Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 12(4)2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37107258

ABSTRACT

Acute kidney injury (AKI) often impairs the function of other organs leading to distant organ injury. The liver is the major organ that regulates metabolism and lipid homeostasis in the body. It has been reported that AKI causes liver injury with increased oxidative stress, inflammatory response and steatosis. In the present study, we investigated the mechanisms by which ischemia-reperfusion-induced AKI caused hepatic lipid accumulation. Kidney ischemia (45 min)-reperfusion (24 h) led to a significant increase in plasma creatinine and transaminase in Sprague Dawley rats, indicating kidney and liver injury. Histological and biochemical analyses revealed hepatic lipid accumulation with a significant elevation of triglyceride and cholesterol levels in the liver. This was accompanied by a decreased AMP-activated protein kinase (AMPK) phosphorylation, indicating the reduced activation of AMPK, which is an energy sensor that regulates lipid metabolism. The expression of AMPK-regulated genes that were responsible for fatty acid oxidation (CPTIα, ACOX) was significantly decreased, while the expression of lipogenesis genes (SREPB-1c, ACC1) was significantly elevated. The oxidative stress biomarker malondialdehyde was elevated in the plasma and liver. Incubation of HepG2 cells with an oxidative stress inducer hydrogen peroxide inhibited AMPK phosphorylation and caused cellular lipid accumulation. This was accompanied by decreased expression of genes responsible for fatty acid oxidation and increased expression of genes responsible for lipogenesis. These results suggest that AKI elicits hepatic lipid accumulation through decreased fatty acid metabolism and increased lipogenesis. Oxidative stress may contribute, in part, to the downregulation of the AMPK signaling pathway leading to hepatic lipid accumulation and injury.

2.
J Anim Sci ; 100(11)2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36104002

ABSTRACT

Feed is the most expensive facet of commercial pork production. In order to reduce feed costs, using high-fiber ingredients has become a common practice. Moderate levels of fiber can maintain intestinal physiological function and promote intestinal health. Oxidative stress is linked to impaired nutrient absorption and growth performance. This study investigated the effects of high-fiber (5.26% crude fiber) and low-fiber (2.46% crude fiber) diets on growth performance and intestinal oxidative stress parameters in growing-finishing pigs. Forty growing pigs with initial body weight (27.07 ± 1.26 kg) were randomly assigned to 2 treatment groups with 10 replicates of 2 pigs per pen. Pigs were weighed on day 35, 42, and 70. The feed intake was recorded daily to calculate growth performance parameters. On day 70, eight pigs in each treatment group were randomly selected and euthanized to obtain jejunum to measure oxidative stress status. Pigs fed a high-fiber diet were heavier than those fed a low-fiber diet on days 35, 42, and 70 (P < 0.05). During the whole feeding period, pigs fed a high-fiber diet had a higher average daily gain than those fed a low-fiber diet (P < 0.05). The low-fiber diet resulted in increased levels of malondialdehyde (P < 0.05) in the jejunum, suggesting that the low-fiber diet contributed to oxidative stress in the jejunum. The low-fiber diet also led to a significant increase in glutathione and oxidized glutathione levels (P < 0.05) in the jejunum, indicating that pigs fed a low-fiber diet needed to produce more antioxidant substances to cope with oxidative stress in the intestine. This was accompanied by a significant increase in the expression of glutathione synthesizing enzymes in the jejunum of the low-fiber group (P < 0.05). These results suggest that the high-fiber diet can improve growth performance and maintain intestinal health in growing-finishing pigs by reducing intestinal oxidative stress.


The gastrointestinal tract provides the location for the digestion and absorption of nutrients. It has physical and chemical barriers to protect body from pathogens and toxins. Oxidative stress tends to weaken the physical and chemical barriers of the intestine, which in turn can lead to intestinal dysfunction. Fiber has been suggested to have beneficial effects on the intestine health of pigs. Revealing how fiber can maintain intestinal health is important for pig production. In the present research, we investigated the effect of fiber on the oxidative stress status and antioxidant content in the pig intestine. Our data revealed that a low-fiber diet contributed to oxidative stress in the pig intestine (jejunum). Pigs fed a high-fiber diet had less intestinal oxidative stress and grew heavier. Pigs fed a low-fiber diet may produce more antioxidants to cope with the increased oxidative stress in the intestine.


