-CCI surgery) and prolonged (from 7-14th days post-CCI surgery) KA post-treatment inhibited CCI-induced mechanical hyperalgesia at all evaluated time points, as per the electronic version of von Frey filaments. The underlying mechanism of KA was dependent on activating the NO/cGMP/PKG/ATP-sensitive potassium channel signaling pathway since L-NAME, ODQ, KT5823, and glibenclamide abolished KA analgesia. KA reduced the activation of primary afferent sensory neurons, as observed by a reduction in CCI-triggered colocalization of pNF-κB and NeuN in DRG neurons. KA treatment also increased the expression of neuronal nitric oxide synthase (nNOS) at the protein level as well as the intracellular levels of NO in DRG neurons. Therefore, our results provide evidence that KA inhibits CCI neuropathic pain by activating a neuronal analgesic mechanism that depends on nNOS production of NO to silence the nociceptive signaling that generates analgesia.

2.
Molecules ; 28(2)2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36677929

ABSTRACT

Arthroplasty is an orthopedic surgical procedure that replaces a dysfunctional joint by an orthopedic prosthesis, thereby restoring joint function. Upon the use of the joint prosthesis, a wearing process begins, which releases components such as titanium dioxide (TiO2) that trigger an immune response in the periprosthetic tissue, leading to arthritis, arthroplasty failure, and the need for revision. Flavonoids belong to a class of natural polyphenolic compounds that possess antioxidant and anti-inflammatory activities. Hesperidin methyl chalcone's (HMC) analgesic, anti-inflammatory, and antioxidant effects have been investigated in some models, but its activity against the arthritis caused by prosthesis-wearing molecules, such as TiO2, has not been investigated. Mice were treated with HMC (100 mg/kg, intraperitoneally (i.p.)) 24 h after intra-articular injection of 3 mg/joint of TiO2, which was used to induce chronic arthritis. HMC inhibited mechanical hyperalgesia, thermal hyperalgesia, joint edema, leukocyte recruitment, and oxidative stress in the knee joint (alterations in gp91phox, GSH, superoxide anion, and lipid peroxidation) and in recruited leukocytes (total reactive oxygen species and GSH); reduced patellar proteoglycan degradation; and decreased pro-inflammatory cytokine production. HMC also reduced the activation of nociceptor-sensory TRPV1+ and TRPA1+ neurons. These effects occurred without renal, hepatic, or gastric damage. Thus, HMC reduces arthritis triggered by TiO2, a component released upon wearing of prosthesis.


Subject(s)
Arthritis , Chalcones , Hesperidin , Mice , Animals , Nociceptors/metabolism , Chalcones/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Arthritis/drug therapy , Oxidative Stress , Antioxidants/pharmacology , Anti-Inflammatory Agents/pharmacology , Hyperalgesia/drug therapy , Cytokines/metabolism
3.
Nat Prod Res ; 36(23): 6081-6084, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35227131

ABSTRACT

Pimenta pseudocaryophyllus (Gomes) Landrum is a Brazilian native plant. The mechanisms by which it promotes analgesia are unknown. We demonstrated the analgesic effect of P. pseudocaryophyllus dried extract (3 mg/kg; i.p.) in the following models of inflammatory pain (maximal inhibition): phenyl-p-benzoquinone (89%), formalin (72% - 1st phase and 96% - 2nd phase for flinches, and 50% - 1st phase and 71% - 2nd phase for licking behavior), complete Freund's adjuvant (95% - flinches and 33% - licking behavior), and carrageenin (56% - mechanical and 85% - thermal hyperalgesia) without motor impairment. Its analgesic effect depends on inhibiting neutrophil recruitment (95% - histopathology, 83% - myeloperoxidase activity, and 80% - LysM-eGFP mice), oxidative stress (86% - GSH and 98% - superoxide anion), and cytokine production (35% - IL-33, 80% - TNF-α, and 95% - IL-1ß). The present study advances in understanding the analgesic mechanisms of P. pseudocaryophyllus.


Subject(s)
Pimenta , Mice , Animals , Neutrophil Infiltration , Pain/drug therapy , Oxidative Stress , Analgesics/pharmacology , Analgesics/therapeutic use , Hyperalgesia , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Inflammation/drug therapy , Cytokines/metabolism
4.
J Ethnopharmacol ; 273: 113980, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-33652112

