Hostname: page-component-7c8c6479df-7qhmt Total loading time: 0 Render date: 2024-03-29T14:45:49.724Z Has data issue: false hasContentIssue false

Beneficial effect of genistein on lowering blood pressure and kidney toxicity in fructose-fed hypertensive rats

Published online by Cambridge University Press:  21 September 2012

Nallasamy Palanisamy*
Affiliation:
Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India
Anuradha C. Venkataraman
Affiliation:
Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India
*
*Corresponding author: Dr N. Palanisamy, fax +91 4144 238080, email nalpybiochem@gmail.com
Rights & Permissions [Opens in a new window]

Abstract

The study evaluates the effects of genistein on blood pressure (BP) and ultrastructural changes in kidney of fructose-fed hypertensive rats. Male Wistar rats were fed a diet containing 60 % starch or 60 % fructose as the source of carbohydrate. After 15 d, rats in each dietary group were divided into two groups and were treated with either genistein (1 mg/kg per d) in dimethylsulfoxide (DMSO) or 30 % DMSO alone. BP, pressor mechanisms, protein kinase C-βII (PKC-βII) expression, endothelial NO synthase (eNOS) expression and renal ultrastructural changes were evaluated after 60 d. Fructose-fed rats displayed significant elevation in BP and heart rate. Significant increase in plasma angiotensin-converting enzyme (ACE) activity, alterations in renal lipid profile, nitrite and kallikrein activity, enhanced expression of membrane-associated PKC-βII and decreased expression of eNOS were observed in them. Histology and electron microscopic studies showed structural changes in the kidney. Genistein administration lowered BP, restored ACE, PKC-βII and eNOS expression and preserved renal ultrastructural integrity. These findings demonstrate that genistein has effects on eNOS activity in renal cells, leading to eNOS activation and NO synthesis. These effects could have been mediated by activation of PKC-βII. The observed benefits of genistein make it a promising candidate for therapy of diabetic kidney disease.

Type
Full Papers
Copyright
Copyright © The Authors 2012 

The fructose-fed rat mimics the human metabolic syndrome in which a constellation of abnormalities such as glucose intolerance, insulin resistance, dyslipidaemia, hyperinsulinaemia and hypertension are described(Reference Reaven1). These risk factors predispose to type 2 diabetes, CVD and renal disease. Renal hypertrophy, arteriolopathy, glomerular hypertension, cortical vasoconstriction and oxidative damage are the pathological findings observed in fructose-fed rats(Reference Sanchez-Lozada, Tapia and Jimenez2). The rise in blood pressure (BP) in this model has been related to defects in vasodilatory mechanisms such as impaired NO production due to decreased endothelial NO synthase (eNOS) activity, activation of selective isoforms of protein kinase C (PKC) and down-regulation of the renal kallikrein–kinin system(Reference Haller, Baur and Quass3Reference Nishimoto, Tomida and Matsui5)

Genistein, a phyto-esterogen with isoflavone structure, is found in a wide variety of plant-derived foods, in particular in soyabean. Genistein exerts an anti-diabetic effect by improving glucose and lipid metabolism in type 2 diabetic subjects(Reference Park, Choi and Cho6). A study has demonstrated the beneficial effects of soya isoflavones on kidney damage induced by nephrotoxins(Reference Kojima, Uesugi and Toda7). Interestingly, we found that genistein improves insulin sensitivity and restores renal function in fructose-fed rats(Reference Palanisamy, Viswanathan and Anuradha8). However, there is a lack of data that directly relate the improvement of BP and renoprotective effect of genistein in the insulin-resistant state. The present study was therefore designed to investigate the effects of genistein on BP, related pressor mechanisms like plasma angiotensin-converting enzyme (ACE), NO and kallikrein and the expression of eNOS and PKC-βII in fructose-fed hypertensive rats. In addition, the effects of genistein on lipid profile and kidney structural changes were studied.

Methods

Chemicals

Anti-PKC-βII antibody was purchased from Sigma Chemicals, while anti-eNOS (Sigma) antibody was obtained as a generous gift from Dr Z. Serfozo, Department of Experimental Zoology, Hungarian Academy of Sciences, Tihany, Hungary. Genistein (Sigma) was obtained as a generous gift from Dr T. Szkudelski, Department of Animal Physiology and Biochemistry, August Cieszkowski University of Agriculture, Poland. All chemicals and solvents used for the study were purchased from SISCO Research Laboratories Private Limited.

