Honokiol protects against renal ischemia/reperfusion injury via the suppression of oxidative stress, iNOS, inflammation and STAT3 in rats

  • Authors:
    • Yongwu Yu
    • Mingxv Li
    • Ning Su
    • Zhiyong Zhang
    • Haidan Zhao
    • Hai Yu
    • Yingluan Xu
  • View Affiliations

  • Published online on: December 9, 2015     https://doi.org/10.3892/mmr.2015.4660
  • Pages: 1353-1360
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Honokiol is the predominant active ingredient in the commonly used traditional Chinese medicine, Magnolia, which has been confirmed in previous studies to exhibit anti‑oxidation, antimicrobial, antitumor and other pharmacological effects. However, its effects on renal ischemia/reperfusion injury (IRI) remain to be elucidated. The present study aimed to examine the effects of honokiol on renal IRI, and to investigate its potential protective mechanisms in the heart. Male adult Wistar albino rats were induced into a renal IRI model. Subsequently, the levels of serum creatinine, blood urea nitrogen (BUN), alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP), and the levels of serum nitrite and the kidney nitrite were examined in the IRI group. The levels of oxidative stress, inducible nitric oxide synthase (iNOS), inflammatory factors and caspase‑3 were evaluated using a series of commercially available kits. The levels of phosphorylated signal transducer and activator of transcription 3 (p‑STAT3) and the protein expression levels of STAT3 were determined using western blotting. Pretreatment with honokiol significantly reduced the levels of serum creatinine, BUN, ALT, AST and ALP, and the level of nitrite in the kidney of the IRI group, compared with the control group. The levels of malondialdehyde, the activity of myeloperoxidase, and the gene expression and activity of iNOS were reduced in the IRI rats, compared with the sham‑operated rats, whereas the levels of superoxide dismutase and catalase were increased following treatment with honokiol in the IRI rats. In addition, the expression levels of tumor necrosis factor‑α and interleukin‑6 in the IRI rats were increased by honokiol. Treatment with honokiol suppressed the protein expression levels of p‑STAT3 and caspase‑3 in the IRI rats. These findings indicated that honokiol protects against renal IRI via the suppression of oxidative stress, iNOS, inflammation and STAT3 in the rat.

Introduction

Investigations in previous years have shown that the predominant cause of acute renal failure is closely associated with renal ischemia/reperfusion injury (IRI), as ischemia-reperfusion can lead to renal vasoconstriction, tubular obstruction, anti-leakage of glomerular filtrate and decreased glomerular filtration rate, leading to impaired renal function, and conditions of shock, heart failure and requirement for kidney transplant. This is often accompanied by renal ischemia and reperfusion, which affect treatment outcomes (1,2). Therefore, reducing or avoiding IRI is one of the areas in renal protection, which has received significant attention.

The generation of excessive reactive oxygen species disrupts normal redox homeostasis of renal tissue, causing a state of oxidative stress in renal tissues (1). Previous studies have demonstrated that, in sustained diabetes, states of hyperglycemia and oxidative stress in vivo, and non-enzymatic glycation (glycosylation) reactions are evident, which are important in the pathogenesis of diabetes and nephropathy (36). Jin et al reported that C-type natriuretic peptide ameliorates IRI-induced acute kidney injury through the inhibition of oxidative stress (7).

The physiological concentration of nitric oxide (NO) is involved in the functional regulation of several vital organisms under normal circumstances, and the pathological induction of renal IRI leads to a significant increase in NO synthesis (8). NO at high concentrations reacts rapidly in the peroxide microenvironment at the site of injury to produce peroxynitrite ion (ONOO-), directly or indirectly leading to the damage of target cells and tissues (9). Inflammatory reactions can markedly promote IRI (10). Certain reports have suggested that IRI is a process of inflammation, although this is debated, and reflects the importance of IRI in inflammatory reactions (11). A cascade network, comprising reactive oxygen species, a substantial number of NO generated by inducible nitric oxide synthase (iNOS) and inflammatory reactions is an important mechanism of IRI (12). A previous study demonstrated that inhibiting the expression of lipopolysaccharide-induced iNOS synthase and inflammation reduces the content of NO in rats with acute myocardial ischemia (13). A previous study demonstrated that the development of IRI is characterized by the activation of the signal transducer and activator of transcription (STAT) pathway, which is involved in signaling associated with various cytokines and growth factors (14).

