Canadian Journal of Physiology and Pharmacology

Heat shock protein 27 and 70 contribute to the protection of Schisandrin B against D-galactosamine-induced liver injury in mice

Journal: Canadian Journal of Physiology and Pharmacology

Manuscript ID cjpp-2015-0419.R1

Manuscript Type: Article

Date Submitted by the Author: 22-Sep-2015

Complete List of Authors: Gao, Zhiying; Medical University Zhang, Jishun;Draft Beijing Chaoyang Hospital, Capital Medical University Li, Libo; Qiqihar Medical University, Department of Pharmacology Shen, Longqing; Qiqihar Medical University Li, Qingyi; Qiqihar Medical University Zou, Yu; Qiqihar Medical University Du, Xiaohui; Qiqihar Medical University Zhao, Zhibo; Qiqihar Medical University

Schisandrin B, Heat shock proteins, D-galactosamine, Liver injury, Keyword: Quercetin

https://mc06.manuscriptcentral.com/cjpp-pubs Page 1 of 27 Canadian Journal of Physiology and Pharmacology

1

Heat shock protein 27 and 70 contribute to the protection of Schisandrin B

against D-galactosamine-induced liver injury in mice

Zhiying Gao 1, Jishun Zhang 2, Libo Li 3* , Longqing Shen 4, Qingyi Li 3, Yu Zou 3,

Xiaohui Du 3, Zhibo Zhao 4

1Polygenic Disease Institute, Qiqihar Medical University, 333 Bukui Street,

Jianhua , Qiqihar 161006, .

2Department of Gastroenterology and Hepatology, Beijing Chaoyang Hospital, Capital Medical University, Draft 5 Jingyuan Road, Shijingshan District, Beijing 100043, China.

3Department of Pharmacology, Qiqihar Medical University, 333 Bukui Street,

Jianhua District, Qiqihar 161006, China.

4School of Basic Medicine, Qiqihar Medical University, 333 Bukui Street, Jianhua

District, Qiqihar 161006, China.

*Corresponding author

Libo Li, Department of Pharmacology, Qiqihar Medical University, 333 Bukui

Street, Jianhua District, Qiqihar 161006, China.

Tel: +864522663159; Fax: +864522663722; Email: [email protected]

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 2 of 27

2

Abstract: Schisandrin B is a hepatoprotective component isolated from a traditional Chinese herb Schisandra chinensis (Turcz.) Baill. This study determined the effect of Schisandrin B on Dgalactosamine induced liver injury and the role of Heat shock protein 27 and 70 against liver injury in mice. Acute liver injury was induced by intraperitoneal injection of Dgalactosamine to mice, and Schisandrin B was given orally. The protein and gene expression of Heat shock protein 27 and 70 were detected by Western blot and realtime quantitative polymerase chain reaction respectively. Liver tissues were subjected to histological evaluation, and activities of alanine aminotransferase and aspartate aminotransferase inDraft the serum were measured. Pretreatment of

Schisandrin B significantly attenuated Dgalactosamineinduced liver injury in mice. This result was evidenced by improved alteration of histopathological hepatic necrosis and reduced alanine aminotransferase and aspartate aminotransferase activities in the serum. The hepatoprotective effect was accompanied with overexpression of Heat shock protein 27 and 70 both at the protein and mRNA levels. However, the aforementioned actions of Schisandrin B were all markedly suppressed by the Heat shock protein inhibitor Quercetin.

Heat shock protein 27 and 70 were involved in the protective effect of

Schisandrin B against Dgalactosamine induced liver injury in mice.

Key words

Schisandrin B; Heat shock proteins; induction; protection; Dgalactosamine;

https://mc06.manuscriptcentral.com/cjpp-pubs Page 3 of 27 Canadian Journal of Physiology and Pharmacology

3

Liver injury; mice; Quercetin

Introduction

Hepatitis has high incidence and mortality worldwide and remains a major public

health threat. This disorder is caused by various agents, such as alcohol, viruses,

environmental toxins, and drugs (Hwang et al. 2005). Liver injury is a basic

pathogenesis of various forms of hepatitis. Longterm presence of liver injury

leads to liver fibrosis, cirrhosis, and even hepatic carcinoma (Zou et al. 2006).

Therefore, rectifying liver injury is an important strategy to cure hepatitis clinically.

Dgalactosamine (DGal)challengedDraft mice is a wellestablished animal model to

investigate acute hepatic injury, which resembles clinical viral hepatitis both in

morphology and function (Decker and Keppler 1972; Keppler et al. 1968). Thus,

this model is widely used to evaluate hepatoprotective activity (Myagmar et al.