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sphagneticola trilobata (L.) Pruski is a plant species belonging to the Asteraceae family. Kaurenoid acid (KA) is a diterpene metabolite and one of the active ingredients of Sphagneticola trilobata (L.) Pruski. Extracts containing KA are used in traditional medicine to treat pain, inflammation, and infection. AIM: The goal of the present study was to investigate the in vivo effects of KA (1-10 mg/kg, per oral gavage) upon LPS inoculation in mice by intraperitoneal (i.p.) or intraplantar (i.pl.; subcutaneous plantar injection) routes at the dose of 200 ng (200 µL or 25 µL, respectively). METHODS: In LPS paw inflammation, mechanical and thermal hyperalgesia MPO activity and oxidative imbalance (TBARS, GSH, ABTS and FRAP assays) were evaluated. In LPS peritonitis we evaluated leukocyte migration, cytokine production, oxidative stress, and NF-κB activation. RESULTS: KA inhibited LPS-induced mechanical and thermal hyperalgesia, MPO activity and modulated redox status in the mice paw. Pre- and post-treatment with KA inhibited migration of neutrophils and monocytes in LPS peritonitis. KA inhibited the pro-inflammatory/hyperalgesic cytokine (e.g., TNF-α, IL-1ß and IL-33) production while enhanced anti-inflammatory/analgesic cytokine IL-10 in peritoneal cavity. In agreement with the effect of KA over pro-inflammatory cytokines it inhibited oxidative stress (total ROS, superoxide production and superoxide positive cells) and NF-κB activation during peritonitis. CONCLUSION: KA efficiently dampens LPS-induced peritonitis and hyperalgesia in vivo, suggesting it as a suitable candidate to control excessive inflammation and pain during gram-negative bacterial infections and bringing mechanistic explanation to the ethnopharmacological application of Sphagneticola trilobata (L.) Pruski in inflammation and infection.


Subject(s)
Analgesics/therapeutic use , Asteraceae/chemistry , Diterpenes/therapeutic use , Lipopolysaccharides/toxicity , Peritonitis/chemically induced , Analgesics/chemistry , Animals , Diterpenes/chemistry , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Lipid Peroxidation , Male , Mice , Molecular Structure , NF-kappa B/genetics , NF-kappa B/metabolism , Pain/drug therapy , Peritonitis/drug therapy , Peroxidase/metabolism
5.
Inflammopharmacology ; 27(6): 1285-1296, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30945072

ABSTRACT

Rutin is a glycone form of the flavonol quercetin and it reduces inflammatory pain in animal models. Therapy with granulocyte colony-stimulating factor (G-CSF) is known by the pain caused as its main side effect. The effect of rutin and its mechanisms of action were evaluated in a model of hyperalgesia induced by G-CSF in mice. The mechanical hyperalgesia induced by G-CSF was reduced by treatment with rutin in a dose-dependent manner. Treatment with both rutin + morphine or rutin + indomethacin, at doses that are ineffectual per se, significantly reduced the pain caused by G-CSF. The nitric oxide (NO)-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG)-ATP-sensitive potassium channel (KATP) signaling pathway activation is one of the analgesic mechanisms of rutin. Rutin also reduced the pro-hyperalgesic and increased anti-hyperalgesic cytokine production induced by G-CSF. Furthermore, rutin inhibited the activation of the nuclear factor kappa-light-chain enhancer of activated B cells (NFκB), which might explain the inhibition of the cytokine production. Treatment with rutin upregulated the decreased mRNA expression of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) combined with enhancement of the mRNA expression of the Nrf2 downstream target heme oxygenase (HO-1). Intraperitoneal (i.p.) treatment with rutin did not alter the mobilization of neutrophils induced by G-CSF. The analgesia by rutin can be explained by: NO-cGMP-PKG-KATP channel signaling activation, inhibition of NFκB and triggering the Nrf2/HO-1 pathway. The present study demonstrates rutin as a promising pharmacological approach to treat the pain induced by G-CSF without impairing its primary therapeutic benefit of mobilizing hematopoietic progenitor cells into the blood.


Subject(s)
Analgesics/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Pain/drug therapy , Rutin/pharmacology , Animals , Cyclic GMP/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Cytokines/biosynthesis , Heme Oxygenase-1/physiology , Hyperalgesia/drug therapy , KATP Channels/physiology , Male , Mice , NF-E2-Related Factor 2/physiology , NF-kappa B/antagonists & inhibitors , Neutrophils/drug effects , Nitric Oxide/physiology , Pain/chemically induced , Signal Transduction/drug effects
6.
Inflamm Res ; 67(11-12): 997-1012, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30370484

ABSTRACT

OBJECTIVE: To evaluate the effect and mechanisms of naringenin in TiO2-induced chronic arthritis in mice, a model resembling prosthesis and implant inflammation. TREATMENT: Flavonoids are antioxidant and anti-inflammatory molecules with important anti-inflammatory effect. Mice were daily treated with the flavonoid naringenin (16.7-150 mg/kg, orally) for 30 days starting 24 h after intra-articular knee injection of 3 mg of TiO2. METHODS: TiO2-induced arthritis resembles cases of aseptic inflammation induced by prosthesis and/or implants. Mice were stimulated with 3 mg of TiO2 and after 24 h mice started to be treated with naringenin. The disease phenotype, treatment toxicity, histopathological damage, oxidative stress, cytokine expression and NFκB were evaluated after 30 days of treatment. RESULTS: Naringenin inhibited TiO2-induced mechanical hyperalgesia (96%), edema (77%) and leukocyte recruitment (74%) without inducing toxicity. Naringenin inhibited histopathological index (HE, 49%), cartilage damage (Toluidine blue tibial staining 49%, and proteoglycan 98%), and bone resorption (TRAP-stained 73%). These effects were accompanied by inhibition of oxidative stress (gp91phox 93%, NBT 83%, and TBARS 41%) cytokine mRNA expression (IL-33 82%, TNFα 76%, pro-IL-1ß 100%, and IL-6 61%), and NFκB activation (100%). CONCLUSION: Naringenin ameliorates TiO2-induced chronic arthritis inducing analgesic and anti-inflammatory responses with improvement in the histopathological index, cartilage damage, and bone resorption.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Arthritis/drug therapy , Flavanones/therapeutic use , Hyperalgesia/drug therapy , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Arthritis/chemically induced , Arthritis/pathology , Chronic Disease , Cytokines/genetics , Flavanones/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Joints/drug effects , Joints/metabolism , Joints/pathology , Male , Mice , NF-kappa B/metabolism , Titanium
7.
J Pharm Pharmacol ; 70(6): 768-777, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29532470