Magnolia officinalis is widely used in traditional Chinese and in Japanese Kampo medicines, which are used clinically to treat bacterial infections, inflammation and gastrointestinal diseases (15). Since 1973, two of the predominant active compounds in Magnolia officinalis have been isolated, magnolol and its isomer honokiol (Fig. 1), and have been investigated in several studies (1619). Previous studies have demonstrated that honokiol has pharmacological functions, including central muscle relaxation, central nervous system inhibition, anti-inflammatory, antibacterial, anti-ulcer, anti-oxidative and anticancer properties, and hormone regulation (2023). Therefore, the present study aimed to evaluate the protective effects of honokiol against IRI and examine its possible mechanism.

Materials and methods

Reagents and kits

Serum creatinine, blood urea nitrogen (BUN) and nitrite commercial kits were purchased from BioAssay Systems (Hayward, CA, USA). Malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), myeloperoxidase (MPO), tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 commercial kits were purchased from Jiancheng Bioengineering Institute (Nanjing, China). TRIzol was purchased from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA). SYBR Green I dye was purchased from Qiagen GmbH (Hilden, Germany). The BIOER Linegene-3320 system was purchased from Hangzhou Bioer Technology, (Hangzhou, China). The Bicinchoninic Acid (BCA) assay kit was purchased from Thermo Fisher Scientific, Inc.

Animals

Male adult Wistar albino rats, weighing 250–300 g, were provided by the Animal Experimental Center of the Navy General Hospital of Chinese PLA and maintained at a room temperature of 23±2°C, with a 12 h light-dark cycle, and were allowed 1 week to acclimatize to the conditions with free access to water and food. The present study was approved by the Experimental Animal Research Committee of the Navy General Hospital of Chinese PLA (Beijing, China) and the ethics committee of the Navy General Hospital of Chinese PLA (Beijing, China).

Induction of renal IRI and experimental protocol

The Wistar rats were used to perform the renal ischemia/reperfusion surgery, as described previously (24,25). Briefly, the rats were anesthetized by intraperitoneal (i.p.) injection of 10 mg/kg xylazine (Jiangsu Biological Technology, Co., Ltd. Jiangsu, China) and 100 mg/kg of ketamine hydrochloride (Sigma-Aldrich, St. Louis, MO, USA). The site of surgery (abdomen) was shaved and swabbed with betadine solution (Beyotime Institute of Biotechnology, Jiangsu, China) and ethanol. Particular care was taken to avoid damage to the organs throughout. A single medial incision was made, the kidney vasculature was exposed and rats were subjected to renal IRI injury by placing a clamp on the vessels for 45 min. In the sham operation group, surgery was performed, but the kidneys were not clamped. After 45 min, the clamp was removed and blood flow perfused into the kidneys. At this stage, the animals exhibiting insufficient restoration of blood flow or with vessel damage were excluded from the experiment.

The rats were randomly allocated into four groups: (1) Sham group (n=10), in which the normal rats received physiological saline (i.p.); (2) sham+honokiol group (Sham+Hon; n=10), in which normal rats received 5 mg/kg, i.p. honokiol; (3) IRI group (IRI; n=10), in which the IRI model rats received physiological saline (i.p.); (4) IRI+honokiol group (IRI+Hon; n=10), in which the IRI model rats received 5 mg/kg, i.p. honokiol.

Assessment of renal function

Rats were sacrificed by decapitation and the blood samples were collected from the rats at the room temperature. The samples were centrifuged at 12,000 × g for 10 min, following which the supernatants were collected. The levels of serum creatinine and BUN in the samples were measured using commercially available kits, according to the manufacturer's protocol (BioAssay Systems). Nitrite levels were measured using a colorimetric assay kit, according to the manufacturer's protocol (BioAssay Systems).

Assessment of hepatic function

Rats were anesthetized with-ketamine (75 mg/kg; Sigma-Aldrich) and xylazine (5 mg/kg; Sigma-Aldrich) and blood samples were collected from the eye socket at room temperature. The samples were centrifuged at 12,000 g for 10 min, following which the supernatants were collected. The levels of alkaline phosphatase (ALP), aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were measured using an autoanalyzer (Pars Azmun, Karaj, Iran).

Assessment of oxidative stress

Rats were sacrificed by decapitation and the areas of ischemia in the renal tissues were collected at room temperature and homogenised using liquid nitrogen and lysed in radioimmunoprecipitation assay bugger (Jiancheng Bioengineering Institute). The samples were centrifuged at 12,000 × g for 10 min, following which the supernatants were collected. The levels of MDA and SOD and the activities of CAT and MPO in the renal ischemic zone of the tissues were measured using colorimetric assay kits (Jiancheng Bioengineering Institute), according to the manufacturer's protocol.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis of iNOS

