Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Free Radic Biol Med ; 53(5): 1123-38, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22683818

ABSTRACT

Mitochondrial reactive oxygen species generation has been implicated in the pathophysiology of ischemia-reperfusion (I/R) injury; however, its exact role and its spatial-temporal relationship with inflammation are elusive. Herein we explore the spatial-temporal relationship of oxidative/nitrative stress and inflammatory response during the course of hepatic I/R and the possible therapeutic potential of mitochondrial-targeted antioxidants, using a mouse model of segmental hepatic ischemia-reperfusion injury. Hepatic I/R was characterized by early (at 2 h of reperfusion) mitochondrial injury, decreased complex I activity, increased oxidant generation in the liver or liver mitochondria, and profound hepatocellular injury/dysfunction with acute proinflammatory response (TNF-α, MIP-1α/CCL3, MIP-2/CXCL2) without inflammatory cell infiltration, followed by marked neutrophil infiltration and a more pronounced secondary wave of oxidative/nitrative stress in the liver (starting from 6 h of reperfusion and peaking at 24 h). Mitochondrially targeted antioxidants, MitoQ or Mito-CP, dose-dependently attenuated I/R-induced liver dysfunction, the early and delayed oxidative and nitrative stress response (HNE/carbonyl adducts, malondialdehyde, 8-OHdG, and 3-nitrotyrosine formation), and mitochondrial and histopathological injury/dysfunction, as well as delayed inflammatory cell infiltration and cell death. Mitochondrially generated oxidants play a central role in triggering the deleterious cascade of events associated with hepatic I/R, which may be targeted by novel antioxidants for therapeutic advantage.


Subject(s)
Antioxidants/therapeutic use , Inflammation/metabolism , Liver Diseases/drug therapy , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/drug therapy , Animals , Antioxidants/pharmacology , Cyclic N-Oxides/pharmacology , Cyclic N-Oxides/therapeutic use , Dose-Response Relationship, Drug , Inflammation/drug therapy , Liver Diseases/metabolism , Liver Diseases/physiopathology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Oxidative Stress/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
2.
Free Radic Biol Med ; 52(8): 1325-33, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22326488

ABSTRACT

(E)-ß-caryophyllene (BCP) is a natural sesquiterpene found in many essential oils of spice (best known for contributing to the spiciness of black pepper) and food plants with recognized anti-inflammatory properties. Recently it was shown that BCP is a natural agonist of endogenous cannabinoid 2 (CB(2)) receptors, which are expressed in immune cells and mediate anti-inflammatory effects. In this study we aimed to test the effects of BCP in a clinically relevant murine model of nephropathy (induced by the widely used antineoplastic drug cisplatin) in which the tubular injury is largely dependent on inflammation and oxidative/nitrative stress. ß-caryophyllene dose-dependently ameliorated cisplatin-induced kidney dysfunction, morphological damage, and renal inflammatory response (chemokines MCP-1 and MIP-2, cytokines TNF-α and IL-1ß, adhesion molecule ICAM-1, and neutrophil and macrophage infiltration). It also markedly mitigated oxidative/nitrative stress (NOX-2 and NOX-4 expression, 4-HNE and 3-NT content) and cell death. The protective effects of BCP against biochemical and histological markers of nephropathy were absent in CB(2) knockout mice. Thus, BCP may be an excellent therapeutic agent to prevent cisplatin-induced nephrotoxicity through a CB(2) receptor-dependent pathway. Given the excellent safety profile of BCP in humans it has tremendous therapeutic potential in a multitude of diseases associated with inflammation and oxidative stress.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney/drug effects , Receptor, Cannabinoid, CB2/physiology , Sesquiterpenes/pharmacology , Animals , Base Sequence , DNA Primers , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Nitrates/metabolism , Oxidative Stress , Polycyclic Sesquiterpenes , Real-Time Polymerase Chain Reaction
3.
Diabetes ; 61(3): 716-27, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315315

ABSTRACT

Endocannabinoids and cannabinoid 1 (CB(1)) receptors have been implicated in cardiac dysfunction, inflammation, and cell death associated with various forms of shock, heart failure, and atherosclerosis, in addition to their recognized role in the development of various cardiovascular risk factors in obesity/metabolic syndrome and diabetes. In this study, we explored the role of CB(1) receptors in myocardial dysfunction, inflammation, oxidative/nitrative stress, cell death, and interrelated signaling pathways, using a mouse model of type 1 diabetic cardiomyopathy. Diabetic cardiomyopathy was characterized by increased myocardial endocannabinoid anandamide levels, oxidative/nitrative stress, activation of p38/Jun NH(2)-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs), enhanced inflammation (tumor necrosis factor-α, interleukin-1ß, cyclooxygenase 2, intracellular adhesion molecule 1, and vascular cell adhesion molecule 1), increased expression of CB(1), advanced glycation end product (AGE) and angiotensin II type 1 receptors (receptor for advanced glycation end product [RAGE], angiotensin II receptor type 1 [AT(1)R]), p47(phox) NADPH oxidase subunit, ß-myosin heavy chain isozyme switch, accumulation of AGE, fibrosis, and decreased expression of sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase (SERCA2a). Pharmacological inhibition or genetic deletion of CB(1) receptors attenuated the diabetes-induced cardiac dysfunction and the above-mentioned pathological alterations. Activation of CB(1) receptors by endocannabinoids may play an important role in the pathogenesis of diabetic cardiomyopathy by facilitating MAPK activation, AT(1)R expression/signaling, AGE accumulation, oxidative/nitrative stress, inflammation, and fibrosis. Conversely, CB(1) receptor inhibition may be beneficial in the treatment of diabetic cardiovascular complications.