2004; Visen et al. 1998).

Schisandrin B (Fig. 1) is an active dibenzocyclooctadiene lignan component

that is isolated from the fruit of a wellknown Chinese herb Schisandra chinensis

(Turcz.) Baill., and is particularly effective in viral and chemicalinduced hepatitis

(Chan 2012; Liu 1989). SchB possesses a significant hepatoprotective effect

against injuries caused by chemicals, such as carbon tetrachloride (CCl 4) (Chiu

et al. 2007; Ip et al. 1995) and ethanol (Lam et al. 2010) induced hepatotoxicity,

as well as injuries caused by drugs, such as tacrine (Pan et al. 2002) and

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 4 of 27

4

menadione (Ip et al. 2000) induced liver damage in animals. SchB also has beneficial effects on TNF alphainduced hepatic apoptosis in mice (Ip et al. 2001) and on oxidative injury or lipoperoxidative damage in vitro (Chiu and Ko 2006;

Zhang et al. 1992).

Heat shock proteins (HSPs) are a highly conserved family of proteins that facilitate proper folding or clearance of damaged proteins in the cytoplasm (Hartl

1996; Jäättelä 1999). HSPs are molecular chaperones that are essential in maintaining cellular homeostasis during normal cell growth and stressful conditions (Fink 1999; Hartl 1996). Among the HSPs, inducible HSP27 and

HSP70 have mostly been investigatedDraft for their ability to protect cells and tissues from damage caused by pathological agents (Jäättelä 1999; Kültz 2005) including liver injury (Fujimori et al. 1997; Fujisawa et al. 2013; Mikami et al.

2004). Hepatic HSP27 and HSP70 have been recently reported to be involved in protecting against DGalinduced hepatocyte apoptosis (Bao and Liu 2010a,

2010 b). HSPs also play a role in the hepatoprotective mechanism of SchB.

Previous studies report that SchB protects against CCl 4 hepatotoxicity (Tang et al. 2003) and TNF αtriggered hepatic apoptosis (Ip et al. 2001) in mice or rats through an HSPinducing effect.

Our recent study has demonstrated that pretreatment with SchB significantly increases HSP27 and HSP70 expression at both the protein and gene levels in normal mice (Li et al. 2014). Furthermore, the hepatoprotection of SchB against

https://mc06.manuscriptcentral.com/cjpp-pubs Page 5 of 27 Canadian Journal of Physiology and Pharmacology

5

acetaminophen hepatotoxicityinduced liver injury is also attributed to its

induction of hepatic HSP27 and HSP70 (Li et al. 2014). In the present study, we

used a DGalinduced liver injury mouse model to determine whether the

hepatoprotective effect of SchB is a result of the induction of hepatic HSPs.

Materials and methods

Chemicals and reagents

SchB was purchased from Push BioTechnology Co. Ltd. (Chengdu, China).

DGal and Quercetin (Que) withDraft a purity of more than 98% by HPLC were

obtained from SigmaAldrich (St. Louis, MO, USA). SchB and Que were

separately suspended in 0.5% [w/v] sodium carboxymethyl cellulose for oral

administration. DGal was dissolved in physiological saline for injection. All other

chemicals and reagents used were of analytical grade.

Animals and treatment

Male ICR mice (18 g to 22 g) were obtained from the National Laboratory Animal

Center (Changchun, China). The animals were initially acclimated for 5 days and

maintained in a 12h light/dark cycle of 22 °C to 24 °C and a relative humidity of

50%. Food and water were given ad libitum. The animal care and experimental

protocols were approved by Guide for the Care and Use of Laboratory Animals

(1996, published by National Academy Press, 2101 Constitution Ave. NW,

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 6 of 27

6

Washington, DC 20055, USA) and the Animal Care and Ethical Committee of

Qiqihar Medical University, China (Ethics no. 2011200409).

Mice were randomly divided into six groups ( n=12) as follows: Control, DGal,

DGal + Que, SchB + DGal, SchB + DGal + Que, and Que . SchB was administered intragastrically at a dose of 200 mg/kg three times in 1 day, with an interval of 8 h. Que was given orally to mice at a dose of 200 mg/kg combined with SchB. DGal was intraperitoneally injected once at a dosage of 800 mg/kg

12 h before the last administration of SchB . The control mice were given equal volume of vehicles. Mice were sacrificed by decapitation 4 h after the last dose of SchB . Blood samples were thenDraft harvested to determine aminotransferase activities. Liver tissues were used for subsequent analyses.