The ischemic zone tissue samples were collected at room temperature and homogenised using liquid nitrogen and lysed in radioimmunoprecipitation assay bugger (Jiancheng Bioengineering Institute). The samples were centrifuged at 12,000 g for 10 min, following which the supernatants were collected. Total RNA (1 µg) was isolated from the supernatants using TRIzol, according to the manufacturer's protocol (Invitrogen; Thermo Fisher Scientific, Inc.). cDNA was obtained following the RT reaction using SYBR Green I dye (Qiagen GmbH). qPCR amplifications were performed using a BIOER Linegene-3320 system (Hangzhou Bioer Technology). The sequences of the primers used were as follows: iNOS, forward 5′-AGTGATGGCAAGCACGACTTC-3′ and reverse 5′-TCTGTCACTCGCTCACCACGG-3′; and β-actin, forward 5′-AAGGGACTTCCTGTAACAATGCA-3′ and reverse 5′-CTGGAACGGTGAAGGTGACA-3′. The cycling conditions were as follows: 5 min at 95°C, 40 cycles of 30 sec at 95°C, 45 sec at 60°C, and 30 sec at 72°C, followed by a cycle of 10 min at 72°C. The expression levels was quantified by Ct value: Ct=−1/lg(1+Ex)*lgX0+lgN/lg(1+Ex) (26,27).

Assessment of iNOS activity

The ischemic zone tissue samples were collected at room temperature and homogenised using liquid nitrogen and lysed in radioimmunoprecipitation assay bugger (Jiancheng Bioengineering Institute). The samples were centrifuged at 12,000 g for 10 min and the supernatants were collected. The renal ischemic zone tissues were homogenized and nuclear proteins were quantified using a BCA assay (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The supernatant was incubated with 0.6 ml reaction buffer (containing 210 mM sucrose, 40 mM NaCl, 2 mM EGTA and 30 mM HEPES; Beyotime Institute of Biotechnology) and 1 mmol/l ethylene glycol tetraacetic acid (Amresco LLC, Solon, OH, USA) at room temperature for 30 min. The activity of iNOS was then determined at 530 nm using a TECH M200 microplate reader (Tecan Group, Ltd., Männedorf, Switzerland).

Assessment of inflammatory cytokines

The ischemic zone tissue samples were collected at room temperature and homogenised using liquid nitrogen and lysed in radioimmuno-precipitation assay buffer (Jiancheng Bioengineering Institute). The samples were centrifuged at 12,000 g for 10 min and the supernatants were collected. The levels of TNF-α and IL-6 in the were measured using a colorimetric assay kit, according to the manufacturer's protocol (Jiancheng Bioengineering Institute).

Western blot analysis of the protein expression of STAT3

The ischemic zone tissue samples were collected at room temperature. The samples were centrifuged at 12,000 g for 10 min and the supernatants were collected. The renal ischemic zone tissues were homogenized, and nuclear proteins were extracted using a BCA assay (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The proteins (50 µg) were electrophoresed on 12% SDS-polyacrylamide gels (Jiangsu Biological Technology, Co., Ltd.) and transferred into nitrocellulose membranes (Abcam, Cambridge, UK) at 4°C for 2 h. The membranes were blocked with Tris-buffered saline-0.05% Tween 20 (TBST) containing 5% skim milk powder for 1 h at room temperature. Following blocking, the membranes were incubated with rabbit polyclonal anti-phosphorylated (p-)STAT3 (sc-135649; 1:1,500; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), rabbit polyclonal anti-STAT3 (sc-7179; 1:1,000; Santa Cruz Biotechnology, Inc.) and rabbit polyclonal anti-β-actin (sc-7210; 1:500; Santa Cruz Biotechnology, Inc.) overnight at 4°C with agitation. Subsequently, the membranes were washed with TBST for 1 h at room temperature and incubated with goat anti-rabbit IgG-PerCP-Cy5.5 secondary antibody (sc-45101; 1:1,000; Santa Cruz Biotechnology, Inc.). Finally, the membranes were visualized using an enhanced chemiluminescence system (Pierce Biotechnology, Rockford, IL, USA). Bands were exposed in a ChemiDoc-It TS2 imager Imager (UVP, LLC, Upland, CA, USA) and analyzed using Image J version 2 software (National Institutes of Health, Bethesda, MD, USA).

Assessment of caspase-3 activity

The ischemic zone tissue samples were collected at room temperature. The samples were centrifuged at 12,000 g for 10 min and the supernatants were collected. The renal ischemic zone tissues were homogenized and nuclear proteins were extracted using a BCA assay (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Equal quantities of protein (30 µg) and Ac-LEHD-pNA (Beyotime Institute of Biotechnology) were incubated at 37°C for 2 h in the dark, following which the activity of caspase-3 was measured using a SpectraMax M2 Microplate Autoreader (Bio-Tek Instruments Inc., Winooski, VT, USA at an absorbance of 405 nm.