Subject(s)
Animal Feed , Dietary Fiber , Swine , Animals , Animal Feed/analysis , Dietary Fiber/metabolism , Diet/veterinary , Intestines/physiology , Oxidative Stress , Glutathione/metabolism , Animal Nutritional Physiological Phenomena
3.
Antioxidants (Basel) ; 11(6)2022 May 25.
Article in English | MEDLINE | ID: mdl-35739943

ABSTRACT

Oxidative stress is a major mediator of adverse outcomes in acute kidney injury (AKI). Deficiency of micronutrients, such as folate, is common in AKI. Our previous study reported that AKI impaired kidney reabsorption of folate and decreased plasma folate level in rats. The present study investigated the effect of 5-methyltetrahydrofolate (5-MTHF), a biologically active form of folate/folic acid, on AKI-impaired kidney function and oxidative stress. Sprague-Dawley rats developed AKI after kidney ischemia (45 min) and reperfusion (24 h). Injection of 5-MTHF (3 µg/kg body weight) improved kidney function and attenuated oxidative stress with a restoration of glutathione and a reduction of lipid peroxidation in the kidney. Injection of 5-MTHF activated transcription factor Nrf2 and increased the expression of glutathione synthesizing enzymes, superoxide dismutase-1 and heme oxygenase-1 in the kidney. Simulated ischemia-reperfusion through hypoxia-reoxygenation increased oxidative stress in proximal tubular cells. Incubation of cells with 5-MTHF alleviated cell injury and increased antioxidant enzyme expression and intracellular glutathione levels. Inhibition of Nrf2 expression through siRNA transfection abolished the effect of 5-MTHF against oxidative stress. These results suggest that low-dose folic acid can improve kidney function through activation of Nrf2 and restoration of antioxidant defence. Micronutrient supplements may improve clinical outcomes in AKI.

4.
Antioxidants (Basel) ; 11(3)2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35326122

ABSTRACT

Impaired hepatic lipid metabolism is a hallmark of non-alcoholic fatty liver disease (NAFLD), which has no effective treatment option. Recently, Notch signaling has been identified as an important mediator of hepatic lipid metabolism. Lingonberry (Vaccinium vitis-idaea L.) is an anthocyanin-rich fruit with significant lipid-lowering properties. In this study, we examined how lingonberry influenced Notch signaling and fatty acid metabolism in a mouse model of NAFLD. Mice (C57BL/6J) fed a high-fat diet (HFD) for 12 weeks developed fatty liver and activated hepatic Notch1 signaling. Lingonberry supplementation inhibited hepatic Notch1 signaling and improved lipid profile by improving the expression of the genes involved in hepatic lipid metabolism. The results were verified using a palmitic-acid-challenged cell model. Similar to the animal data, palmitic acid impaired cellular lipid metabolism and induced Notch1 in HepG2 cells. Lingonberry extract or cyanidin-3-glucoside attenuated Notch1 signaling and decreased intracellular triglyceride accumulation. The inhibition of Notch in the hepatocytes attenuated sterol-regulatory-element-binding-transcription-factor-1 (SREBP-1c)-mediated lipogenesis and increased the expression of carnitine palmitoyltransferase-I-alpha (CPTIα) and acyl-CoA oxidase1 (ACOX1). Taken together, lingonberry's hepatoprotective effect is mediated by, in part, improving hepatic lipid metabolism via inhibiting Notch1 signaling in HFD-induced fatty liver.