ABSTRACT

OBJECTIVES: To evaluate the analgesic effect of Glucantime (antimoniate N-methylglucamine) in Leishmania amazonensis infection and complete Freund's adjuvant (CFA), chronic paw inflammation model, in BALB/c mice. METHODS: Two models of chronic inflammatory pain in BALB/c mice paw were used: infection with L. amazonensis and CFA stimulation. Both animals models received daily treatment with Glucantime (10 mg/kg, i.p.) and during the treatment was measured the mechanical hyperalgesia with electronic version of von Frey filaments. After the treatment, the paw skin sample was collected for analysis of myeloperoxidase (MPO) and N-acetyl-ß-glucosaminidase (NAG) activity, and IL-1ß, TNF-α, IL-6, IFN-γ and IL-10 cytokines production by ELISA. KEY FINDINGS: Leishmania amazonensis-induced chronic inflammation with significant increase in mechanical hyperalgesia, MPO and NAG activity, and IL-1ß, TNF-α and IL-6 production in the paw skin. Glucantime (10 mg/kg, i.p.) inhibited L. amazonensis-induced mechanical hyperalgesia and IL-1ß and IL-6 cytokines productions. In chronic inflammatory model induced by CFA, Glucantime treatment during 7 days inhibited CFA-induced mechanical hyperalgesia, MPO and NAG activity, and IL-1ß, TNF-α, IL-6 and IFN-γ production as well as increased IL-10 production. CONCLUSIONS: Our data demonstrated that Glucantime reduced the chronic inflammatory pain induced by L. amazonensis and CFA stimuli by inhibiting the hyperalgesic cytokines production.


Subject(s)
Chronic Pain/drug therapy , Inflammation/drug therapy , Leishmaniasis, Cutaneous/drug therapy , Meglumine/therapeutic use , Organometallic Compounds/therapeutic use , Acetylglucosaminidase/metabolism , Animals , Chronic Pain/complications , Cytokines/metabolism , Freund's Adjuvant , Inflammation/chemically induced , Inflammation/complications , Male , Meglumine Antimoniate , Mice , Peroxidase/metabolism , Skin/metabolism
8.
J Nutr Biochem ; 53: 81-95, 2018 03.
Article in English | MEDLINE | ID: mdl-29197723

ABSTRACT

Titanium dioxide (TiO2) is a common component of orthopedic prosthesis. However, prosthesis wear releases TiO2, which induces inflammation and osteolysis in peri-prosthetic tissues. Quercetin is a flavonoid widely present in human diet, which presents biological activities such as antinociceptive, anti-inflammatory and antioxidant effects. Therefore, the effect of intraperitoneal treatment with quercetin in TiO2-induced arthritis model was evaluated. In the first set of experiments, mice received injection of TiO2 (0.1-3 mg/knee joint) and articular mechanical hyperalgesia, edema and histopathology analysis were performed in a 30 days protocol. The dose of 3 mg of TiO2 showed the most harmful effect, and was chosen to the following experiments. Subsequently, mice received 3 mg of TiO2 followed by post-treatment with quercetin during 30 days. Quercetin (10-100 mg/kg) inhibited in a dose-dependent manner TiO2-induced knee joint mechanical hyperalgesia, edema and leukocyte recruitment and did not induce damage in major organs such as liver, kidney and stomach. The dose of 30 mg/kg was chosen for the subsequent analysis, and reduced histopathological changes such as leukocyte infiltration, vascular proliferation and synovial hyperplasia (pannus formation) on day 30 after TiO2 challenge. The protective analgesic and anti-inflammatory mechanisms of quercetin included the inhibition of TiO2-induced neutrophil and macrophage recruitment, proteoglycan degradation, oxidative stress, cytokine production (TNF-α, IL-1ß, IL-6, and IL-10), COX-2 mRNA expression, and bone resorption as well as activation of Nrf2/HO-1 signaling pathway. These results demonstrate the potential therapeutic applicability of the dietary flavonoid quercetin to reduce pain and inflammatory damages associated with prosthesis wear process-induced arthritis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/drug therapy , Knee Joint/drug effects , Quercetin/pharmacology , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/adverse effects , Analgesics, Non-Narcotic/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Arthritis, Experimental/chemically induced , Bone Resorption/chemically induced , Bone Resorption/drug therapy , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Injections, Intra-Articular , Injections, Intraperitoneal , Kidney/cytology , Kidney/drug effects , Knee Joint/pathology , Liver/cytology , Liver/drug effects , Male , Mice , Oxidative Stress/drug effects , Quercetin/administration & dosage , Quercetin/adverse effects , Titanium/administration & dosage , Titanium/toxicity
9.
Inflammopharmacology ; 2017 May 15.
Article in English | MEDLINE | ID: mdl-28508104