Animals

Adult male Wistar rats of body weight 150–160 g were obtained from the Central Animal House, Rajah Muthiah Medical College and Hospital (RMMCH), Annamalai University. They were housed in a well-ventilated animal room under controlled conditions on a 12 h light–12 h dark cycle. Animals received the standard pellet diet (Karnataka State Agro Corporation Limited, Agro Feeds Division) and water ad libitum. The experimental procedures were done according to the guidelines of the Committee for the Purpose of Control and Supervision of Experiments on Animals and were approved by the Institutional Ethical Committee of Animal Care, RMMCH, Annamalai University, Annamalai Nagar, India (no. 160/1999/CPCSEA/425).

Experimental design

After the acclimatisation period of 7 d, the rats were divided into four groups of twelve rats each as follows: (1) Group 1 rats were fed the control diet containing 60 % maize starch, 20 % protein, 0·7 % methionine, 5 % ground nut oil, 10·6 % wheat bran, 3·5 % salt mixture and 0·2 % vitamin mixture and given the vehicle (0·5 ml 30 % dimethylsulfoxide/d) by oral administration for 60 d, (2) Group 2 rats were fed the high-fructose diet, which had similar composition to that of the control diet, except that starch was replaced by fructose, and administered dimethylsulfoxide for 60 d, as described for rats in Group 1; (3) Group 3 rats were fed the high-fructose diet and administered genistein (1 mg/kg per d in 0·5 ml dimethylsulfoxide) from day 16 until day 60 by oral administration and (4) Group 4 rats were fed the control diet and administered genistein from the 16th day till the 60th day of the experimental period, as described for Group 3.

On day 60, the animals (n 6) were kept in individual metabolism cages, and 24 h urine samples were collected in sealed beakers with added preservative and killed by cervical dislocation after administering anaesthesia (ketamine hydrochloride 30 mg/kg, intramuscular). The abdomen was cut open and kidneys were dissected, washed in ice-cold saline and weighed. Portions of kidney were cut and homogenised in 0·1-m Tris–HCl buffer (pH 7·4) and used for the assays.

Blood pressure measurement

BP was measured in conscious rats (n 6) by the indirect tail-cuff method using a fully automatic BP analyser (Doc-NIBP200A, BIO PAC Systems). The rats were kept in a constant temperature (32°C) chamber for 30 min and then placed in a rat holder. A total of eight BP measurements were carried out in each animal. An average of six such readings was taken as the individual (systolic/diastolic) BP, neglecting the maximal and minimal values. The sum of the diastolic pressure and one-third of the pulse pressure was taken as the mean arterial pressure.

Biochemical analysis

Total nitrite (nitrate+nitrite) as a measure of NO∙(Reference Rock, Astier and Lab9), plasma ACE activity(Reference Holmquist, Bünning and Riordan10) and the activity of kallikrein(Reference Trautschold, Werle and Schweitzer11) were measured. Plasma and renal tissue cholesterol, TAG and NEFA were measured by methods outlined elsewhere(Reference Raghuramulu, Madhavan Nair and Kalyanasundaram12).

Immunoblot analysis of protein kinase C-βII and endothelial nitric oxide synthase

The cytosol and membrane fractions of the kidney were prepared as follows: 500 mg of kidney were homogenised in 5 ml of ice-cold extraction buffer (40 mm-sucrose, 50 mm-HEPES, 280 mm-NaCl and 20 mm-NaOH and protease inhibitor cocktail; Sigma Chemical Company). The homogenate was centrifuged at 1000 g for 5 min at 4°C. The supernatant obtained was used as the total cell lysate fraction. The total cell lysate was centrifuged again at 100 000 g for 60 min at 4°C. The supernatant obtained was used as the cytosolic fraction, while the pellet was resuspended in ice-cold lysis buffer (50 mm-HEPES, 480 mm-NaCl, 20 mm-NaOH and 2 % SDS) with protease inhibitor cocktail and centrifuged at 100 000 g for 60 min at 4°C. The pellet was discarded and the supernatant was used as the membrane fraction. Protein concentration was measured by the method of Lowry et al. (Reference Lowry, Rosebrough and Farr13). The sample (100 μg protein) was solubilised in 2 ×  Laemmli sample buffer. The total cell lysate (for eNOS expression) and the cytoplasmic and membrane proteins (for PKC-βII expression) were subjected to electrophoresis in SDS-PAGE gel (6 % for eNOS and 8 % for PKC-βII) and blotted onto nitrocellulose membranes in a transferring buffer at 80 mA in a transfer apparatus for 1 h and 30 min. The membranes were then pre-incubated in the blocking buffer (PBS containing 0·05 % Tween-20 (PBS-T)) and 5 % non-fat dried milk for 2 h at room temperature and then probed with rabbit polyclonal anti-eNOS primary antibody (1:750 dilution) or mouse monoclonal anti-PKC-βII primary antibody (1:1000 dilution) overnight at 4°C and washed with PBS-T thrice and incubated with either goat anti-rabbit secondary antibody (1:4000) or goat anti-mouse secondary antibody (1:5000 dilution) (Genei) for 2 h and again washed with PBS-T thrice. Antigen–antibody complexes were detected using a chemiluminescence kit (Thermo Fisher Scientific, Inc.) and quantified by Image J, a public domain Java image processing software (Wayne Rasband, National Institutes of Health). β-Actin was used as the house keeping internal control. The data for test proteins are normalised with corresponding β-actin values and are presented as arbitrary units.