5.
Front Immunol ; 12: 678914, 2021.
Article in English | MEDLINE | ID: mdl-34149715

ABSTRACT

Acute or chronic kidney disease can cause micronutrient deficiency. Patients with end-stage renal disease, kidney transplantation or on dialysis have reduced circulating levels of folate, an essential B vitamin. However, the molecular mechanism is not well understood. Reabsorption of folate in renal proximal tubules through folate transporters is an important process to prevent urinary loss of folate. The present study investigated the impact of acute kidney injury (AKI) on folate transporter expression and the underlying mechanism. AKI was induced in Sprague-Dawley rats that were subjected to kidney ischemia (45 min)-reperfusion (24 h). Both male and female rats displayed kidney injury and low plasma folate levels compared with sham-operated rats. The plasma folate levels were inversely correlated to plasma creatinine levels. There was a significant increase in neutrophil gelatinase-associated lipocalin (NGAL) and IL-6 mRNA expression in the kidneys of rats with ischemia-reperfusion, indicating kidney injury and increased inflammatory cytokine expression. Ischemia-reperfusion decreased mRNA and protein expression of folate transporters including folate receptor 1 (FOLR1) and reduced folate carrier (RFC); and inhibited transcription factor Sp1/DNA binding activity in the kidneys. Simulated ischemia-reperfusion through hypoxia-reoxygenation or Sp1 siRNA transfection in human proximal tubular cells inhibited folate transporter expression and reduced intracellular folate levels. These results suggest that ischemia-reperfusion injury downregulates renal folate transporter expression and decreases folate uptake by tubular cells, which may contribute to low folate status in AKI. In conclusion, ischemia-reperfusion injury can downregulate Sp1 mediated-folate transporter expression in tubular cells, which may reduce folate reabsorption and lead to low folate status.


Subject(s)
Folic Acid Transporters/genetics , Folic Acid/blood , Kidney Diseases/etiology , Kidney Diseases/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Animals , Biomarkers , Disease Models, Animal , Female , Folic Acid Transporters/metabolism , Gene Expression Regulation , Immunohistochemistry , Kidney Diseases/pathology , Kidney Function Tests , Kidney Tubules, Proximal/metabolism , Rats , Reperfusion Injury/pathology
6.
Am J Physiol Renal Physiol ; 300(1): F189-98, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980407

ABSTRACT

Hyperhomocysteinemia, a condition of elevated blood homocysteine (Hcy) levels, is a metabolic disease. It is a common clinical finding in patients with chronic kidney diseases and occurs almost uniformly in patients with end-stage renal disease. Hyperhomocysteinemia is also a risk factor for cardiovascular disease. Our recent studies indicate that hyperhomocysteinemia can lead to renal injury by inducing oxidative stress. Oxidative stress is one of the important mechanisms contributing to Hcy-induced tissue injury. Folic acid supplementation is regarded as a promising approach for prevention and treatment of cardiovascular disease associated with hyperhomocysteinemia due to its Hcy-lowering effect. However, its effect on the kidney is not clear. The aim of this study was to examine the effect of folic acid supplementation on Hcy-induced superoxide anion production via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in the kidney during hyperhomocysteinemia. Hyperhomocysteinemia was induced in male Sprague-Dawley rats fed a high-methionine diet for 12 wk with or without folic acid supplementation. A group of rats fed a regular diet was used as control. There was a significant increase in levels of superoxide anions and lipid peroxides in kidneys isolated from hyperhomocysteinemic rats. Activation of NADPH oxidase was responsible for hyperhomocysteinemia-induced oxidative stress in the kidney. Folic acid supplementation effectively antagonized hyperhomocysteinemia-induced oxidative stress via its Hcy-lowering and Hcy-independent effect. In vitro study also showed that 5-methyltetrahydrofolate, an active form of folate, effectively reduced Hcy-induced superoxide anion production via NADPH oxidase. Xanthine oxidase activity was increased and superoxide dismutase (SOD) activity was decreased in the kidney of hyperhomocysteinemic rats, which might also contribute to an elevation of superoxide anion level in the kidney. Folic acid supplementation attenuated xanthine oxidase activity and restored SOD activity in the kidney of hyperhomocysteinemic rats. These results suggest that folic acid supplementation may offer renal protective effect against oxidative stress.


