Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell Mol Biol (Noisy-le-grand) ; 69(10): 9-16, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37953590

ABSTRACT

Soluble epoxide hydrolase (sEH) inhibition has currently emerged as a therapeutic target in the treatment of various neuroinflammatory neurodegenerative diseases, including multiple sclerosis. Previously, we reported that treatment of mice with a sEH-selective inhibitor, 1-(1-propanoylpiperidin-4-yl)-3-[4-(trifluoromethoxy)phenyl]urea; TPPU), ameliorated chronic experimental autoimmune encephalomyelitis (EAE) induced by myelin oligodendrocyte glycoprotein 35-55 peptide immunization followed by injection of pertussis toxin to mice via regulating pro-inflammatory and anti-inflammatory pathways in the central nervous system. This study tested the hypothesis that the pro-inflammatory G protein-coupled receptor (GPR) 75 and anti-apoptotic phospholipase C (PLC) signaling pathways also contribute to the ameliorating effect of TPPU on chronic EAE. Brains and spinal cords of phosphate-buffered saline-, dimethyl sulfoxide-, or TPPU (3 mg/kg)-treated mice were used for the measurement of sEH, GPR75, Gaq/11, activator protein (AP)-1, PLC ß4, phosphoinositide 3-kinase (PI3K) p85a, Akt1, mitogen-activated protein kinase kinase (MEK) 1/2, extracellular signal-regulated kinase (ERK) 1/2, cyclic adenosine monophosphate-response element-binding protein (CREB) 1, B-cell lymphoma (Bcl)-2, semaphorin (SEMA) 3A, and myelin proteolipid protein (PLP) expression and/or activity by using the immunoblotting method. Expression of sEH, GPR75, Gaq/11, c-jun, phosphorylated c-Jun, and SEMA3A was lower, while PLCß4, phosphorylated PI3K p85a, phosphorylated Akt1, phosphorylated MEK1/2, phosphorylated ERK1/2, phosphorylated CREB1, Bcl-2, and myelin PLP expression was higher in the tissues of TPPU (3 mg/kg)-treated mice as compared with the EAE and vehicle control groups. Inhibition of sEH by TPPU ameliorates chronic EAE through suppressing pro-inflammatory GPR75/Gaq/11/AP-1 pathway and reducing expression of the remyelination inhibitor, SEMA3A, as well as increasing anti-apoptotic PLC/PI3K/Akt1/MEK1/2/ERK1/2/CREB1/Bcl-2 pathway activity and myelin PLP expression.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Phospholipases , Receptors, G-Protein-Coupled , Signal Transduction , Animals , Mice , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/metabolism , Mice, Inbred C57BL , Myelin Proteolipid Protein/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Semaphorin-3A , Receptors, G-Protein-Coupled/metabolism
2.
Cell Mol Biol (Noisy-le-grand) ; 69(9): 15-23, 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37807339

ABSTRACT

The nucleotide-binding oligomerization domain-like receptor X1 (NLRX1) has been associated with various anti-inflammatory mechanisms. We investigated whether the NLRX1 ligand docosahexaenoic acid (DHA) ameliorates lipopolysaccharide (LPS)-induced inflammatory hyperalgesia by interacting with tumor necrosis factor receptor-associated factor 6 (TRAF6)/inhibitor of kB (IkB) kinase (IKK)/IkB-a/nuclear factor-κB (NF-κB) signaling pathway in the central nervous system. Reaction time to thermal stimuli within 30 seconds was measured in male mice injected with saline, lipopolysaccharide (LPS), and/or DHA after 6 hours using the hot plate test. Co-immunoprecipitation and immunoblotting studies were performed to determine the activation of the TRAF6/IKK/IkB-a/NF-kB pathway in the brains and spinal cords of animals. Latency to the thermal stimulus was reduced by 30% in LPS-injected endotoxemic mice compared with saline-injected mice. Treatment with DHA significantly improved latency compared with endotoxemic mice. In the brain and spinal cord of LPS-injected mice, treatment with DHA also prevented the increase in the expression and/or activity of (1) IKKa/IKKß, IKKg, and K63 U in the NLRX1-immunoprecipitated tissues, (2) IKKa/IKKß, K63 U, and K48 U in the IKKg-immunoprecipitated tissues, and (3) IkB-α, NF-kB p65, and interleukin-1ß associated with decreased IkB-α expression. These findings suggest that inhibition of IKK/IkB-a/NF-kB signaling by dissociation of NLRX1 from TRAF6 in response to LPS treatment contributes to the protective effect of DHA against inflammatory hyperalgesia.