Statistical analysis

All statistical data are presented as the mean ± standard error of the mean, and statistical analyses were performed using SPSS software, version 17.0 (SPSS, Inc., Chicago, IL, USA). Statistical analysis was conducted with three or more groups using one-way analysis of variance and Dunnett's test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of honokiol on renal function

The levels of serum creatinine and BUN in the IRI group were significantly increased, compared with those in the sham group (Fig. 2A and B). The elevation in the levels of serum creatinine and BUN in the IRI rats were reduced significantly with honokiol pretreatment in the IRI+Hon group, compared with the IRI group (Fig. 2A and B).

Effect of honokiol on hepatic function

As shown in Fig. 3A–C, the serum levels of ALT, AST and ALP were elevated following IRI, compared with the respective levels in the sham-operated group. The administration of honokiol prior to IRI was observed to significant reduce the serum levels of ALT, AST and ALP, compared with the levels in the IRI group (Fig. 3A–C).

Honokiol decreases the levels of nitrite in the kidney

As shown in Fig. 4A, the level of serum nitrite in the IRI group was higher, compared with the levels in the Sham group, Sham+Hon group and IRI+Hon group. However, the differences identified between the groups were not statistically significant (Fig. 4A). The level of nitrite in the kidney of the IRI group was increased significantly, compared with that in the sham group (Fig. 4B). Honokiol administration prior to IRI led to a decrease in kidney nitrite levels, compared with the IRI group (Fig. 4B).

Honokiol reduces levels of oxidative stress

IRI caused a significant increase in the serum level of MDA and activity of MPO, compared with the sham group, and honokiol treatment decreased the levels of MDA and activity of MPO, compared with the IRI group (Fig. 5A). The activities of SOD and CAT decreased in the IRI group, compared with the sham group (Fig. 5B and C). However, the administration of honokiol significantly increased the activities of SOD and CAT, compared with the IRI group (Fig. 5B and C). Honokiol treatment decreased the activity of MPO, compared with the IRI group (Fig. 5D).

Honokiol reduces the expression and activity of iNOS

The expression and activity levels of iNOS in the IRI group were significantly augmented, compared with those in the Sham group, exhibiting significant increases (Fig. 6A and B). By contrast, treatment with honokiol prior to IRI caused a significant reduction in the expression and activity of iNOS, compared with the IRI group (Fig. 6A and B).

Honokiol reduces levels of inflammatory cytokines

The levels of TNF-α and IL-6 in the IRI group increased significantly, compared with those in the Sham group (Fig. 7A and B). These elevated levels of TNF-α and IL-6 in the IRI rats were reduced significantly by honokiol pretreatment (Fig. 7A and B).

Honokiol decreases the protein expression of STAT3

In order to elucidate the mechanistic basis of the effects of honokiol In the IRI rats, the protein expression levels of STAT3 were measured using western blotting. The results suggested that the protein expression of p-STAT3 was promoted in the IRI group, compared with the Sham group (Fig. 8A and B). The increased protein expression of p-STAT3 by IRI was reversed in the rats pretreated with honokiol (Fig. 8A and B).

Honokiol reduces levels of caspase-3

The results of the present study showed that increased activity of caspase-3 was observed in the IRI group, compared with the Sham group (Fig. 9). In addition. honokiol administration prior to IRI significantly reduced the levels of caspase-3 levels, compared with the levels observed in the IRI group (Fig. 9).

Discussion

The kidney is an organ with a high level of perfusion, and is particularly sensitive to ischemia and reperfusion. When ischemia persists for a certain duration, following which perfusion recovers, organizational structural and functional recovery may be impaired and kidney dysfunction and structural damage may be aggravated, which is known as IRI (28). Following shock, heart failure, cardiopulmonary bypass or kidney transplantation, IRI usually affects the treatment outcome, particularly in kidney transplant recipients, in which IRI is one of the causes of surgical failure. At present, the pathogenesis of IRI remains to be fully elucidated. In the present study, honokiol pretreatment reduced the levels of serum creatinine, BUN, ALT, AST and ALP, and the levels of nitrite in the kidneys of IRI rats. Therefore, honokiol may be a potential drug for treatment following IRI.

Oxidative stress is one of the important pathogenetic mechanisms of IRI. In ischemia, a high level of ATP is decomposed, leading to the accumulation of hypoxanthine, recovery of blood perfusion causes the generation of a substantial number of reactive oxygen free radicals, and peroxy radicals and their degradation products cause tissue damage by lipid peroxidation of mitochondria and lipid membranes (29). In the present study, honokiol treatment depressed serum levels of MDA and MPO, and increased the activities of SOD and CAT in the IRI rats. Hsu et al reported that honokiol protected against heatstroke in diabetic rats through reducing inflammation and oxidative stress (30).

