Sheng-Di-Da-Huang Decoction Inhibited Inflammation Expressed in Microglia after Intracerebral Hemorrhage in Rats

Min Cai, Zhonghai Yu, Wen Zhang, Li Yang, Jun Xiang, Jingsi Zhang, Zhennian Zhang, Ting Wu, Xiangting Li, Maodong Fu, Xuxia Bao, Xiaofei Yu, Dingfang Cai, Min Cai, Zhonghai Yu, Wen Zhang, Li Yang, Jun Xiang, Jingsi Zhang, Zhennian Zhang, Ting Wu, Xiangting Li, Maodong Fu, Xuxia Bao, Xiaofei Yu, Dingfang Cai

Abstract

Objects: Sheng-Di-Da-Huang Decoction was used as an effective hemostatic agent in ancient China. However, its therapeutic mechanism is still not clear. Inflammatory injury plays a critical role in ICH-induced secondary brain injury. After hemolysis, hematoma components are released, inducing microglial activation via TLR4, which initiates the activation of transcription factors (such as NF-κB) to regulate expression of proinflammatory cytokine genes. This study aimed to verify the anti-inflammatory effects of Sheng-Di-Da-Huang Decoction on ICH rats.

Materials and methods: Intracerebral hemorrhage was induced by injection of bacterial collagenase (0.2 U) in rats. Neurological deficits, brain water content, Evans blue extravasation, expression of TLR4, NF-κB, Iba-1 positive cells (activated microglia), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) were examined 1, 3, 7, and 14 days after collagenase injection. MR images were also studied.

Results: Sheng-Di-Da-Huang Decoction remarkably improved neurological function, reduced brain water content as well as Evans blue extravasation, downregulated expression of TLR4, NF-κB, TNF-α, and IL-1β, and inhibited microglial activation.

Conclusions: Sheng-Di-Da-Huang Decoction reduced inflammation reaction after ICH through inhibited inflammation expressed in microglia.

Figures

Figure 1
Figure 1
(a) Neurological deficit score (maximum total score is 13) of rats after injection of collagenase. (b) The result of corner turn test. Data were presented as mean ± SEM, n=5 rats for each group. ∗ P<0.05; ∗∗ <0.01 compared with ICH group.
Figure 2
Figure 2
Result of brain water contents for each group at different time points. Data were presented as mean ± SEM, n=5 rats for each group. ∗ P<0.05; ∗∗ <0.01 compared with ICH group.
Figure 3
Figure 3
Results of Evans blue extravasation in each group at different time points. Data were presented as mean ± SEM, n=5 rats for each group. ∗ P<0.05; ∗∗ <0.01 compared with ICH group.
Figure 4
Figure 4
Results of MRI study. The images, taken at the different time points, were from a single rat. (a) Representative T2-weighted MR images of intracerebral hematoma in rat brain at the level of the collagenase injection. (b) Representative SWI images of intracerebral hematoma in rat brains surrounding collagenase injection. n=5 rats for each group at each time point. The images were from different rats in the same group.
Figure 5
Figure 5
(a) Representative immunofluorescence images of Iba-1, ×200 magnified. (b) Iba-1 positive cells percentage (green fluorescence represents Iba-1 positive cell). Data were presented as mean ± SEM, n=5 rats for each group. ∗ P<0.05; ∗∗ <0.01 compared with ICH group.
Figure 6
Figure 6
Detection of TLR4 and NF-κB in the hemorrhagic area using Western blotting. (a) The bands of GAPDH, TLR4, and NF-κB at 1 day. (b) The bands of GAPDH, TLR4, and NF-κB at 3 days. (c) The bands of GAPDH, TLR4, and NF-κB at 7 days. (d) The bands of GAPDH, TLR4, and NF-κB at 14 days. (e) Quantitative results of the bands for TLR4 relative to GAPDH at each time point. (f) Quantitative results of the bands for NF-κB relative to GAPDH at each time point. Data are presented as mean ± SEM, n=5 rats for each group. ∗ P<0.05; ∗∗ <0.01 compared with ICH group. Results of TNF-α (g) and IL-1β (h) concentrations in each group at different time points. Data were presented as mean ± SEM, n=5 rats for each group. ∗ P<0.05; ∗∗ <0.01 compared with ICH group.