Western blot analysis

A total of 30 g of sample proteins extracted from liver tissue was separated by sodium dodecylsulfate–polyacrylamide gel electrophoresis (SDSPAGE) in a

12% polyacrylamide gel and then transferred to a polyvinylidene difluoride membrane (Pall Corp., Port Washington, NY, USA). The membranes were blocked with 5% (w/v) skim milk in Trisbuffered saline with Tween 20 (TBST) for

1 h at room temperature and then probed with primary antibodies at 4 °C overnight. The antibodies were rabbit antiHSP27 (Cell Signaling, MA, USA), mouse antiHSP70 and mouse antiβactin (internal control) (Santa Cruz

Biotechnology, Santa Cruz, CA, USA). The following day, blots were incubated

https://mc06.manuscriptcentral.com/cjpp-pubs Page 7 of 27 Canadian Journal of Physiology and Pharmacology

7

with horseradish peroxidaseconjugated secondary antibodies in TBST for 2 h at

room temperature. The protein bands were visualized using an enhanced

chemiluminescence plus kit (Beijing Applygen Technologies, Inc., Beijing, China)

and analyzed using GelPro Analyzer 4.0 software (Media Cybernetics, Rockville,

MD, USA). Each experiment was carried out in triplicate.

Real-time quantitative PCR (qPCR) assay

Total RNA isolated from the liver tissue was used to synthesize the

complementary DNA with an ExScript RT kit . Messenger RNA (mRNA) levels

were measured by realtime qPCR using SYBR Premix Ex Taq in an ABI 7300

realtime qPCR system (AppliedDraft Biosystems, Foster City, CA, USA). The kits

and primers were obtained from Takara Biotechnology of China (Dalian, China).

βactin primers were used as the internal control. The following primer

sequences were employed: HSP27 sense primer,

5′GTCCCTGGACGTCAACCACT3′, and HSP27 antisense primer,

5′GAGATGTAGCCATGTTCGTCCT3′; HSP70 sense primer,

5′CAGAGGCCAGGGCTGGATTA3′, and HSP70 antisense primer,

5′ACACATGCTGGTGCTGTCACTTC3′; βactin sense primer,

5′CATCCGTAAAGACCTCTATGCCAAC3′, and βactin antisense primer,

5′ATGGAGCCACCGAT CCACA3′. The thermal cycling parameters were as

follows: 1 cycle of 95 °C for 30 s, 40 cycles of 5 s at 95 °C, and 31 s at 60 °C.

Threshold cycle (Ct) data were collected using Sequence Detection Software

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 8 of 27

8

version 1.2.3 (Applied Biosystems). Relative quantification of gene expression was analyzed by the 2 −∆∆ Ct method. Ct values of the HSP genes were normalized to the Ct values of βactin to obtain delta Ct (∆Ct). Fold change in

HSP genes relative to the βactin internal control was determined by the

−∆∆Ct following equation: Fold change = 2 , where ∆∆Ct = (Ct HSP gene − Ct βactin ) −

(Ct control − Ct βactin). Each experiment was conducted in triplicate.

Determination of the aminotransferase activities

The activities of alanine aminotransferase (ALT) and aspartate aminotransferase

(AST) in serum were measured using an assay kit (Beijing BHKT Clinical

Reagent Co. Ltd., Beijing, China)Draft according to the manufacturer’s instructions.

Histopathological analysis

The paraffin method was used to analyze hepatic histology. In brief, the fresh right lobe of the mouse livers was immediately fixed in 10% formalin. The tissue was then dehydrated with different grades of ethanol, embedded in paraffin, and cut into 5 m sections. The paraffin was removed with xylene and ethanol, and the slide was stained with hematoxylin and eosin. Morphological evaluation was performed under a light microscope equipped with the Nikon D600 digital camera and then analyzed by ImagePro Plus 7.0 software (Media Cybernetics).

The pathological investigator was blinded to the treatments in the experiment.

Statistical analysis

All data were expressed as mean ± standard deviation (SD). SPSS 13.0

https://mc06.manuscriptcentral.com/cjpp-pubs Page 9 of 27 Canadian Journal of Physiology and Pharmacology

9

software (SPSS Inc., Chicago, IL, USA) was used for statistical analysis.

Intergroup differences were detected using oneway ANOVA, followed by

Dunnett’s test. Pvalues <0.05 were considered statistically significant.