ABSTRACT

We investigated the anti-inflammatory and analgesic effects of quercetin in monosodium urate crystals (MSU)-induced gout arthritis, and the sensitivity of quercetin effects to naloxone, an opioid receptor antagonist. Mice were treated with quercetin, and mechanical hyperalgesia was assessed at 1-24 h after MSU injection. In vivo, leukocyte recruitment, cytokine levels, oxidative stress, NFκB activation, and gp91phox and inflammasome components (NLRP3, ASC, Pro-caspase-1, and Pro-IL-1ß) mRNA expression by qPCR were determined in the knee joints at 24 h after MSU injection. Inflammasome activation was determined, in vitro, in lipopolysaccharide-primed macrophages challenged with MSU. Quercetin inhibited MSU-induced mechanical hyperalgesia, leukocyte recruitment, TNFα and IL-1ß production, superoxide anion production, inflammasome activation, decrease of antioxidants levels, NFκB activation, and inflammasome components mRNA expression. Naloxone pre-treatment prevented all the inhibitory effects of quercetin over MSU-induced gout arthritis. These results demonstrate that quercetin exerts analgesic and anti-inflammatory effect in the MSU-induced arthritis in a naloxone-sensitive manner.

10.
Free Radic Biol Med ; 108: 487-499, 2017 07.
Article in English | MEDLINE | ID: mdl-28419865

ABSTRACT

Septic arthritis is a severe and rapidly debilitating disease associated with severe joint pain, inflammation and oxidative stress. Nitroxyl (HNO) has become a nitrogen oxide of significant interest due to its pharmacological endpoints that are potentially favorable for treating varied diseases. However, whether HNO also serves as a treatment to septic arthritis is currently unknown. The aim of this study was to investigate the effect of the HNO donor, Angeli's salt (AS), in the outcome of chronic Staphylococcus aureus (S. aureus)-induced septic arthritis in mice. Daily treatment with AS inhibited mechanical hyperalgesia and inflammation (edema, leukocyte migration, cytokines release and NF-κB activation, and oxidative stress) resulting in reduced disease severity (clinical course, histopathological changes, proteoglycan levels in the joints, and osteoclastogenesis). In addition, AS decreased the number of S. aureus colony forming unities in synovial tissue, enhanced the bactericidal effect of macrophages and inhibited the worsening of systemic inflammatory response (leukocyte counts in the lung and systemic proinflammatory cytokine concentration). Our results suggest for the first time the therapeutic potential of AS in a model of septic arthritis by mechanisms involving microbicidal effects, anti-inflammatory actions and reduction of disease severity.


Subject(s)
Antioxidants/therapeutic use , Arthritis, Infectious/drug therapy , Inflammation/drug therapy , Lung/immunology , Nitrites/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus/immunology , Animals , Hyperalgesia , Lung/drug effects , Lung/microbiology , Male , Mice , NF-kappa B/metabolism , Nitrogen Oxides/metabolism , Oxidative Stress , Signal Transduction
11.
J Nutr Biochem ; 33: 8-14, 2016 07.
Article in English | MEDLINE | ID: mdl-27260463

ABSTRACT

Lipopolysaccharide (LPS) is the major structural component of Gram-negative bacteria cell wall and a highly pro-inflammatory toxin. Naringenin is found in Citrus fruits and exhibits antioxidant and anti-inflammatory properties through inhibition of NF-κB activation but its effects in LPS-induced inflammatory pain and leukocyte recruitment were not investigated yet. We investigated the effects of naringenin in mechanical hyperalgesia, thermal hyperalgesia and leukocyte recruitment induced by intraplantar injection of LPS in mice. We found that naringenin reduced hyperalgesia to mechanical and thermal stimuli, myeloperoxidase (MPO, a neutrophil and macrophage marker) and N-acetyl-ß-D-glucosaminidase (NAG, a macrophage marker) activities, oxidative stress and cytokine (TNF-α, IL-1ß, IL-6, and IL-12) production in the paw skin. In the peritoneal cavity, naringenin reduced neutrophil and mononuclear cell recruitment, and abrogated MPO and NAG activity, cytokine and superoxide anion production, and lipid peroxidation. In vitro, pre-treatment with naringenin inhibited superoxide anion and cytokine (TNF-α, IL-1ß, IL-6, and IL-12) production by LPS-stimulated RAW 264.7 macrophages. Finally, we demonstrated that naringenin inhibited NF-κB activation in vitro and in vivo. Therefore, naringenin is a promising compound to treat LPS-induced inflammatory pain and leukocyte recruitment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dietary Supplements , Flavanones/therapeutic use , Hyperalgesia/prevention & control , Leukocytes, Mononuclear/immunology , Neutrophils/immunology , Transcription Factor RelA/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antioxidants/therapeutic use , Behavior, Animal , Biomarkers/metabolism , Flavanones/metabolism , Hot Temperature/adverse effects , Hyperalgesia/immunology , Hyperalgesia/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Oxidative Stress/drug effects , RAW 264.7 Cells , Skin/drug effects , Skin/immunology , Skin/metabolism , Stress, Mechanical , Transcription Factor RelA/metabolism
12.
Neuropharmacology ; 105: 508-519, 2016 06.
Article in English | MEDLINE | ID: mdl-26907804