Subject(s)
I-kappa B Kinase , NF-kappa B , Male , Mice , Animals , NF-kappa B/metabolism , I-kappa B Kinase/metabolism , I-kappa B Kinase/pharmacology , Lipopolysaccharides/pharmacology , TNF Receptor-Associated Factor 6/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Ligands , Signal Transduction , Mitochondrial Proteins/metabolism
3.
J Cardiovasc Pharmacol ; 80(2): 276-293, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35323151

ABSTRACT

ABSTRACT: The orphan receptor, G protein-coupled receptor (GPR) 75, which has been shown to mediate various effects of 20-hydroxyeicosatetraenoic acid (20-HETE), is considered as a therapeutic target in the treatment of cardiovascular diseases in which changes in the production of 20-HETE play a key role in their pathogenesis. Our previous studies showed that 20-HETE mimetic, N -(20-hydroxyeicosa-5[Z],14[Z]-dienoyl)glycine (5,14-HEDGE), protects against vascular hyporeactivity, hypotension, tachycardia, and arterial inflammation induced by lipopolysaccharide (LPS) in rats. This study tested the hypothesis that the GPR75 signaling pathway mediates these effects of 5,14-HEDGE in response to systemic exposure to LPS. Mean arterial pressure reduced by 33 mm Hg, and heart rate increased by 102 beats/min at 4 hours following LPS injection. Coimmunoprecipitation studies demonstrated that (1) the dissociation of GPR75/Gα q/11 and GPR kinase interactor 1 (GIT1)/protein kinase C (PKC) α, the association of GPR75/GIT1, large conductance voltage and calcium-activated potassium subunit ß (MaxiKß)/PKCα, MaxiKß/proto-oncogene tyrosine-protein kinase (c-Src), and epidermal growth factor receptor (EGFR)/c-Src, MaxiKß, and EGFR tyrosine phosphorylation were decreased, and (2) the association of GIT1/c-Src was increased in the arterial tissues of rats treated with LPS. The LPS-induced changes were prevented by 5,14-HEDGE. N -[20-Hydroxyeicosa-6( Z ),15( Z )-dienoyl]glycine, a 20-HETE antagonist, reversed the effects of 5,14-HEDGE in the arterial tissues of LPS-treated rats. Thus, similar to 20-HETE, by binding to GPR75 and activating the Gα q/11 /PKCα/MaxiKß, GIT1/PKCα/MaxiKß, GIT1/c-Src/MaxiKß, and GIT1/c-Src/EGFR signaling pathways, 5,14-HEDGE may exert its protective effects against LPS-induced hypotension and tachycardia associated with vascular hyporeactivity and arterial inflammation.


Subject(s)
Arteritis , Hypotension , Shock, Septic , Animals , Cell Cycle Proteins/metabolism , ErbB Receptors/metabolism , Glycine , Hydroxyeicosatetraenoic Acids/metabolism , Hypotension/chemically induced , Hypotension/prevention & control , Lipopeptides , Lipopolysaccharides/toxicity , Protein Kinase C-alpha/metabolism , Protein Kinase C-alpha/pharmacology , Rats , Shock, Septic/chemically induced , Shock, Septic/drug therapy , Shock, Septic/prevention & control , Signal Transduction , Tachycardia , Tyrosine/pharmacology , Tyrosine/therapeutic use
4.
J Cardiovasc Pharmacol ; 79(3): 355-367, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34840266

ABSTRACT

ABSTRACT: The present study aimed to explore the contribution of mammalian target of rapamycin (mTOR) in deoxycorticosterone acetate (DOCA) salt-induced hypertension and related pathophysiological changes in cardiovascular and renal tissues. DOCA salt loading resulted in an increase in systolic blood pressure, diastolic blood pressure, and mean blood pressure along with the activity of ribosomal protein S6, the effector protein of mTOR. Treatment with rapamycin, the selective inhibitor of mTOR, initiated at the fourth week of DOCA- salt administration normalized the systolic blood pressure and attenuated ribosomal protein S6 activity in the heart, aorta, and kidney. Cardiac and vascular hypertrophy, oxidative stress, and infiltration of macrophages (CD68+), the marker of inflammation, were also reduced in rapamycin-treated, DOCA-salt, hypertensive rats. In addition, renal hypertrophy and dysfunction were also reduced with rapamycin-treated hypertensive rats. Moreover, these pathophysiological changes in DOCA-salt hypertensive rats were associated with increased NADPH oxidase (NOX) activity, gp91phox (formerly NOX2) expression, ERK1/2, and p38 MAPK activities in the heart, aorta, and kidney were minimized by rapamycin. These data indicate that mTOR plays an important role in regulating blood pressure and the development of cardiovascular and renal pathophysiological changes, most likely due to increased NOX expression/activity, ERK1/2, and p38 MAPK activity with macrophages infiltration in the heart, kidney, and aorta. Pharmacological inhibition of mTOR and related signaling pathways could serve as a novel target for the treatment of hypertension.