Subject(s)
Folic Acid/therapeutic use , Hyperhomocysteinemia/complications , Kidney/drug effects , Kidney/metabolism , NADPH Oxidases/antagonists & inhibitors , Superoxides/metabolism , Acetophenones/pharmacology , Animals , Cells, Cultured , Humans , Hyperhomocysteinemia/chemically induced , Male , Methionine/administration & dosage , Oxidative Stress , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Xanthine Oxidase/metabolism
7.
Am J Physiol Renal Physiol ; 294(1): F236-44, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17977907

ABSTRACT

Hyperhomocysteinemia, or an elevation of blood homocysteine (Hcy) levels, is associated with cardiovascular disorders. Although kidney dysfunction is an important risk factor causing hyperhomocysteinemia, the direct effect of Hcy on the kidney is not well documented. There is a positive association between an elevation of blood Hcy levels and the development of chronic kidney disease. Inflammatory response such as increased chemokine expression has been implicated as one of the mechanisms for renal disease. Monocyte chemoattractant protein-1 (MCP-1) is a potent chemokine that is involved in the inflammatory response in renal disease. Nuclear factor-kappaB (NF-kappaB) plays an important role in upregulation of MCP-1 expression. We investigated the effect of hyperhomocysteinemia on MCP-1 expression and the molecular mechanism underlining such an effect in rat kidneys as well as in proximal tubular cells. Hyperhomocysteinemia was induced in rats fed a high-methionine diet for 12 wk. The MCP-1 mRNA expression and MCP-1 protein levels were significantly increased in kidneys isolated from hyperhomocysteinemic rats. The NF-kappaB activity was significantly increased in the same kidneys. Pretreatment of hyperhomocysteinemic rats with a NF-kappaB inhibitor abolished hyperhomocysteinemia-induced MCP-1 expression in the kidney. To confirm the causative role of NF-kappaB activation in MCP-1 expression, human kidney proximal tubular cells were transfected with decoy NF-kappaB oligodeoxynucleotide to inhibit NF-kappaB activation. Such a treatment prevented Hcy-induced MCP-1 mRNA expression in tubular cells. Our results suggest that hyperhomocysteinemia stimulates MCP-1 expression in the kidney via NF-kappaB activation. Such an inflammatory response may contribute to renal injury associated with hyperhomocysteinemia.


Subject(s)
Chemokine CCL2/metabolism , Homocysteine/metabolism , Kidney Tubules, Proximal/metabolism , Kidney/metabolism , NF-kappa B/metabolism , Animals , Cells, Cultured , Chemokine CCL2/genetics , Homocysteine/pharmacology , Humans , Hyperhomocysteinemia/chemically induced , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Kidney/pathology , Kidney Tubules, Proximal/pathology , Male , Methionine , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
8.
Can J Physiol Pharmacol ; 85(11): 1053-62, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18066107

ABSTRACT

The activation of c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in the induction of cell apoptosis. We previously reported that magnesium tanshinoate B (MTB), a compound purified from a Chinese herb danshen (Salvia miltiorrhiza), could inhibit ischemia/reperfusion-induced myocyte apoptosis in the heart. The objective of the present study was to investigate whether MTB can prevent oxidized lipoprotein-induced apoptosis in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were incubated with copper-oxidized very low density lipoprotein (Cu-OxVLDL) or copper-oxidized low density lipoprotein (Cu-OxLDL). Treatment of cells with Cu-OxVLDL or Cu-OxLDL resulted in a 3-fold increase in the JNK activity. The amount of cytochrome c released and the activity of caspase-3 in cells treated with Cu-OxVLDL or Cu-OxLDL were significantly elevated, indicating the occurrence of apoptosis. The presence of MTB was able to abolish the JNK activation, cytochrome c release, and caspase-3 activation induced by Cu-OxVLDL or Cu-OxLDL, resulting in a marked reduction in apoptosis in endothelial cells. The data from this study indicate that oxidized lipoproteins induce apoptosis in endothelial cells. We postulate that the inhibition of the JNK signaling pathway by MTB is a key mechanism that protects these cells from oxidized lipoprotein-induced apoptosis.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Lipoproteins, LDL/toxicity , Magnesium/pharmacology , Phenanthrolines/pharmacology , Caspase 3/metabolism , Cells, Cultured , Cytochromes c/metabolism , Endothelial Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism
9.
Can J Physiol Pharmacol ; 84(1): 141-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16845898