ABSTRACT

Naringenin is a flavonoid widely consumed by humans that present anti-inflammatory activity and low toxicity. Recently, the analgesic effect of naringenin has been demonstrated in neuropathic pain models. Herein, we tested the analgesic effects of naringenin in several models of inflammatory pain. Mice received treatment with naringenin (16.7-150 mg/kg, per oral), or with the controls anti-inflammatory drugs indomethacin (5 mg/kg, intraperitoneal) or dipyrone (80 mg/kg, intraperitoneal) prior the inflammatory stimuli injection. For acute pain, we used acetic acid- and PBQ-induced visceral pain (abdominal writhings), and formalin-, capsaicin-, and CFA-induced paw flinching and licking. By using an electronic version of von Frey filaments, we also investigated the effects of naringenin in pain intensity to a mechanical stimulus (mechanical hyperalgesia) after carrageenan, capsaicin, CFA, or PGE2 intraplantar injection. Naringenin (50 mg/kg) reduced acute pain behaviors induced by all tested stimuli, including both phases of formalin test, suggesting a direct nociceptor modulatory effect of this compound besides its anti-inflammatory activity. Accordingly, naringenin also inhibited the increased sensitivity to mechanical stimulus induced by carrageenan, capsaicin, and PGE2. Daily treatment with naringenin during 7 days also reduced CFA-induced mechanical hyperalgesia without gastric or hepatic toxicity. The mechanisms of naringenin involve the inhibition of carrageenan-induced oxidative stress, hyperalgesic cytokines (IL-33, TNF-α, and IL-1ß) production and NF-κB activation in the paw skin. Naringenin also activated the analgesic NO-cyclic GMP-PKG-ATP sensitive K(+) channel signaling pathway to inhibit carrageenan-induced mechanical hyperalgesia and neutrophil recruitment. These results suggest that naringenin inhibits both inflammatory pain and neurogenic inflammation.


Subject(s)
Analgesics/pharmacology , Flavanones/pharmacology , Hyperalgesia/drug therapy , Visceral Pain/drug therapy , Animals , Cytokines/metabolism , Dipyrone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Hyperalgesia/metabolism , Indomethacin/pharmacology , Injections, Intraperitoneal , KATP Channels/metabolism , Liver/drug effects , Male , Mice , Motor Activity/drug effects , NF-kappa B/metabolism , Neutrophils/drug effects , Nociceptors/drug effects , Stomach/drug effects , Visceral Pain/metabolism
13.
PLoS One ; 11(2): e0149656, 2016.
Article in English | MEDLINE | ID: mdl-26895409

ABSTRACT

Pimaradienoic acid (PA; ent-pimara-8(14),15-dien-19-oic acid) is a pimarane diterpene found in plants such as Vigueira arenaria Baker (Asteraceae) in the Brazilian savannas. Although there is evidence on the analgesic and in vitro inhibition of inflammatory signaling pathways, and paw edema by PA, its anti-inflammatory effect deserves further investigation. Thus, the objective of present study was to investigate the anti-inflammatory effect of PA in carrageenan-induced peritoneal and paw inflammation in mice. Firstly, we assessed the effect of PA in carrageenan-induced leukocyte recruitment in the peritoneal cavity and paw edema and myeloperoxidase activity. Next, we investigated the mechanisms involved in the anti-inflammatory effect of PA. The effect of PA on carrageenan-induced oxidative stress in the paw skin and peritoneal cavity was assessed. We also tested the effect of PA on nitric oxide, superoxide anion, and inflammatory cytokine production in the peritoneal cavity. PA inhibited carrageenan-induced recruitment of total leukocytes and neutrophils to the peritoneal cavity in a dose-dependent manner. PA also inhibited carrageenan-induced paw edema and myeloperoxidase activity in the paw skin. The anti-inflammatory mechanism of PA depended on maintaining paw skin antioxidant activity as observed by the levels of reduced glutathione, ability to scavenge the ABTS cation and reduce iron as well as by the inhibition of superoxide anion and nitric oxide production in the peritoneal cavity. Furthermore, PA inhibited carrageenan-induced peritoneal production of inflammatory cytokines TNF-α and IL-1ß. PA presents prominent anti-inflammatory effect in carrageenan-induced inflammation by reducing oxidative stress, nitric oxide, and cytokine production. Therefore, it seems to be a promising anti-inflammatory molecule that merits further investigation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asteraceae/chemistry , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Diterpenes/pharmacology , Nitric Oxide/biosynthesis , Oxidative Stress/drug effects , Animals , Brazil , Carrageenan/antagonists & inhibitors , Diterpenes/chemistry , Edema , Interleukin-1beta/biosynthesis , Male , Mice , Neutrophil Infiltration/drug effects , Peritoneal Cavity , Peroxidase/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
14.
Parasitol Res ; 115(4): 1557-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26711452