Subject(s)
Desoxycorticosterone Acetate , Hypertension , Acetates/adverse effects , Animals , Blood Pressure , Desoxycorticosterone Acetate/adverse effects , Hypertension/chemically induced , Hypertension/drug therapy , Hypertension/metabolism , Hypertrophy , Inflammation , Male , Mammals/metabolism , Oxidative Stress , Rats , Ribosomal Protein S6/metabolism , Sirolimus/adverse effects , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Can J Physiol Pharmacol ; 99(9): 921-934, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33641344

ABSTRACT

Neuroinflammation plays a critical role during sepsis triggered by microglial activation. Mammalian target of rapamycin (mTOR) has gained attraction in neuroinflammation, however, the mechanism remains unclear. Our goal was to assess the effects of mTOR inhibition by rapamycin on inflammation, microglial activation, oxidative stress, and apoptosis associated with the changes in the inhibitor-κB (IκB)-α/nuclear factor-κB (NF-κB)/hypoxia-inducible factor-1α (HIF-1α) pathway activity following a systemic challenge with lipopolysaccharide (LPS). Rats received saline (10 mL/kg), LPS (10 mg/kg), and (or) rapamycin (1 mg/kg) intraperitoneally. Inhibition of mTOR by rapamycin blocked phosphorylated form of ribosomal protein S6, NF-κB p65 activity by increasing degradation of IκB-α in parallel with HIF-1α expression increased by LPS in the kidney, heart, lung, and brain tissues. Rapamycin attenuated the increment in the expression of tumor necrosis factor-α and interleukin-1ß, the inducible nitric oxide synthase, gp91phox, and p47phox in addition to nitrite levels elicited by LPS in tissues or sera. Concomitantly, rapamycin treatment reduced microglial activation, brain expression of caspase-3, and Bcl-2-associated X protein while it increased expression of B cell lymphoma 2 induced by LPS. Overall, this study supports the hypothesis that mTOR contributes to the detrimental effect of LPS-induced systemic inflammatory response associated with neuroinflammation via IκB-α/NF-κB/HIF-1α signaling pathway.


Subject(s)
Inflammation/drug therapy , Neuroinflammatory Diseases/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , I-kappa B Proteins/physiology , Lipopolysaccharides , Male , Microglia/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/physiology , Transcription Factor RelA/physiology
6.
Drug Dev Res ; 82(6): 815-825, 2021 09.
Article in English | MEDLINE | ID: mdl-33559150

ABSTRACT

Epoxyeicosatrienoic acids (EETs) have anti-inflammatory effects and soluble epoxide hydrolase (sEH) inhibition might be a useful therapeutic approach to manage inflammatory disorders. The purpose of the study was to investigate whether nucleotide-binding and oligomerization domain-like receptor (NLR) C4 inflammasome-related pro-inflammatory and anti-inflammatory signaling pathways in the central nervous system (CNS) participates in the effect of trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea (TPPU), a potent sEH inhibitor, to prevent hyperalgesia in the LPS-induced pain mouse model. The latency of pain within 30 s was measured by the hot plate test in male mice injected with saline, lipopolysaccharide (LPS) (10 mg/kg), and/or TPPU (0.3, 0.5, or 1 mg/kg) after 6 h. Hyperalgesia induced by LPS was associated with decreased 14,15-dihydroxyeicosatrienoic acid and interleukin (IL)-1ß levels and enhanced expression of NLRC4, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), caspase-1 p20, IL-1ß, and caspase-11 p20 in the brains and spinal cords of the animals. Besides the increased expression of nicotinamide adenine dinucleotide phosphate oxidase (NOX) subunits (gp91phox and p47phox ) and nitrotyrosine, a decrease in NLRC3, inducible nitric oxide synthase (iNOS), and neuronal NOS (nNOS) expression was also observed in the tissues of LPS-treated mice. TPPU at 0.5 mg/kg dose prevented the changes induced by LPS. Likely, decreased activity of pro-inflammatory NLRC4/ASC/pro-caspase-1 and caspase-11 inflammasomes and NOX in addition to enhanced levels of anti-inflammatory EETs and expression of NLRC3, iNOS, and nNOS in the CNS of mice participates in the protective effect of TPPU against LPS-induced hyperalgesia.


Subject(s)
Inflammasomes , Lipopolysaccharides , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Epoxide Hydrolases/metabolism , Epoxide Hydrolases/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Intercellular Signaling Peptides and Proteins/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Pain , Signal Transduction , Urea
7.
Neurochem Res ; 46(3): 624-637, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33389386