Subject(s)
Diabetic Cardiomyopathies/etiology , Heart/physiopathology , Inflammation/etiology , Myocardium/pathology , Oxidative Stress , Receptor, Cannabinoid, CB1/physiology , Animals , Apoptosis , Arachidonic Acids/analysis , Diabetic Cardiomyopathies/physiopathology , Endocannabinoids , Fibrosis , Glycation End Products, Advanced/analysis , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Polyunsaturated Alkamides/analysis , Receptor, Angiotensin, Type 1/analysis , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Ventricular Function, Left
4.
Free Radic Biol Med ; 52(2): 497-506, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22120494

ABSTRACT

Cisplatin is a widely used antineoplastic agent; however, its major limitation is the development of dose-dependent nephrotoxicity whose precise mechanisms are poorly understood. Here we show not only that mitochondrial dysfunction is a feature of cisplatin nephrotoxicity, but also that targeted delivery of superoxide dismutase mimetics to mitochondria largely prevents the renal effects of cisplatin. Cisplatin induced renal oxidative stress, deterioration of mitochondrial structure and function, an intense inflammatory response, histopathological injury, and renal dysfunction. A single systemic dose of mitochondrially targeted antioxidants, MitoQ or Mito-CP, dose-dependently prevented cisplatin-induced renal dysfunction. Mito-CP also prevented mitochondrial injury and dysfunction, renal inflammation, and tubular injury and apoptosis. Despite being broadly renoprotective against cisplatin, Mito-CP did not diminish cisplatin's antineoplastic effect in a human bladder cancer cell line. Our results highlight the central role of mitochondrially generated oxidants in the pathogenesis of cisplatin nephrotoxicity. Because similar compounds seem to be safe in humans, mitochondrially targeted antioxidants may represent a novel therapeutic approach against cisplatin nephrotoxicity.


Subject(s)
Acute Kidney Injury/prevention & control , Antineoplastic Agents/adverse effects , Antioxidants/pharmacology , Cisplatin/adverse effects , Cyclic N-Oxides/pharmacology , Organophosphorus Compounds/pharmacology , Ubiquinone/analogs & derivatives , Acute Kidney Injury/chemically induced , Animals , Antioxidants/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/therapeutic use , Cytoprotection , Electron Transport Complex IV/metabolism , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Kidney Tubules/drug effects , Kidney Tubules/enzymology , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , NADH Dehydrogenase/metabolism , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/therapeutic use , Oxidative Stress/drug effects , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
5.
Free Radic Biol Med ; 51(9): 1774-88, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21884784

ABSTRACT

Cisplatin is a commonly used chemotherapeutic drug, the clinical use of which is limited by the development of dose-dependent nephrotoxicity. Enhanced inflammatory response, oxidative stress, and cell death have been implicated in the development of cisplatin-induced nephropathy; however, the precise mechanisms are elusive. Overactivation of the nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) by oxidative DNA damage under various pathological conditions promotes cell death and up-regulation of key proinflammatory pathways. In this study, using a well-established model of nephropathy, we have explored the role of PARP-1 in cisplatin-induced kidney injury. Genetic deletion or pharmacological inhibition of PARP-1 markedly attenuated the cisplatin-induced histopathological damage, impaired renal function (elevated serum BUN and creatinine levels), and enhanced inflammatory response (leukocyte infiltration; TNF-α, IL-1ß, F4/80, adhesion molecules ICAM-1/VCAM-1 expression) and consequent oxidative/nitrative stress (4-HNE, 8-OHdG, and nitrotyrosine content; NOX2/NOX4 expression). PARP inhibition also facilitated the cisplatin-induced death of cancer cells. Thus, PARP activation plays an important role in cisplatin-induced kidney injury, and its pharmacological inhibition may represent a promising approach to preventing the cisplatin-induced nephropathy. This is particularly exciting because several PARP inhibitors alone or in combination with DNA-damaging anticancer agents show considerable promise in clinical trials for treatment of various malignancies (e.g., triple-negative breast cancer).


Subject(s)
Acute Kidney Injury/chemically induced , Cisplatin/toxicity , Inflammation/chemically induced , Poly(ADP-ribose) Polymerases/metabolism , Acute Kidney Injury/metabolism , Animals , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...