ABSTRACT

Experimental models of mouse paw infection with L. amazonensis show an induction of a strong inflammatory response in the skin, and parasitic migration may occur to secondary organs with consequent tissue injury. There are few studies focusing on the resolution of damage in secondary organs caused by Leishmania species-related cutaneous leishmaniasis. We investigated the propolis treatment effect on liver inflammation induced by Leishmania amazonensis infection in the mouse paw. BALB/c mice were infected in the hind paw with L. amazonensis (10(7)) promastigote forms. After 15 days, animals were treated daily with propolis (5 mg/kg), Glucantime (10 mg/kg), or with propolis plus Glucantime combined. After 60 days, mice were euthanized and livers were collected for inflammatory process analysis. Liver microscopic analysis showed that propolis reduced the inflammatory process compared to untreated infected control. There was a decrease of liver myeloperoxidase and N-acetyl-ß-glucosaminidase activity levels, collagen fiber deposition, pro-inflammatory cytokine production, and plasma aspartate transaminase and alanine transaminase levels. Furthermore, propolis treatment enhanced anti-inflammatory cytokine levels and reversed hepatosplenomegaly. Our data demonstrated that daily low doses of Brazilian propolis reduced the secondary chronic inflammatory process in the liver caused by L. amazonensis subcutaneous infection in a susceptible mice strain.


Subject(s)
Inflammation/drug therapy , Leishmaniasis, Cutaneous/parasitology , Propolis/therapeutic use , Animals , Cytokines/biosynthesis , Inflammation/parasitology , Leishmania mexicana/drug effects , Leishmaniasis, Cutaneous/complications , Liver/drug effects , Liver/parasitology , Male , Meglumine , Meglumine Antimoniate , Mice , Mice, Inbred BALB C , Organometallic Compounds
15.
Eur J Pharmacol ; 749: 62-72, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25584775

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) is a therapeutic approach to increase peripheral neutrophil counts after anti-tumor therapies. Pain is the major side effect of G-CSF. Intraplantar administration of G-CSF in mice induces mechanical hyperalgesia. However, the peripheral mechanisms involved in this effect were not elucidated. Therefore, the participation of pronociceptive cytokines tumor necrosis factor (TNF) alpha (TNFα), interleukin (IL)-1 beta (IL-1ß) and antinociceptive cytokine IL-10 in G-CSF-induced mechanical hyperalgesia in mice was investigated. G-CSF-induced mechanical hyperalgesia was inhibited by systemic and local treatment with etanercept and IL-1 receptor antagonist (IL-1ra) or TNF receptor 1 (TNFR1) deficiency and increased in IL-10 deficient mice. In agreement, G-CSF injection induced significant TNFα, IL-1ß and IL-10 production in paw tissue. G-CSF-induced hyperalgesia was dose-dependently inhibited by thalidomide (5-45mg/kg) and pentoxifylline (0.5-13.5mg/kg), and treatment with these drugs inhibited G-CSF-induced TNFα, IL-1ß and IL-10 production. The combined treatment with pentoxifylline or thalidomide with morphine, at doses that are ineffective as single treatment, diminished G-CSF-induced hyperalgesia through inhibiting cytokine production. Indomethacin also reduces G-CSF hyperalgesia alone or combined with pentoxifylline or thalidomide. Thus, G-CSF-induced hyperalgesia might be mediate by peripheral production of pronociceptive cytokines TNFα and IL-1ß and down-regulated by IL-10. Systemic IL-1ra reduced G-CSF-induced increase of peripheral neutrophil counts. However, local treatment with morphine, IL-1ra or etanercept, and systemic treatment with indomethacin, etanercept, thalidomide and pentoxifylline did not alter G-CSF-induced mobilization of neutrophils. Therefore, this study advances in the understanding of G-CSF-induced hyperalgesia and suggests therapeutic approaches for its control.


