Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci ; 208: 221-231, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30036488

ABSTRACT

AIMS: Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used and effective anti-inflammatories despite the well-known side effects such as gastrointestinal damage, acute kidney injury (AKI), and cardiovascular dysfunctions. Diclofenac is among the most prescribed NSAIDs due to its efficient analgesic and anti-inflammatory properties. Patients using diclofenac possess 77% risk increase to develop AKI. Activation of NF-κB contributes to diclofenac-induced AKI, which is in line with the use of glucocorticoids as one of the management choices to treat AKI patients. MAIN METHODS: In this work, we investigate the efficacy of pyrrolidine dithiocarbamate (PDTC) in diclofenac-induced AKI in mice given it is a known NF-κB inhibitor. KEY FINDINGS: We observed that diclofenac increased proteinuria and urine neutrophil gelatinase-associated lipocalin (NGAL), blood levels of urea, creatinine, oxidative stress, C-reactive protein (CRP), and pro-inflammatory cytokine after 24 h of a bolus administration. In renal tissue, diclofenac also induced morphological changes consistent with kidney damage, modulated cytokine production, increased oxidative stress and reduced antioxidant defenses. These alterations induced by diclofenac were accompanied by activation of NF-κB in the kidney. Treatment with PDTC dose-dependently reduced diclofenac-induced blood urea, creatinine, and oxidative stress. In addition, PDTC reduced proteinuria and urine NGAL levels and blood CRP and pro-inflammatory cytokines. In the kidney, PDTC inhibited diclofenac-induced morphological changes, pro-inflammatory cytokine production, oxidative stress, and NF-κB activation, and increased antioxidant defenses and anti-inflammatory cytokine IL-10. SIGNIFICANCE: Our data demonstrate that PDTC ameliorates diclofenac-induced AKI and that targeting NF-κB signaling pathway is a promising therapeutic approach for the treatment of diclofenac-induced AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Antioxidants/pharmacology , Cytokines/metabolism , Diclofenac/toxicity , NF-kappa B/metabolism , Oxidative Stress/drug effects , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Gene Expression Regulation/drug effects , Male , Mice , NF-kappa B/genetics , Signal Transduction/drug effects
2.
Physiol Behav ; 128: 277-87, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24518865

ABSTRACT

The injection of cytokines such as TNF-α induces muscle pain. Herein, it was addressed the role of endogenous TNF-α/TNFR1 signaling in intense acute swimming-induced muscle mechanical hyperalgesia in mice. Mice were exposed to water during 30 s (sham) or to a single session of 30-120 min of swimming. Intense acute swimming induced a dose-dependent (time of exercise-dependent) muscle mechanical hyperalgesia, which peaked after 24 h presenting characteristics of delayed onset muscle soreness (DOMS). The intense acute swimming (120 min)-induced muscle mechanical hyperalgesia was reduced in etanercept (soluble TNF receptor) treated and TNFR1 deficient ((-/-)) mice. TNF-α levels increased 2 and 4 h after intense acute swimming in soleus muscle (but not in gastrocnemius), and spinal cord, respectively. Exercise induced an increase of myeloperoxidase activity and decrease in reduced glutathione levels in an etanercept-sensitive and TNFR1-dependent manners in the soleus muscle, but not in the gastrocnemius muscle. Concluding, TNF-α/TNFR1 signaling mediates intense acute swimming-induced DOMS by an initial role in the soleus muscle followed by spinal cord, inducing muscle inflammatory hyperalgesia and oxidative stress. The knowledge of these mechanisms might contribute to improve the training of athletes, individuals with physical impairment and intense training such as military settings.


Subject(s)
Myalgia/etiology , Physical Exertion/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood Glucose/analysis , Dose-Response Relationship, Drug , Etanercept , Hydrocortisone/blood , Hyperalgesia/chemically induced , Immunoglobulin G/pharmacology , Leukocytes/physiology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/chemistry , Myalgia/chemically induced , Peroxidase/metabolism , Physical Exertion/drug effects , Receptors, Tumor Necrosis Factor , Spinal Cord/chemistry , Swimming/physiology , Tumor Necrosis Factor-alpha/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...