Histology

A portion of kidney tissue removed from each group was fixed in 8 % neutral formalin, dried and embedded in paraffin wax. Sections of 3–5 mm were cut, processed and stained with periodic acid–Schiff stain. The slides were examined under a light microscope.

Transmission electron microscopic examination of kidney

A portion of renal tissue was cut into small pieces and pre-fixed in 2·5 % glutaraldehyde (0·2 mol/l cacodylate buffer, pH 7·4) for 4 h and then post-fixed in 1 % buffered osmium tetroxide for 1 h and embedded in epoxyresin. Ultra-thin sections were stained with uranyl acetate and lead citrate, examined and photographed under a Philips-P201 transmission electron microscope (Koninklijke Philips Electronics).

Statistical analysis

Values are expressed as means and standard deviations. Data within the groups were analysed using one-way ANOVA followed by Duncan's multiple range test. A value of P< 0·05 was considered statistically significant.

Results

Blood pressure, nitrite, kallikrein and angiotensin-converting enzyme

Table 1 presents body and kidney weight, systolic and diastolic BP, mean arterial pressure, heart rate, kallikrein, ACE and total nitrite values. Systolic and diastolic BP, mean arterial pressure, heart rate and ACE activity were significantly (P< 0·05) increased in fructose-fed rats. Kallikrein activity was lower by 21 % and total nitrite by 38 % in fructose-fed rats compared to control rats. Supplementation of genistein restored the levels, which were not significantly different from control rats.

Table 1 Body and kidney weight, systolic and diastolic blood pressure, mean arterial pressure (MAP), heart rate, angiotensin-converting enzyme (ACE), kallikrein and nitrite in plasma and urine of experimental rats (Mean values and standard deviations of six animals)

CON, control rats; FRU, fructose-fed rats; FRU+GEN, fructose+genistein (1 mg/kg body weight per d); CON+GEN, control+genistein (1 mg/kg body weight per d).

* Mean values were significantly different compared to control rats (P< 0·05, Duncan's multiple range test).

Mean values were significantly different compared to fructose-fed rats (P< 0·05, Duncan's multiple range test).

Plasma and kidney lipid content

Fig. 1(a) and (b) represent the levels of plasma and kidney lipids, respectively, in experimental animals. Significant increases were observed in plasma cholesterol, TAG and NEFA in the fructose-fed rats (by 29, 40 and 35 %, respectively). Genistein administration resulted in significant reductions in cholesterol, TAG and NEFA levels in fructose-fed rats. Similarly, the levels of cholesterol, NEFA and TAG were significantly higher in the kidney of fructose-fed rats than in those fed the control diet (Fig. 1(b)). Genistein treatment of fructose-fed rats restored the levels of kidney lipids to near-normal values.

Fig. 1 (a) Plasma and (b) kidney lipid profile of experimental animals. CHOL, cholesterol. (a) , Control rats (CON); , fructose-fed rats (FRU); , fructose+genistein (1 mg/kg body weight per d) (FRU+GEN); , control+genistein (1 mg/kg body weight per d) (CON+GEN). (b) , CON; , FRU; , FRU+GEN; , CON+GEN. * Mean values were significantly different compared to CON (P< 0·05; ANOVA followed by Duncan's multiple range test). † Mean values were significantly different compared to FRU (P< 0·05; ANOVA followed by Duncan's multiple range test).