NO in the body is generated by NOS, and the level of NO is closely associated with the level and activity of NOS. In normal kidney tissues, endothelial NOS is predominantly expressed in the renal blood vessels and capillaries; neuronal NOS is predominantly expressed in the juxtaglomerular macula densa; and iNOS is expressed in the renal medullary thick ascending limb, proximal tubule, distal convoluted tubule and interlobular arteries, arcuate arteries and blood vessels, and other areas of the glomerulus (31). Under a pathophysiological state, iNOS exhibits high levels of expression in mesangial cells, epithelial cells, smooth muscle cells and renal tubular epithelial cells, and there are high levels of infiltrated inflammatory cells, including in the glomeruli and renal interstitium (32). The present study showed that honokiol significantly decreased the gene expression and activity of iNOS in the IRI rats. Previous studies have suggested that honokiol prevents the inflammatory response and the expression of iNOS in human osteoarthritis chondrocytes (33), and the level of iNOS is attenuated by honokiol in septic rats (34).

Inflammatory reactions, including a series of complex pathological processes, develop and interconnect with each other, which can be roughly divided into the following four processes: Leukocyte activation, chemotaxis, leukocyte-endothelial cell adhesion and migration. In IRI, polymorphonuclear cells within the kidney or proximal tubule, activating factors, including bradykinin, histamine, leukotrienes and platelet-activating factors, which are generated by mesangial cells, pro-inflammatory cytokines, including TNF-α, IL-1β, IL-6, and monocyte chemoattractant protein-1, macrophage inflammatory protein-2, interferon-inducible protein 10 and other chemokines, lead to leukocyte activation and subsequent chemotaxis to the site of injury (35). Subsequently, following interaction with vascular endothelial cells, the leukocytes roll and then adhere closely to the skin layer in the endoderm of blood vessels, migrating into the skin layer and ultimately penetrating the endoderm to reach the extravascular tissue where it is exerts its effects (36). Inflammatory cells directly damage cells following reperfusion by the release of oxidase and hydrolytic enzymes, and adhered neutrophils block the capillary bed, which further increases the circulatory disorder. The present study showed that honokiol significantly reduced the levels of TNF-α and IL-6 in the IRI rats. Chiang et al demonstrated that honokiol protects against eccentric exercise-induced skeletal muscle damage by inhibiting oxidative stress and inflammation in rats (37), and Munroe et al indicated that the anti-inflammatory effects of honokiol decreased the levels of TNF-α and IL-6 in a mouse model of allergic asthma (38).

The STAT pathway is important in cytokine signaling, and STAT transcription factors exist in the cytoplasm during the resting state, which can be activated by cytokines, growth factors, and reactive oxygen species, as intracellular signal transduction proteins and transcription factors. Once phosphorylated by Janus kinase, STATs form homologous or heterologous dimers, which are then translocated into the nucleus and combine with DNA, initiating gene transcription. Ishikawa et al reported that honokiol induces cell cycle arrest and apoptosis through inhibiting the DNA binding of nuclear factor-ϰB and STAT3 (39). Previous studies have suggested that the STAT signaling pathway and IRI have a specific association. IRI can produce large quantities of reactive oxygen species, and when it exceeds the processing ability of antioxidant enzymes, reactive oxygen species accumulate, leading to excessive oxidative stress and cell damage, and the direct activation of STAT3 (40). In conditions of hypoxia without reperfusion, ATP may not be sufficient to make STAT3 fully phosphorylated; however, following reperfusion, ATP levels rise again, and STAT3 can be activated to a maximum degree by phosphorylation (41). In the present study, honokiol suppressed the protein expression of p-STAT3 in the IRI rats. Yu et al suggested that honokiol exerts pro-apoptotic effects on transformed Barrett's cells through inhibition of STAT3 (42). Luan et al reported that honokiol induces cell cycle arrest and apoptosis through inhibiting the DNA binding of nuclear factor-κB and STAT3 (43). In the present study, honokiol administration significantly decreased the levels of caspase-3 in the IRI rats. Weng et al suggested that honokiol attenuates the severity of acute pancreatitis and lung injury through suppression of apoptosis and caspase-3 activity (44). Honokiol also inhibits the activation of caspase-3 and caspase-9 in H2O2-induced apoptosis in human lens epithelial cells (45).