ABSTRACT

A selective RXR agonist, bexarotene, has been shown to have anti-inflammatory, anti-nociceptive, and neuroprotective effects in several models of numerous neurological diseases characterized by systemic inflammation. The mechanisms underlying these effects remains unknown. To elucidate these mechanisms, we investigated whether the TLR4/MyD88/TAK1/NF-κB/COX-2 signaling pathway in the CNS mediates the effect of bexarotene to prevent hyperalgesia in the LPS-induced inflammatory pain mouse model. The reaction time to thermal stimuli within 30 s was evaluated by the hot plate test in male mice treated with saline, LPS (10 mg/kg), DMSO, and/or bexarotene (0.1, 1, 3, or 10 mg/kg) after 6 h. The latency to the thermal stimulus (18.11 ± 1.36 s) in the LPS-treated mice was significantly decreased by 30% compared with saline-treated mice (25.84 ± 1.99 s). Treatment with bexarotene only at a dose of 10 mg/kg showed a significant increase in the latency by 22.49 ± 1.00 s compared with LPS-treated mice. Bexarotene also prevented the reduction in RXRα protein expression associated with a rise in the expression of TLR4, MyD88, phosphorylated TAK1, NF-κB p65, phosphorylated NF-κB p65, COX-2, and IL-1ß proteins, in addition to COX-2 activity and levels of PGE2 and IL-1ß in the brains and spinal cords of the LPS-treated animals. Likely, decreased activity of TLR4/MyD88/TAK1/NF-κB/COX-2 signaling pathway in addition to increased pro-inflammatory cytokine formation in the CNS of mice participates in the protective effect of bexarotene against hyperalgesia induced by LPS.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Bexarotene/therapeutic use , Hyperalgesia/prevention & control , Retinoid X Receptors/agonists , Signal Transduction/drug effects , Animals , Cyclooxygenase 2/metabolism , Hyperalgesia/chemically induced , Lipopolysaccharides , MAP Kinase Kinase Kinases/metabolism , Male , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , NF-kappa B p50 Subunit/metabolism , Toll-Like Receptor 4/metabolism
8.
Inflammopharmacology ; 28(6): 1509-1524, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32128702

ABSTRACT

We aimed to determine the effect of soluble epoxide hydrolase (sEH) inhibition on chronic experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS), associated with changes in inflammasome-dependent and -independent inflammatory and anti-inflammatory pathways in the CNS of mice. C57BL/6 mice were used to induce chronic EAE by using an injection of MOG35-55 peptide/PT. Animals were observed daily and scored for EAE signs for 25 days after immunization. Following the induction of EAE, the scores were increased after 9 days and reached peak value as determined by ≥ 2 or ≤ 3 with 8% mortality rate on day 17. On day 17, mice were administered daily PBS, DMSO, or TPPU (a potent sEH inhibitor) (1, 3, or 10 mg/kg) until the end of the study. TPPU only at 3 mg/kg dose decreased the AUC values calculated from EAE scores obtained during the disease compared to EAE and vehicle control groups. On day 25, TPPU also caused an increase in the PPARα/ß/γ and NLRC3 proteins and a decrease in the proteins of TLR4, MyD88, NF-κB p65, p-NF-κB p65, iNOS/nNOS, COX-2, NLRC4, ASC, caspase-1 p20, IL-1ß, caspase-11 p20, NOX subunits (gp91phox and p47phox), and nitrotyrosine in addition to 14,15-DHET and IL-1ß levels compared to EAE and vehicle control groups. Our findings suggest that pharmacological inhibition of sEH attenuates chronic EAE likely because of enhanced levels of anti-inflammatory EETs in addition to PPARα/ß/γ and NLRC3 expression associated with suppressed inflammatory TLR4/MyD88/NF-κB signalling pathway, NLRC4/ASC/pro-caspase-1 inflammasome, caspase-11 inflammasome, and NOX activity that are responsible for inflammatory mediator formation in the CNS of mice.


Subject(s)
Anti-Inflammatory Agents/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Epoxide Hydrolases/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Signal Transduction/physiology , Animals , Central Nervous System/metabolism , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
10.
Prostaglandins Other Lipid Mediat ; 145: 106377, 2019 12.
Article in English | MEDLINE | ID: mdl-31586592

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Septic shock, the most common form of vasodilatory shock, is a subset of sepsis in which circulatory and cellular/metabolic abnormalities are severe enough to increase mortality. Inflammatory shock constitutes the hallmark of sepsis, but also a final common pathway of any form of severe long-term tissue hypoperfusion. The pathogenesis of inflammatory shock seems to be due to circulating substances released by pathogens (e.g., bacterial endotoxins) and host immuno-inflammatory responses (e.g., changes in the production of histamine, bradykinin, serotonin, nitric oxide [NO], reactive nitrogen and oxygen species, and arachidonic acid [AA]-derived eicosanoids mainly through NO synthase, cyclooxygenase, and cytochrome P450 [CYP] pathways, and proinflammatory cytokine formation). Therefore, refractory hypotension to vasoconstrictors with end-organ hypoperfusion is a life threatening feature of inflammatory shock. This review summarizes the current knowledge regarding the role of eicosanoids derived from CYP pathway of AA in animal models of inflammatory shock syndromes with an emphasis on septic shock in addition to potential therapeutic strategies targeting specific CYP isoforms responsible for proinflammatory/anti-inflammatory mediator production.