Protein kinase C-βII and endothelial nitric oxide synthase expression

Fig. 2(a) and (b) depict the expression of PKC-βII in the cytosolic and membrane fractions, respectively, and give the densitometry data of the respective blots. Significant increase in PKC-βII expression was observed in both the fractions in the fructose-fed group compared to the control group. Expression was significantly reduced in the fructose-fed genistein-treated group, as compared to the genistein untreated fructose-fed group.

Fig. 2 Representative immunoblot of protein kinase C-βII (PKC-βII) isoenzyme in kidney (a) cytosol and (b) membrane and representative densitometry analysis of Western blots a and b, respectively. Values are expressed as relative intensity and means of six independent experiments, and standard deviations represented by vertical bars. CON, control rats; FRU, fructose-fed rats; FRU+GEN, fructose+genistein (1 mg/kg body weight per d); CON+GEN, control+genistein (1 mg/kg body weight per d). * Values were significantly different compared to CON (P< 0·05; ANOVA followed by Duncan's multiple range test). † Values were significantly different compared to FRU (P< 0·05; ANOVA followed by Duncan's multiple range test). AU, arbitrary units.

Fig. 3 depicts a representative immunoblot showing eNOS expression in the kidney of experimental rats, together with a bar plot giving the results of densitometry analysis of the protein bands. The eNOS expression was significantly lower in the fructose-fed group, as compared to the control group. Protein expression was similar in all other groups.

Fig. 3 Representative immunoblot of endothelial nitric oxide synthase (eNOS) in kidney cell lysate and representative densitometry analysis of the bands. Values are expressed as relative intensity and means of six independent experiments, and standard deviations represented by vertical bars. CON, control rats; FRU, fructose-fed rats; FRU+GEN, fructose+genistein (1 mg/kg body weight per d); CON+GEN, control+genistein (1 mg/kg body weight per d). * Mean values were significantly different compared to CON (P< 0·05; ANOVA followed by Duncan's multiple range test). † Mean values were significantly different compared to FRU (P< 0·05; ANOVA followed by Duncan's multiple range test). AU, arbitrary units.

Structural studies

Fig. 4(a)–(d) represents the kidney sections of animals analysed by periodic acid–Schiff staining (40 ×  magnification). Under a light microscope, kidneys from fructose-fed animals showed glomerular hypertrophy, glomerular sclerosis and interstitial damage (Fig. 4(b)). Kidney sections from fructose+genistein-treated animals showed reduced pathological changes (Fig. 4(c)) compared to those observed in fructose-treated animals. Kidney sections from the control group and the control group treated with genistein showed normal glomerular and tubular histology (Fig. 4(a) and (d)).

Fig. 4 Representative micrographs of kidney tissue stained with periodic acid–Schiff from (a) control rats, (b) fructose-fed rats, (c) fructose+genistein-treated rats (1 mg/kg body weight per d) and (d) control+genistein-treated rats (1 mg/kg body weight per d). (a–d) Magnification 400 × . (A colour version of this figure can be found online at http://www.journals.cambridge.org/bjn).

Fig. 5 presents the transmission electron microscopic images showing renal architecture and podocyte foot processes of experimental animals. Podocyte and basement membrane from control (Fig. 5(a)) and control plus genistein (Fig. 5(d))-treated groups show similar architecture. Fructose-fed rat kidney shows extensive disruption of podocytes and effacement. Arrows show basement membrane thickening and the presence of excess matrix proteins (Fig. 5(b)). Treatment with genistein significantly prevented these pathological structural changes (Fig. 5(c)). Podocytes and basement membrane appear normal in genistein-treated control animals.

Fig. 5 (a–d) Transmission electron microscopic examination of rat kidney ( × 10 000). (a) An electron micrograph from a control rat. Regularly arranged podocyte foot processes with normal basement membrane are observed. (b) An electron micrograph from a fructose-fed rat. Abnormal foot processes effacement, podocyte appear broadened, basement membrane thickening and extensive deposition of matrix proteins (blue arrows) are seen. (c) An electron micrograph from a genistein-treated fructose-fed rat shows basement membrane within normal limits; some podocyte foot processes appear broadened. (d) Electron micrograph from a genistein-treated control rat shows normal structure of podocytes and basement membrane.