Results

SchB increased the expression of hepatic HSP27 and HSP70 proteins in

D-Gal-challenged mice

Western blot analysis showed that the hepatic HSP27 and HSP70 proteins in

DGalchallenged mice markedlyDraft increased compared with the normal control

group. Oral administration of SchB at 200 mg/kg increased the expression of

hepatic HSP27 and HSP70 proteins in mice more significantly, by approximately

4.9 and 3.8fold, respectively. This result indicates that SchB exerted more

potent inducing effect on HSP27 than HSP70. However, Que inhibited the

inducing effect of SchB and DGal both on HSP27 and HSP70. Compared with

the normal control group, Que itself exhibited no noticeable effect on the

expression of HSP27 and HSP70 proteins (Fig. 2).

SchB increased the expression of hepatic HSP27 and HSP70 mRNAs in

D-Gal-challenged mice

Realtime qPCR analysis showed that hepatic HSP27 and HSP70 mRNAs in

DGaltreated mice markedly increased. SchB at 200 mg/kg upregulated the

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 10 of 27

10

expression of hepatic HSP27 and HSP70 mRNAs by approximately 6.1 and

5.3fold, respectively, indicating that the gene level results were in accordance with HSP27 and HSP70 proteins. Increased levels of HSP27 and HSP70 mRNAs induced by SchB (200 mg/kg) were also markedly suppressed by Que

(200 mg/kg) treatment. Combined with the same observation in the protein expression, these results suggest Que is a feasible inhibitory tool in this study

(Fig. 3).

SchB-induced hepatic HSP27 and HSP70 attenuated hepatoxicity in mice treated with D-Gal

Biochemical assay showed thatDraft the serum activities of ALT and AST in

DGALtreated mice were significantly greater than those in the control group.

Oral administration with SchB (200 mg/kg) before DGAL injection in mice significantly suppressed the serum activities of ALT and AST by 60.30% and

55.61%, respectively, showing the hepatoprotective effect of SchB. However, the activities of ALT and AST relapsed when Que was coadministered with SchB, without difference compared with the DGALtreated mice. By contrast, Que neither increased the serum activities of ALT and AST nor affected the increase of such activities induced by DGAL treatment (Fig. 4).

Protective effect of hepatic HSP27 and HSP70 induced by SchB against

D-Gal-induced liver injury in histology in mice

The histological assay demonstrated the normal hepatic lobular architecture and

https://mc06.manuscriptcentral.com/cjpp-pubs Page 11 of 27 Canadian Journal of Physiology and Pharmacology

11

cell structure of mice liver in the control group. Injection of DGAL resulted in

significant morphological changes in liver histology. These changes included

disseminated hepatocyte necrosis, congestion, ballooning, and destruction

around the central vein, as well as immigration of inflammatory cells, which were

markedly attenuated by SchB pretreatment in mice. However, the

hepatoprotective effect of SchB was suppressed by Que (200 mg/kg) treatment,

as indicated by heavier aggravated lesions compared with the SchB + DGal

group. Que per se exhibited neither protective nor injurious effect in mice (Fig.

5).

Draft

Discussion

We investigated a possible molecular mechanism of the hepatoprotective effect

of SchB in a mouse model with DGalinduced acute liver injury. SchB

attenuated DGalinduced hepatic injury by overexpression of HSP27 and

HSP70 in mice.

DGalinduced acute liver injury is a widely used animal model that closely

resembles human viral hepatitis (Decker and Keppler 1972; Keppler et al. 1968).

Injection with DGal decreases uracil nucleotides in the liver, markedly depleting

hepatic UDPglucuronic acid. DGal inhibits hepatic glucuronidation, disrupting

the synthesis of essential uridylate nucleotides. Reduction of these nucleotides

eventually impairs protein and glycoprotein synthesis, causing progressive

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 12 of 27

12

damage of cellular membranes. This leads to changes in membrane permeability and ultimately enzyme leakage from the cells (AbdulHussain et al.

1991; Keppler et al. 1970). In this study, we used DGalinduced acute liver injury model to investigate the actions and mechanisms of SchB. Pretreatment with SchB protected the liver against DGalinduced hepatotoxicity in mice, as demonstrated by the amelioration of histopathological hepatic necrosis and the reduction in ALT and AST activities.