ABSTRACT

Folic acid supplementation is a promising approach for patients with cardiovascular diseases associated with hyperhomocysteinemia. We have demonstrated that homocysteine (Hcy) activates nuclear factor-kappaB (NF-kappaB), a transcription factor that plays an important role in inflammatory responses. The aim of the present study was to investigate the effect of folic acid on Hcy-induced NF-kappaB activation in macrophages. Hcy treatment (100 micromol/L) resulted in NF-kappaB activation and increased monocyte chemoattractant protein-1 (MCP-1) expression in THP-1 derived macrophages. Hcy-induced NF-kappaB activation was associated with a significant increase in the intracellular superoxide anion levels. There was a significant increase in phosphorylation and membrane translocation of NADPH oxidase p47phox subunit in Hcy-treated cells. Addition of folic acid (200 ng/mL) to the culture medium abolished NADPH oxidase-dependent superoxide anion generation in macrophages by preventing phosphorylation of p47phox subunit. Consequently, Hcy-induced NF-kappaB activation and MCP-1 expression was inhibited. Such an inhibitory effect of folic acid was independent of its Hcy-lowering ability. Taken together, these results suggest that folic acid treatment can effectively inhibit Hcy-induced oxidative stress and inflammatory responses in macrophages. This may represent one of the mechanisms by which folic acid supplementation exerts a protective effect in cardiovascular disorders.


Subject(s)
Chemokine CCL2/metabolism , Folic Acid/pharmacology , Macrophages/metabolism , NF-kappa B/antagonists & inhibitors , Superoxides/antagonists & inhibitors , Cell Line , Chemokine CCL2/genetics , Gene Expression Regulation/drug effects , Homocysteine/pharmacology , Humans , NADPH Oxidases/metabolism , NF-kappa B/metabolism , RNA, Messenger/metabolism , Superoxides/metabolism
10.
Biochem J ; 398(1): 73-82, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16626305

ABSTRACT

Hyperhomocysteinaemia is an independent risk factor for cardiovascular diseases due to atherosclerosis. The development of atherosclerosis involves reactive oxygen species-induced oxidative stress in vascular cells. Our previous study [Wang and O (2001) Biochem. J. 357, 233-240] demonstrated that Hcy (homocysteine) treatment caused a significant elevation of intracellular superoxide anion, leading to increased expression of chemokine receptor in monocytes. NADPH oxidase is primarily responsible for superoxide anion production in monocytes. In the present study, we investigated the molecular mechanism of Hcy-induced superoxide anion production in monocytes. Hcy treatment (20-100 microM) caused an activation of NADPH oxidase and an increase in the superoxide anion level in monocytes (THP-1, a human monocytic cell line). Transfection of cells with p47phox siRNA (small interfering RNA) abolished Hcy-induced superoxide anion production, indicating the involvement of NADPH oxidase. Hcy treatment resulted in phosphorylation and subsequently membrane translocation of p47phox and p67phox subunits leading to NADPH oxidase activation. Pretreatment of cells with PKC (protein kinase C) inhibitors Ro-32-0432 (bisindolylmaleimide XI hydrochloride) (selective for PKCalpha, PKCbeta and PKCgamma) abolished Hcy-induced phosphorylation of p47phox and p67phox subunits in monocytes. Transfection of cells with antisense PKCbeta oligonucleotide, but not antisense PKCalpha oligonucleotide, completely blocked Hcy-induced phosphorylation of p47phox and p67phox subunits as well as superoxide anion production. Pretreatment of cells with LY333531, a PKCbeta inhibitor, abolished Hcy-induced superoxide anion production. Taken together, these results indicate that Hcy-stimulated superoxide anion production in monocytes is regulated through PKC-dependent phosphorylation of p47phox and p67phox subunits of NADPH oxidase. Increased superoxide anion production via NADPH oxidase may play an important role in Hcy-induced inflammatory response during atherogenesis.