Discussion

The fructose-fed rat is a well established model of the metabolic syndrome associated with renal dysfunction and hypertension(Reference Vasdev, Gill and Parai14). In the present study, fructose feeding caused deleterious effects on pressure-regulatory mechanisms and kidney structure. Several processes that contribute to a rise in BP such as sympathetic nervous system overactivation, increased production of vasoconstrictors, defects in NO and increased formation of reactive oxygen species are reported in fructose-fed rats. It has been suggested that elevation of BP in this model is a sequence of hyperinsulinaemia, as insulin induces inappropriate Na retention, activation of the sympathetic nervous system and the renin–angiotensin system(Reference Bhanot, Salh and Verma15, Reference Vasdev, Longerich and Gill16).

ACE catalyses the formation of angiotensin II, the aldosterone-stimulating peptide, from angiotensin I. A previous study has shown that genistein decreased ACE expression dose-dependently in rat aortic endothelial cells via oestrogen receptors(Reference Xu, Yang and Li17). The antihypertensive and beneficial cardiovascular effects of genistein have been reported by Montenegro et al. (Reference Montenegro, Pessa and Tanus-Santos18). The observed mechanisms were inhibition of plasma ACE, both in vivo and in vitro, reduction in the hypertensive response to angiotensin I and increase in the hypotensive response to bradykinin.

The kallikrein enzyme acts on kininogen substrates to release kinins, which bind to the bradykinin B2 receptors, and cause vasodilatation by NO production. Defects in vasodilatory mechanism in fructose-fed rats have been found to be associated with decreased NO production(Reference Kamata and Yamashita19) and infusion of sodium nitroprusside, an NO donor, improves insulin sensitivity in fructose-fed rats(Reference Oshida, Tachi and Morishita20). Vascular protective effects and regulation of eNOS by genistein have been observed in animal and human studies(Reference Mahn, Borras and Knock21, Reference Räthel, Leikert and Vollmar22). Genistein enhances eNOS gene transcription and protein synthesis in spontaneously hypertensive rats(Reference Si and Liu23). Stimulation of NO release and reduction in peripheral vascular resistance by genistein could influence BP and the distribution of blood flow in this model.

Several studies reveal that insulin could regulate the expression of eNOS gene through activation of phosphatidyl inositol 3 kinase (PI3) kinase(Reference Kuboki, Jiang and Takahara24). We earlier showed that genistein promotes insulin action in high fructose-fed rats by assessing the homeostasis model assessment values (control rats, 9·39; fructose-fed rats, 42·89; fructose+genistein (1 mg/kg body weight per d), 11·16; control+genistein (1 mg/kg body weight per d), 8·87), although we did not measure PI3 kinase activation by genistein. Although genistein is a known tyrosine kinase inhibitor, studies have shown that at physiological (low dose) concentration, it induces tyrosine phosphorylation of insulin-like growth factor 1 receptor and insulin receptor substrate-1 in cancer cell lines(Reference Chen and Wong25). We suggest that genistein by its insulin sensitivity effects might activate eNOS expression partly via the activation of PI3 kinase.

Podocytes are visceral epithelial cells that share the characteristics of both a mesenchymal and an epithelial cell. The podocyte slit diaphragm is the final filtration barrier in the glomerular endothelial basement membrane–podocyte interface. Podocytopathy is present early in the natural history of diabetic nephropathy and plays an important role in micro- and macro-albuminuria. Broadening of foot processes with eventual effacement and dysfunction of podocytes were observed in fructose-fed rats. In addition, podocyte abnormalities were mitigated by genistein treatment.

A large body of evidence has demonstrated that oxidative stress is an important mediator of fructose-induced membrane and structural damage of kidney(Reference Choi, Park and Kim26). Genistein might have recovered the kidney from structural alterations by its antioxidative capacity. This has been reported earlier by us using haematoxylin and eosin staining(Reference Palanisamy, Viswanathan and Anuradha8). Periodic acid–Schiff staining not only shows glycogen content, but also detects structural deterioration characterised by disorganisation of contractile and cytoskeletal proteins. Genistein was found to improve the overall renal structure.

Elevated circulating lipids may contribute to renal disease progression(Reference Vaziri and Norris27). In contention with this, we observed marked increases in lipid levels in both plasma and kidney of fructose-fed rats. Lipids bind to extracellular matrix molecules and are peroxidised, thereby increasing reactive oxygen species production(Reference Morena, Patrier and Jaussent28). These processes affect the structure and function of the kidney. Genistein has been shown to be a PPARγ agonist to improve lipid metabolism in murine RAW 264.7 cells and in obese Zucker rats(Reference Mezei, Banz and Steger29). The lipid-lowering effect of genistein observed in the present study could be one of the mechanisms for its renoprotective action.