Subject(s)
Arachidonic Acid/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Nitric Oxide Synthase/metabolism , Shock/metabolism , Animals , Humans , Inflammation/metabolism , Inflammation/pathology , Shock/pathology
11.
Can J Physiol Pharmacol ; 97(12): 1193-1203, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31505123

ABSTRACT

Mammalian target of rapamycin (mTOR) has been recognized with potential immunomodulatory properties playing an important role in various physiopathological processes including ischemia-reperfusion (I/R) injury. I/R injury stimulate reactive oxygen and nitrogen species by activating nicotinamide adenine dinucleotide phosphate oxidase and inducible nitric oxide synthase, respectively. Controversial results have been obtained in different I/R models following localized I/R; however, the precise role of the mTOR signaling pathway remains undefined. The objective of the current study was to evaluate the role of the mTOR in oxidative-nitrosative stress and inflammation in hindlimb I/R-induced injury in target and remote organ injuries. In rats subjected to I/R, an increased expression of ribosomal protein S6 (rpS6), inhibitor κB (IκB)-α, nuclear factor-κB (NF-κB) p65, inducible nitric oxide synthase, cyclooxygenase 2, gp91phox, and levels of tumor necrosis factor α, nitrite, nitrotyrosine, malondialdehyde and the activities of myeloperoxidase and catalase in the tissues and (or) sera were detected. Treatment with rapamycin, a selective inhibitor of mTOR, reversed all the I/R-induced changes as manifested by its anti-inflammatory and antioxidant effects in kidney and gastrocnemius muscle of rats. Collectively, these findings suggest that rapamycin protects against I/R-induced oxidative-nitrosative stress and inflammation leading to organ injuries via suppression of mTOR/IκB-α/NF-κB signaling pathway.


Subject(s)
Hindlimb/blood supply , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Reperfusion Injury/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Biomarkers/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , NF-KappaB Inhibitor alpha/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6/metabolism , Transcription Factor RelA/metabolism
12.
Clin Exp Pharmacol Physiol ; 46(2): 173-182, 2019 02.
Article in English | MEDLINE | ID: mdl-30347478

ABSTRACT

We have previously demonstrated that the activation of the spleen tyrosine kinase (Syk)/inhibitory-κB (IκB)-α/nuclear factor-κB (NF-κB) p65 signalling pathway contributes to hypotension and inflammatory response in a rat models of zymosan (ZYM)-induced non-septic shock. The purpose of this study was to further examine the possible mechanism underlying the effect of inhibition of Syk by BAY61-3606 via NF-κB activity at the level of nuclear translocation regarding the production of vasodilator and proinflammatory mediators in lipopolysaccharide (LPS) (septic)- and ZYM (non-septic)-induced shock. Administration of LPS (10 mg/kg, ip) or ZYM (500 mg/kg, ip) to male Wistar rats decreased mean arterial pressure and increased heart rate that was associated with an increase in the activities of cyclooxygenase and nitric oxide synthase, tumour necrosis factor-α, and interleukin-8 levels, and NF-κB activation and nuclear translocation in sera and/or cardiovascular and renal tissues. BAY61-3606 (3 mg/kg, ip), the selective Syk inhibitor, given 1 hour after LPS- or ZYM injection reversed all the above-mentioned effects. These results suggest that Syk contributes to the LPS- or ZYM-induced hypotension and inflammation associated with transactivation of NF-κB in septic and non-septic shock.


Subject(s)
Hypotension/drug therapy , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Niacinamide/analogs & derivatives , Pyrimidines/pharmacology , Shock, Septic/drug therapy , Syk Kinase/antagonists & inhibitors , Zymosan/pharmacology , Animals , Cyclooxygenase 2/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Hypotension/metabolism , Hypotension/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Interleukin-8/metabolism , Male , NF-KappaB Inhibitor alpha/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
13.
Clin Exp Pharmacol Physiol ; 45(2): 155-165, 2018 02.
Article in English | MEDLINE | ID: mdl-28949404

ABSTRACT

Spleen tyrosine kinase (Syk), a non-receptor tyrosine kinase, plays an important role in allergic diseases and inflammation. Syk triggers several intracellular signalling cascades including Toll-like receptor signalling to activate inflammatory responses following fungal infection but the role of this enzyme in zymosan (ZYM)-induced non-septic shock and its impacts on hypotension and inflammation in rats is not well understood. This study was conducted to determine the effects of Syk inhibition on ZYM-induced alterations in the expression and/or activities of Syk, inhibitor ĸB (IĸB)-α, and nuclear factor-ĸB (NF-ĸB) p65. We also examined the effect of Syk inhibition on inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, and tumour necrosis factor (TNF)-α, and activity of myeloperoxidase (MPO) that contribute to hypotension and inflammation. Administration of ZYM (500 mg/kg, ip) to male Wistar rats decreased blood pressure and increased heart rate. These changes were associated with increased expression and/or activities of Syk, NF-κB p65, iNOS and COX-2 and decreased expression of IκB-α with enhanced levels of nitrite, nitrotyrosine, 6-keto-PGF1α , and TNF-α and activity of MPO in renal, cardiac and vascular tissues. ZYM administration also elevated serum and tissue nitrite levels. The selective Syk inhibitor BAY 61-3606 (3 mg/kg, ip) given 1 hour after ZYM injection reversed all of these changes induced by ZYM. These results suggest that Syk/IĸB-α/NF-ĸB pathway activation contributes to hypotension and inflammation caused by the production of vasodilator and proinflammatory mediators in the zymosan-induced non-septic shock model.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Niacinamide/analogs & derivatives , Pyrimidines/therapeutic use , Shock/chemically induced , Syk Kinase/metabolism , Animals , Gene Expression Regulation/drug effects , I-kappa B Kinase/genetics , Male , NF-kappa B/genetics , Niacinamide/therapeutic use , Rats , Rats, Wistar , Shock/drug therapy , Syk Kinase/antagonists & inhibitors , Syk Kinase/genetics , Zymosan/toxicity
14.
Inflammation ; 41(1): 337-355, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29188497