In conclusion, the present study demonstrated the protect effect of honokiol on renal IRI through the suppression of oxidative stress, iNOS, inflammation and STAT3 in the rats. These results may be of potential clinical relevance, and the protective effect of honokiol as a clinical therapeutic strategy may be of value in the future.

References

1 

Senbel AM, AbdelMoneim L and Omar AG: Celecoxib modulates nitric oxide and reactive oxygen species in kidney ischemia/reperfusion injury and rat aorta model of hypoxia/reoxygenation. Vascul Pharmacol. 62:24–31. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Cai Y, Xu H, Yan J, Zhang L and Lu Y: Molecular targets and mechanism of action of dexmedetomidine in treatment of ischemia/reperfusion injury. Mol Med Rep. 9:1542–1550. 2014.PubMed/NCBI

3 

Cetin N, Suleyman H, Sener E, Demirci E, Gundogdu C and Akcay F: The prevention of ischemia/reperfusion induced oxidative damage by venous blood in rabbit kidneys monitored with biochemical, histopatological and immunohistochemical analysis. J Physiol Pharmacol. 65:383–392. 2014.PubMed/NCBI

4 

Cetin N, Suleyman H, Sener E, Demirci E, Gundogdu C and Akcay F: The prevention of ischemia/reperfusion induced oxidative damage by venous blood in rabbit kidneys monitored with biochemical, histopatological and immunohistochemical analysis. J Physiol Pharmacol. 65:383–392. 2014.PubMed/NCBI

5 

Jiang G, Liu X, Wang M, Chen H, Chen Z and Qiu T: Oxymatrine ameliorates renal ischemia-reperfusion injury from oxidative stress through Nrf2/HO-1 pathway. Acta Cir Bras. 30:422–429. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Zhuan-Yun LI, Xue-Ping Y, Bin L, Reheman HN, Yang G, Zhan S and Qi MA: Auricularia auricular-judae polysaccharide attenuates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress and inflammation. Biomed Rep. 3:478–482. 2015.PubMed/NCBI

7 

Jin X, Zhang Y, Li X, Zhang J and Xu D: C-type natriuretic peptide ameliorates ischemia/reperfusion-induced acute kidney injury by inhibiting apoptosis and oxidative stress in rats. Life Sci. 117:40–45. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Tripatara P, Patel NS, Webb A, Rathod K, Lecomte FM, Mazzon E, Cuzzocrea S, Yaqoob MM, Ahluwalia A and Thiemermann C: Nitrite-derived nitric oxide protects the rat kidney against ischemia/reperfusion injury in vivo: Role for xanthine oxidoreductase. J Am Soc Nephrol. 18:570–580. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Reyes-Ocampo J, Ramirez-Ortega D, Vazquez Cervantes GI, Pineda B, Montes de Oca Balderas P, González-Esquivel D, Sánchez-Chapul L, Lugo-Huitrón R, Silva-Adaya D, Ríos C, et al: Mitochondrial dysfunction related to cell damage induced by 3-hydroxykynurenine and 3-hydroxyanthranilic acid: Non-dependent-effect of early reactive oxygen species production. Neurotoxicology. 50:81–91. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Koc M, Kumral ZN, Özkan N, Memi G, Kaçar Ö, Bilsel S, Çetinel Ş and Yeğen BÇ: Obestatin improves ischemia/reperfusion-induced renal injury in rats via its antioxidant and anti-apoptotic effects: Role of the nitric oxide. Peptides. 60:23–31. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Yan R, Li Y, Zhang L, Xia N, Liu Q, Sun H and Guo H: Augmenter of liver regeneration attenuates inflammation of renal ischemia/reperfusion injury through the NF-kappa B pathway in rats. Int Urol Nephrol. 47:861–868. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Garcia-Criado FJ, Rodriguez-Barca P, Garcia-Cenador MB, Rivas-Elena JV, Grande MT, Lopez-Marcos JF, Mourelle M and López-Novoa JM: Protective effect of new nitrosothiols on the early inflammatory response to kidney ischemia/reperfusion and transplantation in rats. J Interferon Cytokine Res. 29:441–450. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Chen YY, Yeh CH, So EC, Sun DP, Wang LY and Hsing CH: Anticancer drug 2-methoxyestradiol protects against renal ischemia/reperfusion injury by reducing inflammatory cytokines expression. Biomed Res Int. 2014:4315242014.PubMed/NCBI

14 

Hsu YH, Li HH, Sung JM, Chen WT, Hou YC and Chang MS: Interleukin-19 mediates tissue damage in murine ischemic acute kidney injury. PLoS One. 8:e560282013. View Article : Google Scholar : PubMed/NCBI