Hyperglycaemia and angiotensin II are potent inducers of PKC-βII activation(Reference Koya and King30). Activated PKC-βII translocates to the membrane and triggers the production of endothelin 1, a vasoactive peptide that causes endothelial dysfunction, vascular smooth muscle contraction and proliferation, leading to a rise in BP(Reference Pfaff and Vallon31). Fructose-fed rats exhibited hyperglycaemia and enhanced ACE activity in the present study. Lowering of glucose and inhibition of ACE by genistein could have contributed to the reduction in PKC-βII activation and BP. The BP-lowering effect of genistein has been shown already in stroke-prone spontaneously hypertensive rats(Reference Xu, Ikeda and Yamori32), ovariectomised hypertensive rats(Reference Nevala, Lassila and Finckenberg33) and in spontaneously hypertensive rats(Reference Vera, Galisteo and Villar34). Important vascular effects of genistein include an increase in inducible NO synthase activity, inhibition of platelet aggregation and arterial vasorelaxation(Reference Mishra, Abbot and Choudhury35) and prevention of vascular alterations(Reference Bermejo, Zarzuelo and Duarte36).

In conclusion, the inhibitory effect on ACE and PKC-βII activation and increased availability of kinins and NO could be the contributory mechanism for the BP-lowering effect and renoprotection by genistein. It remains to be seen whether the insulin sensitivity effect of genistein may underlie the renoprotective action.

Acknowledgements

The financial support in the form of a Senior Research Fellowship to Dr N. P. from the Indian Council of Medical Research, New Delhi is gratefully acknowledged. A. C. V. designed the study protocol and edited the draft manuscript; N. P. conducted the experiments, analysed the data and wrote the draft of the manuscript. All authors read and approved the final manuscript. The authors declare no conflict of interest.