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that can activate or inhibit the expression of many target genes by forming a heterodimer complex with the retinoid X receptor (RXR). The aim of this study was to investigate effects of bexarotene, a selective RXRα agonist, on the changes in renal, cardiac, hepatic, and pulmonary expression/activity of inducible nitric oxide synthase (iNOS) and cytochrome P450 (CYP) 4F6 in relation to PPARα/ß/γ-RXRα heterodimer formation in a rat model of septic shock. Rats were injected with dimethyl sulfoxide or bexarotene 1 h after administration of saline or lipopolysaccharide (LPS). Mean arterial pressure (MAP) and heart rate (HR) were recorded from rats, which had received either saline or LPS before and after 1, 2, 3, and 4 h. Serum iNOS, LTB4, myeloperoxidase (MPO), and lactate dehydrogenase (LDH) levels as well as tissue iNOS and CYP4F6 mRNA expression in addition to PPARα/ß/γ and RXRα proteins were measured. LPS-induced decrease in MAP and increase in HR were associated with a decrease in PPARα/ß/γ-RXRα heterodimer formation and CYP4F6 mRNA expression. LPS also caused an increase in systemic iNOS, LTB4, MPO, and LDH levels as well as iNOS mRNA expression. Bexarotene at 0.1 mg/kg (i.p.) prevented the LPS-induced changes, except tachycardia. The results suggest that increased formation of PPARα/ß/γ-RXRα heterodimers and CYP4F6 expression/activity in addition to decreased iNOS expression contributes to the beneficial effect of bexarotene to prevent the hypotension associated with inflammation and tissue injury during rat endotoxemia.


Subject(s)
Arterial Pressure/drug effects , Hypotension/prevention & control , Inflammation/drug therapy , Retinoid X Receptor alpha/agonists , Shock, Septic/drug therapy , Tetrahydronaphthalenes/pharmacology , Animals , Bexarotene , Cytochrome P450 Family 4/genetics , Cytochrome P450 Family 4/metabolism , Disease Models, Animal , Heart Rate/drug effects , Hypotension/chemically induced , Hypotension/metabolism , Hypotension/physiopathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/physiopathology , Kidney/drug effects , Kidney/metabolism , L-Lactate Dehydrogenase/blood , Leukotriene B4/blood , Lipopolysaccharides , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Multiprotein Complexes , Myocardium/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Peroxidase/blood , Peroxisome Proliferator-Activated Receptors/metabolism , Rats, Wistar , Retinoid X Receptor alpha/metabolism , Shock, Septic/chemically induced , Shock, Septic/metabolism , Shock, Septic/physiopathology , Signal Transduction/drug effects
15.
Inflammation ; 41(1): 276-298, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29110153

ABSTRACT

Mammalian target of rapamycin (mTOR), a serine/threonine kinase regulate variety of cellular functions including cell growth, differentiation, cell survival, metabolism, and stress response, is now appreciated to be a central regulator of immune responses. Because mTOR inhibitors enhanced the anti-inflammatory activities of regulatory T cells and decreased the production of proinflammatory cytokines by macrophages, mTOR has been a pharmacological target for inflammatory diseases. In this study, we examined the role of mTOR in the production of proinflammatory and vasodilator mediators in zymosan-induced non-septic shock model in rats. To elucidate the mechanism by which mTOR contributes to non-septic shock, we have examined the activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system caused by mTOR/mitogen-activated protein kinase kinase (MEK1)/extracellular signal-regulated kinase (ERK1/2)/inhibitor κB kinase (IKKß)/inhibitor of κB (IκB-α)/nuclear factor-κB (NF-κB) signalling pathway activation. After 1 h of zymosan (500 mg/kg, i.p.) administration to rats, mean arterial blood pressure (MAP) was decreased and heart rate (HR) was increased. These changes were associated with increased expression and/or activities of ribosomal protein S6, MEK1, ERK1/2, IKKß, IκB-α and NF-κB p65, and NADPH oxidase system activity in cardiovascular and renal tissues. Rapamycin (1 mg/kg, i.p.), a selective mTOR inhibitor, reversed these zymosan-induced changes in these tissues. These observations suggest that activation of mTOR/MEK1/ERK1/2/IKKß/IκB-α/NF-κB signalling pathway with proinflammatory and vasodilator mediator formation and NADPH oxidase system activity contributes to systemic inflammation in zymosan-induced non-septic shock. Thus, mTOR may be an optimal target for the treatment of the diseases characterized by the severe systemic inflammatory response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , I-kappa B Proteins/metabolism , Inflammation/prevention & control , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Protein Kinase Inhibitors/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Zymosan , Animals , Arterial Pressure/drug effects , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Male , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Rats, Wistar , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors , Vasodilation/drug effects
16.
Eur J Pharmacol ; 802: 7-19, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28228357