Subject(s)
Granulocyte Colony-Stimulating Factor/adverse effects , Hyperalgesia/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Analgesics, Opioid/pharmacology , Animals , Etanercept , Hyperalgesia/chemically induced , Immunoglobulin G/pharmacology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-1beta/antagonists & inhibitors , Leukocyte Count , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Pentoxifylline/pharmacology , Receptors, Tumor Necrosis Factor , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Skin/drug effects , Skin/metabolism , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
16.
J Nat Prod ; 77(11): 2488-96, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25394199

ABSTRACT

Pimaradienoic acid (1) is a pimarane diterpene (ent-pimara-8(14),15-dien-19-oic acid) extracted at high amounts from various plants including Vigueira arenaria Baker. Compound 1 inhibited carrageenan-induced paw edema and acetic acid-induced abdominal writhing, which are its only known anti-inflammatory activities. Therefore, it is important to further investigate the analgesic effects of 1. Oral administration of 1 (1, 3, and 10 mg/kg) inhibited the acetic acid-induced writhing. This was also observed at 10 mg/kg via sc and ip routes. Both phases of the formalin- and complete Freund's adjuvant (CFA)-induced paw flinch and time spent licking the paw were inhibited by 1. Compound 1 inhibited carrageenan-, CFA-, and PGE2-induced mechanical hyperalgesia. Treatment with 1 inhibited carrageenan-induced production of TNF-α, IL-1ß, IL-33, and IL-10 and nuclear factor κB activation. Pharmacological inhibitors also demonstrated that the analgesic effects of 1 depend on activation of the NO-cyclic GMP-protein kinase G-ATP-sensitive potassium channel signaling pathway. Compound 1 did not alter plasma levels of AST, ALT, or myeloperoxidase activity in the stomach. These results demonstrate that 1 causes analgesic effects associated with the inhibition of NF-κB activation, reduction of cytokine production, and activation of the NO-cyclic GMP-protein kinase G-ATP-sensitive potassium channel signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Diterpenes/pharmacology , Acetic Acid/pharmacology , Analgesics/pharmacology , Carrageenan/pharmacology , Cyclic GMP/metabolism , Diterpenes/chemistry , Edema/chemically induced , Freund's Adjuvant/pharmacology , Hyperalgesia/drug therapy , Interleukin-10/metabolism , Interleukin-1beta/metabolism , KATP Channels/drug effects , Molecular Structure , Pain/drug therapy , Potassium Channels/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
17.
Naunyn Schmiedebergs Arch Pharmacol ; 387(11): 1053-68, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25116441

ABSTRACT

Nitric oxide plays an important role in various biological processes including antinociception. The control of its local concentration is crucial for obtaining the desired effect and can be achieved with exogenous nitric oxide-carriers such as ruthenium complexes. Therefore, we evaluated the analgesic effect and mechanism of action of the ruthenium nitric oxide donor [Ru(HEDTA)NO] focusing on the role of cytokines, oxidative stress and activation of the cyclic guanosine monophosphate/protein kinase G/ATP-sensitive potassium channel signaling pathway. It was observed that [Ru(HEDTA)NO] inhibited in a dose-dependent (1-10 mg/kg) manner the acetic acid-induced writhing response. At the dose of 1 mg/kg, [Ru(HEDTA)NO] inhibited the phenyl-p-benzoquinone-induced writhing response, and formalin- and complete Freund's adjuvant-induced licking and flinching responses. Systemic and local treatments with [Ru(HEDTA)NO] also inhibited the carrageenin-induced mechanical hyperalgesia and increase of myeloperoxidase activity in paw skin samples. Mechanistically, [Ru(HEDTA)NO] inhibited carrageenin-induced production of the hyperalgesic cytokines tumor necrosis factor-α and interleukin-1ß, and decrease of reduced glutathione levels. Furthermore, the inhibitory effect of [Ru(HEDTA)NO] in the carrageenin-induced hyperalgesia and myeloperoxidase activity was prevented by the treatment with ODQ (soluble guanylyl cyclase inhibitor), KT5823 (protein kinase G inhibitor) and glybenclamide (ATP-sensitive potassium channel inhibitor), indicating that [Ru(HEDTA)NO] inhibits inflammatory hyperalgesia by activating the cyclic guanosine monophosphate/protein kinase G/ATP-sensitive potassium channel signaling pathway, respectively. These results demonstrate that [Ru(HEDTA)NO] exerts its analgesic effect in inflammation by inhibiting pro-nociceptive cytokine production, oxidative imbalance and activation of the nitric oxide/cyclic guanosine monophosphate/protein kinase G/ATP-sensitive potassium channel signaling pathway in mice.


Subject(s)
Hyperalgesia/drug therapy , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Ruthenium Compounds/pharmacology , Animals , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Edetic Acid/administration & dosage , Edetic Acid/analogs & derivatives , Edetic Acid/chemistry , Inflammation/drug therapy , KATP Channels/metabolism , Male , Mice , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/chemistry , Nociception/drug effects , Oxidative Stress/drug effects , Ruthenium Compounds/administration & dosage , Ruthenium Compounds/chemistry , Signal Transduction/drug effects
18.
Pharmacol Biochem Behav ; 105: 157-65, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23470198