References

1Reaven, GM (1994) Syndrome X: six years later. J Int Med 736, 1322.Google Scholar
2Sanchez-Lozada, LG, Tapia, E, Jimenez, A, et al. (2007) Fructose-induced metabolic syndrome is associated with glomerular hypertension and renal microvascular damage in rats. Am J Physiol Renal Physiol 292, 423429.Google Scholar
3Haller, H, Baur, E, Quass, P, et al. (1995) High glucose concentrations and protein kinase C isoforms in vascular smooth muscle cells. Kidney Int 47, 10571067.Google Scholar
4Rajasekar, P, Palanisamy, N & Anuradha, CV (2007) Increase in nitric oxide and reductions in blood pressure, protein kinase C beta II and oxidative stress by l-carnitine: a study in the fructose-fed hypertensive rat. Clin Exp Hypertens 29, 517530.CrossRefGoogle ScholarPubMed
5Nishimoto, Y, Tomida, T, Matsui, H, et al. (2002) Decrease in renal medullary endothelial nitric oxide synthase of fructose-fed, salt-sensitive hypertensive rats. Hypertension 40, 190194.Google Scholar
6Park, SA, Choi, MS, Cho, SY, et al. (2006) Genistein and daidzein modulate hepatic glucose and lipid regulating enzyme activities in C57BL/KsJ-db/db mice. Life Sci 79, 12071213.Google Scholar
7Kojima, T, Uesugi, T, Toda, T, et al. (2002) Hypolipidemic action of the soybean isoflavones genistein and genistin in glomerulonephritic rats. Lipids 37, 261265.Google Scholar
8Palanisamy, N, Viswanathan, P & Anuradha, CV (2008) Effect of genistein, a soy isoflavone, on whole body insulin sensitivity and renal damage induced by a high-fructose diet. Ren Fail 30, 645654.Google Scholar
9Rock, E, Astier, C, Lab, C, et al. (1995) Magnesium deficiency in rats induces a rise in plasma nitric oxide. Magnes Res 8, 237242.Google ScholarPubMed
10Holmquist, B, Bünning, P & Riordan, JF (1979) A continuous spectrophotometric assay for angiotensin converting enzyme. Anal Biochem 95, 540548.Google Scholar
11Trautschold, I, Werle, E & Schweitzer, G (1974) Methods in Enzymatic Analysis, vol. 2, New York, NY: Academic Press, Inc.Google Scholar
12Raghuramulu, N, Madhavan Nair, K & Kalyanasundaram, K (2003) A Manual of Laboratory Techniques, 2nd ed.Hyderabad: National Institute of Nutrition Press.Google Scholar
13Lowry, OH, Rosebrough, NJ, Farr, AL, et al. (1951) Protein measurement with the Folin phenol reagent. J Biol Chem 193, 265275.CrossRefGoogle ScholarPubMed
14Vasdev, S, Gill, V, Parai, S, et al. (2002) Dietary vitamin E and C supplementation prevents fructose induced hypertension in rats. Mol Cell Biochem 241, 107114.CrossRefGoogle Scholar
15Bhanot, S, Salh, BS, Verma, S, et al. (1999) In vivo regulation of protein-serine kinases by insulin in skeletal muscle of fructose-hypertensive rats. Am J Physiol 277, 299307.Google Scholar
16Vasdev, S, Longerich, L & Gill, V (2004) Prevention of fructose-induced hypertension by dietary vitamins. Clin Biochem 37, 19.Google Scholar
17Xu, YY, Yang, C & Li, SN (2006) Effects of genistein on angiotensin-converting enzyme in rats. Life Sci 79, 828837.CrossRefGoogle ScholarPubMed
18Montenegro, MF, Pessa, LR & Tanus-Santos, JE (2009) Isoflavone genistein inhibits the angiotensin-converting enzyme and alters the vascular responses to angiotensin I and bradykinin. Eur J Pharmacol 607, 173177.CrossRefGoogle ScholarPubMed
19Kamata, K & Yamashita, K (1999) Insulin resistance and impaired endothelium-dependent relaxation in fructose-hypertensive rats. Am J Hypertens 9, 370376.Google Scholar
20Oshida, Y, Tachi, Y, Morishita, Y, et al. (2000) Nitric oxide decreases insulin resistance induced by high-fructose feeding. Horm Metab Res 32, 339342.CrossRefGoogle ScholarPubMed
21Mahn, K, Borras, C, Knock, GA, et al. (2005) Dietary soy isoflavone induced increases in antioxidant and eNOS gene expression lead to improved endothelial function and reduced blood pressure in vivo. FASEB J 19, 17551757.CrossRefGoogle ScholarPubMed
22Räthel, TR, Leikert, JF, Vollmar, AM, et al. (2005) The soy isoflavone genistein induces a late but sustained activation of the endothelial nitric oxide-synthase system in vitro. Br J Pharmacol 144, 394399.Google Scholar
23Si, H & Liu, D (2008) Genistein, a soy phytoestrogen, upregulates the expression of human endothelial nitric oxide synthase and lowers blood pressure in spontaneously hypertensive rats. J Nutr 138, 297304.Google Scholar
24Kuboki, K, Jiang, ZY, Takahara, N, et al. (2000) Regulation of endothelial constitutive nitric oxide synthase gene expression in endothelial cells and in vivo: a specific vascular action of insulin. Circulation 101, 676681.Google Scholar
25Chen, WF & Wong, MS (2004) Genistein enhances insulin-like growth factor signaling pathway in human breast cancer (MCF-7) cells. J Clin Endocrinol Metab 89, 23512359.Google Scholar
26Choi, HN, Park, YH, Kim, JH, et al. (2011) Renoprotective and antioxidant effects of Saururus chinensis Baill in rats fed a high-fructose diet. Nutr Res Pract 5, 365369.Google Scholar
27Vaziri, ND & Norris, K (2011) Lipid disorders and their relevance to outcomes in chronic kidney disease. Blood Purif 31, 189196.Google Scholar
28Morena, M, Patrier, L, Jaussent, I, et al. (2011) Reduced glomerular filtration rate, inflammation and HDL cholesterol as main determinants of superoxide production in non-dialysis chronic kidney disease patients. Free Radic Res 45, 735745.Google Scholar
29Mezei, O, Banz, WJ, Steger, RW, et al. (2003) Soy isoflavones exert antidiabetic and hypolipidemic effects through the PPAR pathways in obese Zucker rats and murine RAW 264.7 cells. J Nutr 133, 12381243.Google Scholar
30Koya, D & King, GL (1998) Protein kinase C activation and the development of diabetic complications. Diabetes 47, 859866.Google Scholar
31Pfaff, IL & Vallon, V (2002) Protein kinase C beta isoenzymes in diabetic kidneys and their relation to nephroprotective actions of the ACE inhibitor lisinopril. Kidney Blood Press Res 25, 329340.Google Scholar
32Xu, JW, Ikeda, K & Yamori, Y (2004) Genistein inhibits expressions of NADPH oxidase p22 phox and angiotensin II type 1 receptor in aortic endothelial cells from stroke-prone spontaneously hypertensive rats. Hypertens Res 27, 675683.CrossRefGoogle Scholar
33Nevala, R, Lassila, M, Finckenberg, P, et al. (2002) Genistein treatment reduces arterial contractions by inhibiting tyrosine kinases in ovariectomized hypertensive rats. Eur J Pharmacol 452, 8796.Google Scholar
34Vera, R, Galisteo, M, Villar, IC, et al. (2005) Soy isoflavones improve endothelial function in spontaneously hypertensive rats in an estrogen-independent manner: role of nitric-oxide synthase, superoxide, and cyclooxygenase metabolites. J Pharmacol Exp Ther 314, 13001309.Google Scholar
35Mishra, SK, Abbot, SE, Choudhury, Z, et al. (2000) Endothelium-dependent relaxation of rat aorta and main pulmonary artery by the phytoestrogens genistein and daidzein. Cardiovasc Res 46, 539546.Google Scholar
36Bermejo, A, Zarzuelo, A & Duarte, J (2003) In vivo vascular effects of genistein on a rat model of septic shock induced by lipopolysaccharide. J Cardiovasc Pharmacol 42, 329338.Google Scholar
Figure 0