HSP27 and HSP70 are multidimensional proteins that are implicated in a range of diseases; thus, they serve as disease biomarkers and may be a potential drug target (Evans etDraft al. 2010; Vidyasagar et al. 2012). Also, HSP70 has been reported to be a chaperone involved in inflammation, endoplasmic reticulum stress and apoptosis at liver level (Tarantino et al. 2012). Because

HSPs have a cytoprotective effect on tissue injury, we hypothesized that the hepatoprotective effect of SchB is due to the induction of hepatic HSPs. Our previous study confirmed that SchB induces the expression of HSP27 and

HSP70 in mice. Hepatic HSP27 and HSP70 production were enhanced following

SchB administration. HSP27 and HSP70 mRNA and protein expression increased in a time and dosedependent manner. Moreover, the pattern of protein expression paralleled mRNA levels, suggesting that the induction of hepatic HSP27 and HSP70 by SchB is through the upregulation of HSP gene transcription (Li et al. 2014). The present study demonstrated that SchB

https://mc06.manuscriptcentral.com/cjpp-pubs Page 13 of 27 Canadian Journal of Physiology and Pharmacology

13

significantly induced both mRNA levels and protein expression of HSP27 and

HSP70 in DGaltreated mice compared with DGal alone challenged mice,

which showed higher HSP27 and HSP70 expression than the normal control

group.

To investigate whether the upregulation of HSP27 and HSP70 by SchB is

responsible for its hepatoprotective action against DGalinduced liver injury, we

used Que, an inhibitor of HSP biosynthesis (Gonzalez et al. 2009; Hosokawa et

al. 1990; Nagai et al. 1995). The results showed that Que significantly

suppressed the induction of hepatic HSP27 and HSP70 by SchB, but Que itself

did not affect the expression ofDraft hepatic HSP27 and HSP70 in the normal control

or DGalinjected mice. In the SchB (200 mg/kg) pretreated mice, compared with

DGaltreated mice, reduced ALT and AST activities and decreased extent of

hepatic necrosis were observed. This finding suggests that SchB has a

protective effect on DGalinduced liver injury. Simultaneous administration of

Que (200 mg/kg) and SchB (200 mg/kg) in DGaltreated mice attenuated the

protective effect of SchB on ALT and AST activities and hepatic necrosis, but

Que itself exhibited neither hepatoprotective nor injurious effect on mice. Our

results demonstrated that Que treatment markedly suppressed the

hepatoprotective effect of SchB, suggesting that the increased HSP27 and

HSP70 expression following SchB treatment contributed to mitigate the extent of

DGalinduced liver injury.

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 14 of 27

14

In summary, we found that oral administration of SchB markedly alleviated

DGalinduced liver injury in mice, as demonstrated by the reduction in ALT and

AST activities and decreased hepatic necrosis, accompanied by the overexpression of HSP27 and HSP70 in the mouse liver. However, the hepatoprotective effects of SchB were significantly attenuated by Que, an inhibitor of HSP biosynthesis. Our findings revealed that SchB administration provide a significant hepatoprotective effect against DGalinduced liver injury in mice, and HSP27 and HSP70 contribute to the protection of SchB.

Because HSPs occur and function endogenously, the upregulation of HSP function by pharmacological approachDraft may provide a potential prophylactic or therapeutic drug target for treatment of liver diseases (Latchman. 1998), such as inducing HSP expression in the treatment of liver injury. Therefore, SchB, with its

HSPinducing capability, might be a potential candidate compound for hepatitis or other hepatic diseases in the future.

Acknowledgements

This work was supported by the Scientific Research Fund of

Province Education Department of China [grant number: 1155G68]. We also thank the Basic Medical Research Section and Molecular Biological Experiment

Center of Qiqihar Medical University for their help.

Conflict of interest

We declare no conflict of interest.

https://mc06.manuscriptcentral.com/cjpp-pubs Page 15 of 27 Canadian Journal of Physiology and Pharmacology

15

References

AbdulHussain, S. K., and Mehendale, H. M. 1991. Studies on the

agedependent effects of galactosamine in primary rat hepatocyte cultures.

Toxicol. Appl. Pharmacol. 107(3): 504513. PMID: 2000637.

Bao, X. Q., and Liu, G. T. 2010 a. Bicyclol protects HepG2 cells against

Dgalactosamineinduced apoptosis through inducing heat shock protein 27 and

mitochondria associated pathway. Acta Pharmacol. Sin. 31(2): 219226. doi:

10.1038/aps.2009.194. PMID: 20139905.

Bao, X. Q., and Liu, G. T. 2010 b. Involvement of HSP70 in the protection of

bicyclol on apoptosis of HepG2Draft cells intoxicated by dgalactosamine. J. Asian

Nat. Prod. Res. 12(4): 313323. doi: 10.1080/10286021003769924. PMID:

20419542.

Chan, S. W. 2012. Panax ginseng, Rhodiola rosea and Schisandra chinensis. Int.

J. Food Sci. Nutr. 63 Suppl 1: 7581. doi: 10.3109/09637486.2011.627840.

PMID: 22039930.