Subject(s)
Homocysteine/pharmacology , Monocytes/drug effects , Monocytes/enzymology , NADPH Oxidases/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Protein Subunits/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Humans , NADPH Oxidases/genetics , Oligonucleotides, Antisense/metabolism , Phosphoproteins/genetics , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Subunits/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Superoxides/metabolism
11.
Can J Physiol Pharmacol ; 83(4): 321-34, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15877107

ABSTRACT

Traditional Chinese medicine (TCM) has many beneficial effects and has been practiced for several thousand years. It is known to treat the cause of a disease rather than to alleviate its symptoms. Based on a belief that TCM is natural, safe, and of lower cost, consumers worldwide are spending more out-of-pocket money on this form of therapy. This increased spending, and reports of adverse reactions, has drawn the attention of many regulatory agencies. Scientists have called for more evidence-based and scientific research on the risks and benefits of TCM. In Canada, the Natural Health Product Regulations came into effect January 2004. TCM herbal product manufacturers will need to provide products of reputable quality to the market. Many will apply modern technology and good science to support their products. The issues facing producers, scientists, and consumers alike are quality control and assessment, standardization of bioactive components, mechanisms of actions, and integration of the evolved modern Chinese medicine into the healthcare system. Solid science, better regulation of the final product, and better education of consumers are necessary to extract the best of TCM to complement existing conventional medicine to deliver the best healthcare.


Subject(s)
Medicine, Chinese Traditional/trends , Animals , Chromatography , History, Ancient , Humans , Medicine, Chinese Traditional/history , Plants, Medicinal/chemistry , Quality Control
12.
Circ Res ; 94(1): 28-36, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14630727

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for cardiovascular diseases. Our previous studies demonstrated an important interaction between nuclear factor-kappaB (NF-kappaB) activation and homocysteine (Hcy)-induced chemokine expression in vascular smooth muscle cells and macrophages. The objective of the present study was to investigate the in vivo effect of hyperhomocysteinemia on NF-kappaB activation and the underlying mechanism of Hcy-induced NF-kappaB activation in endothelial cells. Hyperhomocysteinemia was induced in Sprague-Dawley rats after 4 weeks of a high-methionine diet. The activated form of NF-kappaB and increased level of superoxide anions were detected in the endothelium of aortas isolated from hyperhomocysteinemic rats. The underlying mechanism of Hcy-induced NF-kappaB activation was investigated in human umbilical cord vein endothelial cells and in human aortic endothelial cells. Incubation of cells with Hcy (100 micromol/L) activated IkappaB kinases (IKKalpha and IKKbeta), leading to phosphorylation and subsequent degradation of IkappaBalpha. As a consequence, NF-kappaB nuclear translocation, enhanced NF-kappaB/DNA binding activity, and increased transcriptional activity occurred. Additional analysis revealed a marked elevation of superoxide anion levels in Hcy-treated cells. Treatment of cells with a superoxide anion scavenger (polyethylene glycol-superoxide dismutase) or IkappaB kinase inhibitor (prostaglandin A(1)) could prevent Hcy-induced activation of IKK kinases and NF-kappaB in endothelial cells. In conclusion, these results suggest that Hcy-induced superoxide anion production may play a potential role for NF-kappaB activation in the early stages of atherosclerosis in the vascular wall via activation of IkappaB kinases.