ABSTRACT

Mammalian target of rapamycin (mTOR), a serine/threonine kinase plays an important role in various pathophysiological processes including cancer, metabolic diseases, and inflammation. Although mTOR participates in Toll-like receptor 4 signalling in different cell types, the role of this enzyme in sepsis pathogenesis and its effects on hypotension and inflammation in endotoxemic rats remains unclear. In this study we investigated the effects of mTOR inhibition on lipopolysaccharide (LPS)-induced changes on expressions and/or activities of ribosomal protein S6 (rpS6), an mTOR substrate, nuclear factor-κB (NF-κB) p65, inhibitor κB (IκB)-α, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2 with production of nitric oxide, peroxynitrite, prostacyclin, and tumor necrosis factor (TNF)-α and activity of myeloperoxidase (MPO), which results in hypotension and inflammation. Injection of LPS (10mg/kg, i.p.) to male Wistar rats decreased blood pressure and increased heart rate that were associated with elevated nitrotyrosine, 6-keto-PGF1α, and TNF-α levels and MPO activity, and increased expressions and/or activities of rpS6, NF-κB p65, iNOS, and COX-2 and decreased expression of IκB-α in renal, cardiac, and vascular tissues. LPS also increased serum and tissue nitrite levels. Rapamycin (1mg/kg, i.p.) given one h after injection of LPS reversed these effects of LPS. These data suggest that the activation of mTOR/IκB-α/NF-κB pathway associated with vasodilator and proinflammatory mediator formation contributes to LPS-induced hypotension and inflammation.


Subject(s)
Hypotension/chemically induced , Hypotension/pathology , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Arterial Pressure/drug effects , Cyclooxygenase 2/metabolism , Epoprostenol/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Heart Rate/drug effects , Hypotension/metabolism , Hypotension/physiopathology , Inflammation/chemically induced , Inflammation/pathology , Male , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Peroxynitrous Acid/biosynthesis , Rats , Rats, Wistar , Ribosomal Protein S6/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
Inflammation ; 40(2): 366-386, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27924425

ABSTRACT

The nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3), an intracellular signaling molecule that senses many environmental- and pathogen/host-derived factors, has been implicated in the pathogenesis of several diseases associated with inflammation. It has been suggested that NLRP3 inflammasome inhibitors may have a therapeutic potential in the treatment of NLRP3-related inflammatory diseases. The aim of this study was to determine whether inhibition of NLRP3 inflammasome prevents inflammatory hyperalgesia induced by lipopolysaccharide (LPS) in mice as well as changes in expression/activity of nuclear factor κB (NF-κB), caspase-1/11, nicotinamide adenine dinucleotide phosphate oxidase (NOX), and endothelial/neuronal/inducible nitric oxide synthase (eNOS/nNOS/iNOS) that may regulate NLRP3/apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)/pro-caspase-1 inflammasome formation and activity by using a selective NLRP3 inflammasome inhibitor, MCC950. Male mice received saline (10 ml/kg; i.p.), LPS (10 mg/kg; i.p.), and/or MCC950 (3 mg/kg; i.p.). Reaction time to thermal stimuli within 1 min was evaluated after 6 h. The mice were killed and the brains, hearts, and lungs were collected for measurement of NF-κB, caspase-1, caspase-11, NLRP3, ASC, NOX subunits (gp91phox; NOX2), and p47phox; NOXO2), nitrotyrosine, eNOS, nNOS, iNOS, and ß-actin protein expression, NOS activity, and interleukin (IL)-1ß levels. LPS-induced hyperalgesia was associated with a decrease in eNOS, nNOS, and iNOS protein expression and activity as well as an increase in expression of NF-κB p65, caspase-1 p20, caspase-11 p20, NLRP3, ASC, gp91phox, p47phox, and nitrotyrosine proteins in addition to elevated IL-1ß levels. The LPS-induced changes were prevented by MCC950. The results suggest that inhibition of NLRP3/ASC/pro-caspase-1 inflammasome formation and activity prevents inflammatory hyperalgesia induced by LPS in mice as well as changes in NF-κB, caspase-11, NOX2, NOXO2, and eNOS/nNOS/iNOS expression/activity.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/therapeutic use , Hyperalgesia/drug therapy , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfones/therapeutic use , Animals , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Caspases/metabolism , Caspases, Initiator , Furans , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Hyperalgesia/pathology , Hyperalgesia/prevention & control , Indenes , Inflammasomes/chemistry , Inflammasomes/drug effects , Inflammation , Lipopolysaccharides , Male , Mice , Mice, Inbred BALB C , NADPH Oxidases/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitric Oxide Synthase/metabolism , Protein Isoforms/metabolism , Sulfonamides , Sulfones/administration & dosage
18.
J Basic Clin Pharm ; 7(2): 49-59, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27057126