Chiu, P. Y., and Ko, K. M. 2006. Schisandrin Binduced increase in cellular

glutathione level and protection against oxidant injury are mediated by the

enhancement of glutathione synthesis and regeneration in AML12 and H9c2

cells. Biofactors, 26(4): 221230. PMID: 17119269.

Chiu, P. Y., Leung, H. Y., Siu, A. H., Poon, M. K., and Ko, K. M. 2007.

Schisandrin B decreases the sensitivity of mitochondria to calcium ioninduced

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 16 of 27

16

permeability transition and protects against carbon tetrachloride toxicity in mouse livers. Biol. Pharm. Bull. 30(6): 11081112. PMID: 17541162.

Decker, K., and Keppler, D. 1972. Galactosamine induced liver injury. Prog. Liver

Dis. 4: 183199. PMID: 4631501.

Evans, C. G., Chang, L., and Gestwicki, J. E. 2010. Heat shock protein 70

(hsp70) as an emerging drug target. J. Med. Chem. 53(12): 45854602. doi:

10.1021/jm100054f. PMID: 20334364.

Fink, A. L. 1999. Chaperonemediated protein folding. Physiol. Rev. 79(2):

425449. PMID: 10221986.

Fujimori, S., Otaka, M., Otani,Draft S., Jin, M., Okuyama, A., Itoh, S., et al. 1997.

Induction of a 72kDa heat shock protein and cytoprotection against thioacetamideinduced liver injury in rats. Dig. Dis. Sci. 42(9): 19871994. PMID:

9331166.

Fujisawa, K., Yabuuchi, C., Izawa, T., Kuwamura, M., Takasu, N., Torii, M., et al.

2013. Expression patterns of heat shock protein 25 in carbon tetrachlorideinduced rat liver injury. Exp. Toxicol. Pathol. 65(5): 469476. doi:

10.1016/j.etp.2012.02.001. PMID: 22417648.

Gonzalez, O., Fontanes, V., Raychaudhuri, S., Loo, R., Loo, J., Arumugaswami,

V., et al. 2009. The heat shock protein inhibitor Quercetin attenuates hepatitis C virus production. Hepatology, 50(6): 17561764. doi: 10.1002/hep.23232. PMID:

19839005.

https://mc06.manuscriptcentral.com/cjpp-pubs Page 17 of 27 Canadian Journal of Physiology and Pharmacology

17

Hartl, F. U. 1996. Molecular chaperones in cellular protein folding. Nature,

381(6583): 571579. doi: 10.1038/381571a0. PMID: 8637592.

Hosokawa, N., Hirayoshi, K., Nakai, A., Hosokawa, Y., Marui, N., Yoshida, M., et

al. 1990. Flavonoids inhibit the expression of heat shock proteins. Cell Struct.

Funct. 15(6): 393401. PMID: 2085852.

Hwang, J. M., Tseng, T. H., Tsai, Y. Y., Lee, H. J., Chou, F. P., Wang, C. J., et al.

2005. Protective effects of baicalein on tertbutyl hydroperoxideinduced hepatic

toxicity in rat hepatocytes. J. Biomed. Sci. 12(2): 389397. doi:

10.1007/s1137300515728. PMID: 15917992.

Ip, S. P., Poon, M. K., Wu, S. S.,Draft Che, C. T., Ng, K. H., Kong, Y. C., et al. 1995.

Effect of schisandrin B on hepatic glutathione antioxidant system in mice:

protection against carbon tetrachloride toxicity. Planta Med. 61(5): 398401. doi:

10.1055/s2006958123. PMID: 7480197.

Ip, S. P., Yiu, H. Y., and Ko, K. M. 2000. Schisandrin B protects against

menadioneinduced hepatotoxicity by enhancing DTdiaphorase activity. Mol.

Cell. Biochem. 208(12): 151155. PMID: 10939639.

Ip, S. P., Che, C. T., Kong, Y. C., and Ko, K. M. 2001. Effects of schisandrin B

pretreatment on tumor necrosis factoralpha induced apoptosis and Hsp70

expression in mouse liver. Cell Stress Chaperones, 6(1): 4448. PMID:

11525242.

Jäättelä, M. 1999. Heat shock proteins as cellular lifeguards. Ann. Med. 31(4):

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 18 of 27

18

261271. PMID: 10480757.

Keppler, D., Lesch, R., Reutter, W., and Decker, K. 1968. Experimental hepatitis induced by Dgalactosamine. Exp. Mol. Pathol. 9(2): 279290. PMID: 4952077.