Table 1 Body and kidney weight, systolic and diastolic blood pressure, mean arterial pressure (MAP), heart rate, angiotensin-converting enzyme (ACE), kallikrein and nitrite in plasma and urine of experimental rats (Mean values and standard deviations of six animals)

Figure 1

Fig. 1 (a) Plasma and (b) kidney lipid profile of experimental animals. CHOL, cholesterol. (a) , Control rats (CON); , fructose-fed rats (FRU); , fructose+genistein (1 mg/kg body weight per d) (FRU+GEN); , control+genistein (1 mg/kg body weight per d) (CON+GEN). (b) , CON; , FRU; , FRU+GEN; , CON+GEN. * Mean values were significantly different compared to CON (P< 0·05; ANOVA followed by Duncan's multiple range test). † Mean values were significantly different compared to FRU (P< 0·05; ANOVA followed by Duncan's multiple range test).

Figure 2

Fig. 2 Representative immunoblot of protein kinase C-βII (PKC-βII) isoenzyme in kidney (a) cytosol and (b) membrane and representative densitometry analysis of Western blots a and b, respectively. Values are expressed as relative intensity and means of six independent experiments, and standard deviations represented by vertical bars. CON, control rats; FRU, fructose-fed rats; FRU+GEN, fructose+genistein (1 mg/kg body weight per d); CON+GEN, control+genistein (1 mg/kg body weight per d). * Values were significantly different compared to CON (P< 0·05; ANOVA followed by Duncan's multiple range test). † Values were significantly different compared to FRU (P< 0·05; ANOVA followed by Duncan's multiple range test). AU, arbitrary units.

Figure 3

Fig. 3 Representative immunoblot of endothelial nitric oxide synthase (eNOS) in kidney cell lysate and representative densitometry analysis of the bands. Values are expressed as relative intensity and means of six independent experiments, and standard deviations represented by vertical bars. CON, control rats; FRU, fructose-fed rats; FRU+GEN, fructose+genistein (1 mg/kg body weight per d); CON+GEN, control+genistein (1 mg/kg body weight per d). * Mean values were significantly different compared to CON (P< 0·05; ANOVA followed by Duncan's multiple range test). † Mean values were significantly different compared to FRU (P< 0·05; ANOVA followed by Duncan's multiple range test). AU, arbitrary units.

Figure 4

Fig. 4 Representative micrographs of kidney tissue stained with periodic acid–Schiff from (a) control rats, (b) fructose-fed rats, (c) fructose+genistein-treated rats (1 mg/kg body weight per d) and (d) control+genistein-treated rats (1 mg/kg body weight per d). (a–d) Magnification 400 × . (A colour version of this figure can be found online at http://www.journals.cambridge.org/bjn).

Figure 5

Fig. 5 (a–d) Transmission electron microscopic examination of rat kidney ( × 10 000). (a) An electron micrograph from a control rat. Regularly arranged podocyte foot processes with normal basement membrane are observed. (b) An electron micrograph from a fructose-fed rat. Abnormal foot processes effacement, podocyte appear broadened, basement membrane thickening and extensive deposition of matrix proteins (blue arrows) are seen. (c) An electron micrograph from a genistein-treated fructose-fed rat shows basement membrane within normal limits; some podocyte foot processes appear broadened. (d) Electron micrograph from a genistein-treated control rat shows normal structure of podocytes and basement membrane.