ABSTRACT

The activation of nitric oxide (NO) production is an analgesic mechanism shared by drugs such as morphine and diclofenac. Therefore, the controlled release of low amounts of NO seems to be a promising analgesic approach. In the present study, the antinociceptive effect of the ruthenium NO donor [Ru(bpy)2(NO)SO3](PF6) (complex I) was investigated. It was observed that complex I inhibited in a dose (0.3-10mg/kg)-dependent manner the acetic acid-induced writhing response. At the dose of 1mg/kg, complex I inhibited the phenyl-p-benzoquinone-induced writhing response and formalin- and complete Freund's adjuvant-induced licking and flinch responses. Additionally, complex I also inhibited transient receptor potential cation channel subfamily V member 1 (TRPV1)-dependent overt pain-like behavior induced by capsaicin. Complex I also inhibited the carrageenin-induced mechanical hyperalgesia and increase of myeloperoxidase activity (MPO) in paw skin samples. The inhibitory effect of complex I in the carrageenin-induced hyperalgesia, MPO activity and formalin was prevented by the treatment with ODQ, KT5823 and glybenclamide, indicating that complex I inhibits inflammatory hyperalgesia by activating the cGMP/PKG/ATP-sensitive potassium channel signaling pathway. The present study demonstrates the efficacy of a novel ruthenium NO donor and its analgesic mechanisms.


Subject(s)
Inflammation/prevention & control , Nitric Oxide Donors/pharmacology , Pain/prevention & control , Potassium Channels/metabolism , Ruthenium Compounds/pharmacology , Signal Transduction , TRPV Cation Channels/physiology , Adenosine Triphosphate/metabolism , Animals , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Electron Transport Complex I/metabolism , Mice , TRPV Cation Channels/metabolism
19.
Pharmacol Biochem Behav ; 105: 183-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23474372

ABSTRACT

Piracetam is a prototype of nootropic drugs used to improve cognitive impairment. However, recent studies suggest that piracetam can have analgesic and anti-inflammatory effects. Inflammatory pain is the result of a process that depends on neutrophil migration, cytokines and prostanoids release and oxidative stress. We analyze whether piracetam has anti-nociceptive effects and its mechanisms. Per oral pretreatment with piracetam reduced in a dose-dependent manner the overt pain-like behavior induced by acetic acid, phenyl-p-benzoquinone, formalin and complete Freund's adjuvant. Piracetam also diminished carrageenin-induced mechanical and thermal hyperalgesia, myeloperoxidase activity, and TNF-α-induced mechanical hyperalgesia. Piracetam presented analgesic effects as post-treatment and local paw treatment. The analgesic mechanisms of piracetam were related to inhibition of carrageenin- and TNF-α-induced production of IL-1ß as well as prevention of carrageenin-induced decrease of reduced glutathione, ferric reducing ability and free radical scavenging ability in the paw. These results demonstrate that piracetam presents analgesic activity upon a variety of inflammatory stimuli by a mechanism dependent on inhibition of cytokine production and oxidative stress. Considering its safety and clinical use for cognitive function, it is possible that piracetam represents a novel perspective of analgesic.


Subject(s)
Analgesics/pharmacology , Cytokines/biosynthesis , Oxidative Stress/drug effects , Piracetam/pharmacology , Animals , Male , Mice , Pain/prevention & control
20.
J Nat Prod ; 75(5): 896-904, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22574649

ABSTRACT

Kaurenoic acid [ent-kaur-16-en-19-oic acid (1)] is a diterpene present in several plants including Sphagneticola trilobata. The only documented evidence for its antinociceptive effect is that it inhibits the writhing response induced by acetic acid in mice. Therefore, the analgesic effect of 1 in different models of pain and its mechanisms in mice were investigated further. Intraperitoneal and oral treatment with 1 dose-dependently inhibited inflammatory nociception induced by acetic acid. Oral treatment with 1 also inhibited overt nociception-like behavior induced by phenyl-p-benzoquinone, complete Freund's adjuvant (CFA), and both phases of the formalin test. Compound 1 also inhibited acute carrageenin- and PGE(2)-induced and chronic CFA-induced inflammatory mechanical hyperalgesia. Mechanistically, 1 inhibited the production of the hyperalgesic cytokines TNF-α and IL-1ß. Furthermore, the analgesic effect of 1 was inhibited by l-NAME, ODQ, KT5823, and glybenclamide treatment, demonstrating that such activity also depends on activation of the NO-cyclic GMP-protein kinase G-ATP-sensitive potassium channel signaling pathway, respectively. These results demonstrate that 1 exhibits an analgesic effect in a consistent manner and that its mechanisms involve the inhibition of cytokine production and activation of the NO-cyclic GMP-protein kinase G-ATP-sensitive potassium channel signaling pathway.


Subject(s)
Acetic Acid/pharmacology , Asteraceae/chemistry , Cyclic GMP-Dependent Protein Kinases/drug effects , Cytokines/drug effects , Diterpenes/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , KATP Channels/drug effects , Pain/chemically induced , Pain/drug therapy , Administration, Oral , Animals , Carbazoles/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cytokines/biosynthesis , Diterpenes/chemistry , Diterpenes/isolation & purification , Dose-Response Relationship, Drug , Formaldehyde/pharmacology , Freund's Adjuvant/adverse effects , Freund's Adjuvant/pharmacology , Glyburide/pharmacology , Injections, Intraperitoneal , Interleukin-8/drug effects , Mice , Molecular Structure , NG-Nitroarginine Methyl Ester/pharmacology , Tumor Necrosis Factor-alpha/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...