Keppler, D. O., Rudigier, J. F., Bischoff, E., and Decker, K. F. 1970. The trapping of uridine phosphates by Dgalactosamine. Dglucosamine, and

2deoxyDgalactose. A study on the mechanism of galactosamine hepatitis. Eur.

J. Biochem. 17(2): 246253. PMID: 5500393.

Kültz, D. 2005. Molecular and evolutionary basis of the cellular stress response.

Annu. Rev. Physiol. 67: 225257. doi:

10.1146/annurev.physiol.67.040403.103635.Draft PMID: 15709958.

Lam, P. Y., Chiu, P. Y., Leung, H. Y., Chen, N., Leong, P. K., and Ko, K. M. 2010.

Schisandrin B cotreatment ameliorates the impairment on mitochondrial antioxidant status in various tissues of longterm ethanol treated rats. Fitoterapia,

81(8): 12391245. doi: 10.1016/j.fitote.2010.08.010. PMID: 20800090.

Latchman, D. S. 1998. Heat shock proteins: protective effect and potential therapeutic use (review). Int. J. Mol. Med. 2(4): 375381. PMID: 9857220.

Li, L., Zhang, T., Zhou, L., Zhou, L., Xing, G., Chen, Y., et al. 2014. Schisandrin B attenuates acetaminopheninduced hepatic injury through heatshock protein 27 and 70 in mice. J. Gastroenterol. Hepatol. 29(3): 640647. doi:

10.1111/jgh.12425. PMID: 24219791.

Liu, G. T. 1989. Pharmacological actions and clinical use of fructus schizandrae.

https://mc06.manuscriptcentral.com/cjpp-pubs Page 19 of 27 Canadian Journal of Physiology and Pharmacology

19

Chin. Med. J (Engl). 102(10): 740749.PMID: 2517053.

Mikami, K., Otaka, M., Goto, T., Miura, K., Ohshima, S., Yoneyama, K., et al.

2004. Induction of a 72kDa heat shock protein and protection against

lipopolysaccharideinduced liver injury in cirrhotic rats. J. Gastroenterol. Hepatol.

19(8): 884890. doi: 10.1111/j.14401746.2004.03401.x. PMID: 15242491.

Myagmar, B. E., Shinno, E., Ichiba, T., and Aniya, Y. 2004. Antioxidant activity of

medicinal herb Rhodococcum vitisidaea on galactosamineinduced liver injury

in rats. Phytomedicine, 11(5): 416423. doi: 10.1016/j.phymed.2003.04.003.

PMID: 15330497.

Nagai, N., Nakai, A., and Nagata,Draft K. 1995. Quercetin suppresses heat shock

response by down regulation of HSF1. Biochem. Biophys. Res. Commun. 208(3):

10991105. doi: 10.1006/bbrc.1995.1447. PMID: 7702609.

Pan, S. Y., Han, Y. F., Carlier, P. R., Pang, Y. P., Mak, D. H., Lam, B. Y., et al.

2002. Schisandrin B protects against tacrine and bis(7)tacrineinduced

hepatotoxicity and enhances cognitive function in mice. Planta Med. 68(3):

217220. doi: 10.1055/s200223145. PMID: 11914957.

Tang, M. H., Chiu, P. Y., and Ko, K. M. 2003. Hepatoprotective action of

schisandrin B against carbon tetrachloride toxicity was mediated by both

enhancement of mitochondrial glutathione status and induction of heat shock

proteins in mice. Biofactors, 19(12): 3342. PMID: 14757975.

Tarantino, G., Finelli, C., Colao, A., Capone, D., Tarantino, M., Grimaldi, E., et al.

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 20 of 27

20

2012. Are hepatic steatosis and carotid intima media thickness associated in obese patients with normal or slightly elevated gammaglutamyltransferase? J.

Transl. Med. 10:50. doi: 10.1186/147958761050. PMID: 22424154.

Vidyasagar, A., Wilson, N. A., and Djamali, A. 2012. Heat shock protein 27

(HSP27): biomarker of disease and therapeutic target. Fibrogenesis Tissue

Repair, 5(1): 7. doi: 10.1186/1755153657. PMID: 22564335.

Visen, P. K., Saraswat, B., and Dhawan, B. N. 1998. Curative effect of picroliv on primary cultured rat hepatocytes against different hepatotoxins: an in vitro study.

J. Pharmacol. Toxicol. Methods, 40(3): 173179. PMID: 10334634.

Zhang, T. M., Wang, B. E., andDraft Liu, G. T. 1992. Effect of schisandrin B on lipoperoxidative damage to plasma membrane of rat liver in vitro. Zhongguo Yao

Li Xue Bao, 13(3): 255258. PMID: 1442110.