Subject(s)
Endothelium, Vascular/metabolism , Hyperhomocysteinemia/metabolism , NF-kappa B/metabolism , Oxidative Stress , Animals , Aorta/chemistry , Aorta/cytology , Cells, Cultured , Endothelium, Vascular/drug effects , Homocysteine/pharmacology , I-kappa B Kinase , I-kappa B Proteins/metabolism , Male , NF-KappaB Inhibitor alpha , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Superoxides/analysis , Superoxides/metabolism
13.
Kidney Int ; 62(4): 1160-70, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12234286

ABSTRACT

BACKGROUND: Renal ischemia/reperfusion injury is a major cause of acute renal failure in both native kidneys and renal allografts. One important feature of such injury is monocyte/macrophage infiltration into the renal tissue. The infiltration of monocytes/macrophages can be induced by chemotactic factors produced by renal cells. Monocyte chemoattractant protein-1 (MCP-1) is a potent chemoattractant protein for monocyte recruitment. The objective of the present study was to investigate mechanisms of elevated MCP-1 expression in rat kidney during ischemia/reperfusion injury. METHODS: The left kidney was subjected to one hour of ischemia followed by reperfusion for various time periods. The expression of MCP-1 mRNA was determined by nuclease protection assay and MCP-1 protein was identified by immunohistochemistry. Activation of a nuclear factor-kappa B (NF-kappaB) was determined by electrophoretic mobility shift assay and the level of lipid peroxides in the kidney was measured. RESULTS: There was a significant increase in MCP-1 expression in the ischemia/reperfusion kidney 2 hours after reperfusion (210% of the control). This increase was accompanied by activation of NF-kappaB, suggesting that this transcription factor might be involved in the event. The number of monocytes was significantly elevated in the kidney 3 days after ischemia/reperfusion. Pretreatment of rats with NF-kappaB inhibitors not only prevented NF-kappaB activation induced by ischemia/reperfusion, but also inhibited MCP-1 mRNA expression. Further analysis revealed that oxidative stress and increased IkappaB-alpha phosphorylation might be an underlying mechanism for NF-kappaB activation and subsequent MCP-1 mRNA expression in the ischemia/reperfusion kidney. CONCLUSION: The present study clearly demonstrates that enhanced MCP-1 expression in rat kidney during ischemia/reperfusion injury is mediated by NF-kappaB activation and oxidative stress. Elevated MCP-1 expression might be responsible for increased monocyte infiltration in the injured kidney.


Subject(s)
Chemokine CCL2/genetics , NF-kappa B/metabolism , Oxidative Stress/physiology , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Animals , Gene Expression/physiology , I-kappa B Proteins/genetics , Kidney/cytology , Kidney/metabolism , Lipid Peroxidation/physiology , Male , Monocytes/cytology , NF-KappaB Inhibitor alpha , Neutrophils/cytology , Rats , Rats, Sprague-Dawley
14.
Clin Exp Pharmacol Physiol ; 29(9): 852-4, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12165055

ABSTRACT

1. Renal ischaemia followed by reperfusion leads to acute renal failure in both native kidneys and renal allografts. Cyclosporine A (CsA) has been used as an immunosuppressive agent in organ transplantation. In the present study, the effect of CsA on ischaemia/reperfusion-induced apoptosis in the kidney was investigated. 2. Ischaemia/reperfusion injury caused widespread apoptosis primarily in the medulla of the kidney. At 1.5 mg/kg per day, CsA significantly reduced the number of apoptotic cells in rat kidney after ischaemia/reperfusion injury. 3. Low-dose CsA treatment did not affect the levels of creatinine in the serum of rats after ischaemia/reperfusion injury.


Subject(s)
Apoptosis/drug effects , Cyclosporine/therapeutic use , Ischemia/drug therapy , Kidney/blood supply , Reperfusion Injury/drug therapy , Animals , Cyclosporine/pharmacology , Ischemia/pathology , Kidney/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...