15 

Seo KH, Nam YH, Lee DY, Ahn EM, Kang TH and Baek NI: Recovery effect of phenylpropanoid glycosides from Magnolia obovata fruit on alloxan-induced pancreatic islet damage in zebrafish (Danio rerio). Carbohydr Res. 416:70–74. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Kumar A, Kumar Singh U and Chaudhary A: Honokiol analogs: A novel class of anticancer agents targeting cell signaling pathways and other bioactivities. Future Med Chem. 5:809–829. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Kumar A, Kumar Singh U and Chaudhary A: Honokiol analogs: A novel class of anticancer agents targeting cell signaling pathways and other bioactivities. Future Med Chem. 5:809–829. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Raison-Peyron N, Cesaire A, Du-Thanh A and Dereure O: Allergic contact dermatitis caused by Magnolia officinalis bark extract in a facial anti-ageing cream. Contact Dermatitis. 72:416–417. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Ghys K, De Palma A, Vandevenne A, Werbrouck J and Goossens A: Magnolia officinalis bark extract, a recently identified contact allergen in 'anti-ageing' cosmetics. Contact Dermatitis. 73:130–132. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Woodbury A, Yu SP, Wei L and Garcia P: Neuro-modulating effects of honokiol: A review. Front Neurol. 4:1302013. View Article : Google Scholar : PubMed/NCBI

21 

Fried LE and Arbiser JL: Honokiol, a multifunctional anti-angiogenic and antitumor agent. Antioxid Redox Signal. 11:1139–1148. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Tian W, Xu D and Deng YC: Honokiol, a multifunctional tumor cell death inducer. Pharmazie. 67:811–816. 2012.PubMed/NCBI

23 

Shen JL, Man KM, Huang PH, Chen WC, Chen DC, Cheng YW, Liu PL, Chou MC and Chen YH: Honokiol and magnolol as multifunctional antioxidative molecules for dermatologic disorders. Molecules. 15:6452–6465. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Mozaffari-Khosravi H, Ahadi Z and Fallah Tafti M: The effect of green tea versus sour tea on insulin resistance, lipids profiles and oxidative stress in patients with type 2 diabetes mellitus: A randomized clinical trial. Iran J Med Sci. 39:424–432. 2014.PubMed/NCBI

25 

Monji A, Mitsui T, Bando YK, Aoyama M, Shigeta T and Murohara T: Glucagon-like peptide-1 receptor activation reverses cardiac remodeling via normalizing cardiac steatosis and oxidative stress in type 2 diabetes. Am J Physiol Heart Circ Physiol. 305:H295–H304. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Zheng Z, Ge Y, Zhang J, et al: PUFA diets alter the microRNA expression profiles in an inflammation rat model. Mol Med Rep. 11:4149–4157. 2015.PubMed/NCBI

27 

Pfaffl MW, Horgan GW and Dempfle L: Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res. 30:e362002. View Article : Google Scholar : PubMed/NCBI

28 

Sagiroglu T, Torun N, Yagci M, Yalta T, Sagiroglu G and Oguz S: Effects of apelin and leptin on renal functions following renal ischemia/reperfusion: An experimental study. Exp Ther Med. 3:908–914. 2012.PubMed/NCBI

29 

Liu ZG, Qi ZC, Liu WL and Wang WZ: Lutein protects against ischemia/reperfusion injury in rat kidneys. Mol Med Rep. 11:2179–2184. 2015.

30 

Hsu CC, Chen LF, Lin MT and Tian YF: Honokiol protected against heatstroke-induced oxidative stress and inflammation in diabetic rats. Int J Endocrinol. 2014:1345752014. View Article : Google Scholar : PubMed/NCBI

31 

Kinaci MK, Erkasap N, Kucuk A, Koken T and Tosun M: Effects of quercetin on apoptosis, NF-κB and NOS gene expression in renal ischemia/reperfusion injury. Exp Ther Med. 3:249–254. 2012.PubMed/NCBI

32 

Zhang J, Li JH, Wang L, Han M, Xiao F, Lan XQ, Li YQ, Xu G and Yao Y: Glucocorticoid receptor agonist dexamethasone attenuates renal ischemia/reperfusion injury by up-regulating eNOS/iNOS. J Huazhong Univ Sci Technolog Med Sci. 34:516–520. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Chen YJ, Tsai KS, Chan DC, Lan KC, Chen CF, Yang RS and Liu SH: Honokiol, a low molecular weight natural product, prevents inflammatory response and cartilage matrix degradation in human osteoarthritis chondrocytes. J Orthop Res. 32:573–580. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Li N, Xie H, Li L, Wang J, Fang M, Yang N and Lin H: Effects of honokiol on sepsis-induced acute kidney injury in an experimental model of sepsis in rats. Inflammation. 37:1191–1199. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhang B, Rong R, Li H, Peng X, Xiong L, Wang Y, Yu X and Mao H: Heat shock protein 72 suppresses apoptosis by increasing the stability of X-linked inhibitor of apoptosis protein in renal ischemia/reperfusion injury. Mol Med Rep. 11:1793–1799. 2015.