Zou, Y., Yang, Y., Li, J., Li, W., and Wu, Q. 2006. Prevention of hepatic injury by a traditional Chinese formulation, BJJN, in mice treated with

BacilleCalmetteGuerin and lipopolysaccharide. J. Ethnopharmacol. 107(3):

442448. doi: 10.1016/j.jep. 2006.04.006. PMID: 16697540.

https://mc06.manuscriptcentral.com/cjpp-pubs Page 21 of 27 Canadian Journal of Physiology and Pharmacology

21

Figure captions:

Fig. 1 Chemical Structure of Schisandrin B

Fig. 2 Schisandrin B (SchB) increased protein expression of hepatic Heat shock

protein (HSP) 27 and HSP70 in Dgalactosamine (DGal)challenged mice. Mice

were orally given SchB (200 mg/kg) three times in 24 h or cotreated with

Quercetin (Que) (200 mg/kg), and DGal (800 mg/kg) was intraperitoneally

injected once 12 h before the last administration of SchB. The control animals

were given the equal volume (20 mL/kg) of vehicles only. Liver tissue samples

were harvested at 4 h after the last dose of SchB, whole liver proteins were extracted, and the protein expressionDraft of hepatic HSP27 and HSP70 was detected by Western blot analysis. (a) Representative Western blot analysis of

HSP27 and HSP70 protein from each group. (b) Levels of hepatic HSP27 and

HSP70 proteins. Protein levels were expressed as mean ± Standard Deviation

(SD) (n=5) in each column. The lanes were normalized to the levels of βactin. * P

<0.05, ** P <0.01 compared with corresponding control group; # P <0.05

compared with corresponding DGal group; && P < 0.01 compared with

corresponding SchB+DGal group. Similar results were obtained with three

replications.

Fig. 3 Schisandrin B (SchB) increased expression of hepatic Heat shock protein

(HSP) 27 and HSP70 mRNA in Dgalactosamine (DGal) challenged mice.

Animal treatment was as described as in Fig. 2. After mice sacrificed, total RNA

was isolated from liver tissue samples using RNAiso reagent and then

subsequently used for cDNA synthesis. The 2 ∆∆ Ct analyses of quantitative

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 22 of 27

22

polymerase chain reaction (qPCR) of HSP27 and HSP70 were obtained from 5 mice per group. Data were expressed as mean ± Standard Deviation (SD). ** P <

0.01 compared with the corresponding vehicletreated mice; ## P <0.01, compared with corresponding DGal group; & P < 0.05 compared with corresponding SchB + DGal group. Similar results were obtained with three replications. Que, Quercetin.

Fig. 4 Schisandrin B (SchB) induced hepatic Heat shock protein (HSP) 27 and

HSP70 attenuated hepatoxicity in mice treated with Dgalactosamine (DGal).

Mice were treated as described as in Fig. 2. Blood samples were harvested and then serum was separated forDraft determination of (a) ALT and (b) AST activities according to the manufacturer’s instruction. Data were expressed as mean ± Standard Deviation (SD) (n=12). ** P < 0.01 compared with the corresponding control group; ## P <0.01, compared with corresponding DGal group; && P < 0.01 compared with corresponding SchB + DGal group. Que,

Quercetin; ALT, alanine aminotransferase; AST, aspartate aminotransferase.

Fig. 5 Protective effect of hepatic Heat shock protein (HSP) 27 and HSP70 induced by Schisandrin B (SchB) against Dgalactosamine (DGal) induced liver injury in histology in mice. Mice were treated as described as in Fig. 2. Liver sections were stained with hematoxylineosin for morphological evaluation

(magnification: 100 ×)

https://mc06.manuscriptcentral.com/cjpp-pubs Page 23 of 27 Canadian Journal of Physiology and Pharmacology

Draft

81x70mm (300 x 300 DPI)

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 24 of 27

Draft

182x196mm (300 x 300 DPI)

https://mc06.manuscriptcentral.com/cjpp-pubs Page 25 of 27 Canadian Journal of Physiology and Pharmacology

Draft

86x71mm (300 x 300 DPI)

https://mc06.manuscriptcentral.com/cjpp-pubs Canadian Journal of Physiology and Pharmacology Page 26 of 27

Draft

153x236mm (300 x 300 DPI)

https://mc06.manuscriptcentral.com/cjpp-pubs Page 27 of 27 Canadian Journal of Physiology and Pharmacology

Draft

110x67mm (300 x 300 DPI)

https://mc06.manuscriptcentral.com/cjpp-pubs