ABSTRACT

OBJECTIVES: There is a need to remove excess iron with iron chelation therapy (ICT) to avoid the serious clinical sequelae associated with iron overload in patients with beta thalassemia major (BTM) and sickle cell anemia (SCA). Due to the effects of the diseases and their treatments, ICT is still a major reason for unsatisfactory compliance. The aim of this single-center observational study was to evaluate the quality of life, clinical effectiveness, and satisfaction in pediatric and adult patients with BTM and SCA receiving deferasirox (DFX) chelation therapy. METHODS: In this study, 37 pediatric and 35 adult patients with BTM or SCA receiving DFX for at least 6 months participated. Upon receipt of Informed Consent Form, Case Report Form, Demographic Data Collection Form, Child Health Questionnaire-Parent Form, Life Quality Survey Short Form-36, and ICT Satisfaction Survey were used to obtain data for the effectiveness of ICT and parameters that may affect compliance to treatment and life quality of the participants. RESULTS: As a main index for the effectiveness of DFX chelation therapy, serum ferritin levels were higher than the normal values in the patients receiving DFX. The increased ferritin levels were also associated with hematological and biochemical abnormalities. Our findings regarding quality of life and satisfaction with DFX chelation therapy indicated that the patients with BTM or SCA had lower scores. Overall, problems with treatment regimen and side effects appeared to be common causes of poor compliance to DFX chelation therapy. CONCLUSIONS: Our findings suggest that health care providers should be aware of the importance of monitoring iron load with timely initiation of DFX chelation therapy and ongoing adjustments to chelation regimens and/or transfusion methods to decrease hospitalizations and improve compliance to ICT of the patients with BTM and SCA.

19.
Inflamm Res ; 65(5): 367-87, 2016 May.
Article in English | MEDLINE | ID: mdl-26875149

ABSTRACT

OBJECTIVES: We have previously demonstrated that downregulation of the MyD88/TAK1-dependent signaling pathway associated with increased CYP4A1 expression and 20-HETE formation participates in the protective effect of N-(20-hydroxyeicosa-5[Z],14[Z]-dienoyl)glycine (5,14-HEDGE), a 20-HETE mimetic, against vascular hyporeactivity, hypotension, tachycardia, inflammation, and mortality in a rodent model of septic shock. The aim of this study was to determine whether increased renal and cardiovascular expression of PPARα/ß/γ and RXRα associated with decreased expression and/or activity of AP-1 and importin-α3 participates in the protective effect of 5,14-HEDGE in response to systemic administration of lipopolysaccharide (LPS). METHODS: Conscious male Wistar rats received saline (4 ml/kg) or LPS (10 mg/kg) at time 0. Blood pressure and heart rate were measured using a tail-cuff device. Separate groups of LPS-treated rats were given 5,14-HEDGE (30 mg/kg) 1 h after injection of saline or LPS. The rats were killed 4 h after saline or LPS administration and the kidney, heart, thoracic aorta, and superior mesenteric artery were collected for measurement of protein expression. RESULTS: Blood pressure fell by 33 mmHg and heart rate rose by 72 beats/min at 4 h after LPS administration. In LPS-treated rats, tissue protein expressions of cytosolic/nuclear PPARα/ß/γ and nuclear RXRα, in addition to nuclear translocation of PPARα/ß/γ proteins, were decreased, while cytosolic/nuclear AP-1 subunit c-jun/phosphorylated c-jun and importin-α3 protein expression as well as their nuclear translocation were increased. The LPS-induced changes were prevented by 5,14-HEDGE. CONCLUSIONS: The results suggest that an increase in the expression of PPARα/ß/γ and RXRα as well as a decrease in AP-1 and importin-α3 expression/activity participates in the protective effect of 5,14-HEDGE against hypotension, tachycardia, and inflammation during endotoxemia and thus have a beneficial effect in septic shock treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lipopeptides/pharmacology , Shock, Septic/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Disease Models, Animal , Hydroxyeicosatetraenoic Acids , Hypotension/drug therapy , Hypotension/metabolism , Kidney/drug effects , Kidney/metabolism , Lipopeptides/therapeutic use , Lipopolysaccharides , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Myocardium/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Rats, Wistar , Retinoid X Receptor alpha/metabolism , Shock, Septic/drug therapy , Tachycardia/drug therapy , Tachycardia/metabolism , Transcription Factor AP-1/metabolism , alpha Karyopherins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...