36 

Asaga T, Ueki M, Chujo K and Taie S: JTE-607, an inflammatory cytokine synthesis inhibitor, attenuates ischemia/reperfusion-induced renal injury by reducing neutrophil activation in rats. J Biosci Bioeng. 106:22–26. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Chiang J, Shen YC, Wang YH, Hou YC, Chen CC, Liao JF, Yu MC, Juan CW and Liou KT: Honokiol protects rats against eccentric exercise-induced skeletal muscle damage by inhibiting NF-kappaB induced oxidative stress and inflammation. Eur J Pharmacol. 610:119–127. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Munroe ME, Businga TR, Kline JN and Bishop GA: Anti-inflammatory effects of the neurotransmitter agonist Honokiol in a mouse model of allergic asthma. J Immunol. 185:5586–5597. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Ishikawa C, Arbiser JL and Mori N: Honokiol induces cell cycle arrest and apoptosis via inhibition of survival signals in adult T-cell leukemia. Biochim Biophys Acta. 1820:879–887. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Di Domenico F, Casalena G, Jia J, Sultana R, Barone E, Cai J, Pierce WM, Cini C, Mancuso C, Perluigi M, et al: Sex differences in brain proteomes of neuron-specific STAT3-null mice after cerebral ischemia/reperfusion. J Neurochem. 121:680–692. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Yang Y, Duan W, Jin Z, Yi W, Yan J, Zhang S, Wang N, Liang Z, Li Y, Chen W, et al: JAK2/STAT3 activation by melatonin attenuates the mitochondrial oxidative damage induced by myocardial ischemia/reperfusion injury. J Pineal Res. 55:275–286. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Yu C, Zhang Q, Zhang HY, Zhang X, Huo X, Cheng E, Wang DH, Arbiser JL, Spechler SJ and Souza RF: Targeting the intrinsic inflammatory pathway: Honokiol exerts proapoptotic effects through STAT3 inhibition in transformed Barrett's cells. Am J Physiol Gastrointest Liver Physiol. 303:G561–569. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Luan HF, Zhao ZB, Zhao QH, Zhu P, Xiu MY and Ji Y: Hydrogen sulfide postconditioning protects isolated rat hearts against ischemia and reperfusion injury mediated by the JAK2/STAT3 survival pathway. Braz J Med Biol Res. 45:898–905. 2012.PubMed/NCBI

44 

Weng TI, Wu HY, Chen BL and Liu SH: Honokiol attenuates the severity of acute pancreatitis and associated lung injury via acceleration of acinar cell apoptosis. Shock. 37:478–484. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Xia J, Wu Z, Yu C, He W, Zheng H, He Y, Jian W, Chen L, Zhang L and Li W: miR-124 inhibits cell proliferation in gastric cancer through down-regulation of SPHK1. J Pathol. 227:470–480. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 13 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu Y, Li M, Su N, Zhang Z, Zhao H, Yu H and Xu Y: Honokiol protects against renal ischemia/reperfusion injury via the suppression of oxidative stress, iNOS, inflammation and STAT3 in rats. Mol Med Rep 13: 1353-1360, 2016
APA
Yu, Y., Li, M., Su, N., Zhang, Z., Zhao, H., Yu, H., & Xu, Y. (2016). Honokiol protects against renal ischemia/reperfusion injury via the suppression of oxidative stress, iNOS, inflammation and STAT3 in rats. Molecular Medicine Reports, 13, 1353-1360. https://doi.org/10.3892/mmr.2015.4660
MLA
Yu, Y., Li, M., Su, N., Zhang, Z., Zhao, H., Yu, H., Xu, Y."Honokiol protects against renal ischemia/reperfusion injury via the suppression of oxidative stress, iNOS, inflammation and STAT3 in rats". Molecular Medicine Reports 13.2 (2016): 1353-1360.
Chicago
Yu, Y., Li, M., Su, N., Zhang, Z., Zhao, H., Yu, H., Xu, Y."Honokiol protects against renal ischemia/reperfusion injury via the suppression of oxidative stress, iNOS, inflammation and STAT3 in rats". Molecular Medicine Reports 13, no. 2 (2016): 1353-1360. https://doi.org/10.3892/mmr.2015.4660