References

    1. Hwang B. Y., Appelboom G., Ayer A., et al. Advances in neuroprotective strategies: Potential therapies for intracerebral hemorrhage. Cerebrovascular Disease. 2011;31(3):211–222. doi: 10.1159/000321870.
    1. Zhou Y., Wang Y., Wang J., Anne Stetler R., Yang Q.-W. Inflammation in intracerebral hemorrhage: from mechanisms to clinical translation. Progress in Neurobiology. 2014;115:25–44. doi: 10.1016/j.pneurobio.2013.11.003.
    1. Smiley S. T., King J. A., Hancock W. W. Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4. The Journal of Immunology. 2001;167(5):2887–2894. doi: 10.4049/jimmunol.167.5.2887.
    1. Aronowski J., Hall C. E. New horizons for primary intracerebral hemorrhage treatment: experience from preclinical studies. Neurological Research. 2005;27(3):268–279. doi: 10.1179/016164105x25225.
    1. Wagner K. R., Xi G., Hua Y., et al. Lobar intracerebral hemorrhage model in pigs: rapid edema development in perihematomal white matter. Stroke. 1996;27(3):490–497. doi: 10.1161/01.str.27.3.490.
    1. Ziai W. C. Hematology and inflammatory signaling of intracerebral hemorrhage. Stroke. 2013;44(1):S74–S78. doi: 10.1161/STROKEAHA.111.000662.
    1. Rosenberg G. A., Navratil M. Metalloproteinase inhibition blocks edema in intracerebral hemorrhage in the rat. Neurology. 1997;48(4):921–926. doi: 10.1212/WNL.48.4.921.
    1. Xu M., Yu X. F., Xiang J., Cai D. F. Neuroprotective effects of, Sheng-Di-Da-Huang Decoction , on intracerebral hemorrhage in rats. Acta Universitatis Traditions Medicalis Sinensis Pharmacologiaeque Shanghai. 2016;30:69–72.
    1. Cai M., Yu Z., Wang L., et al. Tongxinluo reduces brain edema and inhibits post-ischemic inflammation after middle cerebral artery occlusion in rats. Journal of Ethnopharmacology. 2016;181:136–145. doi: 10.1016/j.jep.2016.01.026.
    1. MacLellan C. L., Auriat A. M., McGie S. C., et al. Gauging recovery after hemorrhagic stroke in rats: Implications for cytoprotection studies. Journal of Cerebral Blood Flow & Metabolism. 2006;26(8):1031–1042. doi: 10.1038/sj.jcbfm.9600255.
    1. Hua Y., Schallert T., Keep R. F., Wu J., Hoff J. T., Xi G. Behavioral tests after intracerebral hemorrhage in the rat. Stroke. 2002;33(10):2478–2484. doi: 10.1161/01.STR.0000032302.91894.0F.
    1. MacLellan C. L., Davies L. M., Fingas M. S., Colbourne F. The Influence of Hypothermia on Outcome After Intracerebral Hemorrhage in Rats. Stroke. 2006;37(5):1266–1270. doi: 10.1161/01.STR.0000217268.81963.78.
    1. Liu C., Ma R., Wang L., et al. Rehmanniae Radix in osteoporosis: A review of traditional Chinese medicinal uses, phytochemistry, pharmacokinetics and pharmacology. Journal of Ethnopharmacology. 2017;198:351–362. doi: 10.1016/j.jep.2017.01.021.
    1. Liang A. H., Xue B. Y., Wang J. H., Hao J. D., Yang H., Yi H. A study on hemostatic and immunologic actions of fresh- and dry Dihuang. China Journal of Chinese Materia Medica. 1999;24:663–666.
    1. Zhang R. X., Li M. X., Jia Z. P. Rehmannia glutinosa: review of botany, chemistry and pharmacology. Journal of Ethnopharmacology. 2008;117(2):199–214. doi: 10.1016/j.jep.2008.02.018.
    1. Capasso F., Gaginella TS. Laxative. A Practical Guide. Milano: Springer-Verlag: Italia; 1997.
    1. Cirillo C., Capasso R. Constipation and botanical medicines: An overview. Phytotherapy Research. 2015;29(10):1488–1493. doi: 10.1002/ptr.5410.
    1. Cai D.-F., Dai W., Chen Y.-P., Wen M., Dai H.-L. Effect and mechanism of rhubarb on fibrinolysis in secondary damaged central nerve system of rats with acute hemorrhagic stroke. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2005;25(1):38–41.
    1. Tang Y.-P., Cai D.-F., Liu J. Research on acting mechanism of rhubarb on aquaporin-4 in rats with blood-brain barrier injury after acute cerebral hemorrhage. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2006;26(2):152–156.
    1. Gu J.-W., Hasuo H., Takeya M., Akasu T. Effects of emodin on synaptic transmission in rat hippocampal CA1 pyramidal neurons in vitro. Neuropharmacology. 2005;49(1):103–111. doi: 10.1016/j.neuropharm.2005.02.003.
    1. Liu T., Jin H., Sun Q.-R., Xu J.-H., Hu H.-T. Neuroprotective effects of emodin in rat cortical neurons against β-amyloid-induced neurotoxicity. Brain Research. 2010;1347:149–160. doi: 10.1016/j.brainres.2010.05.079.
    1. Legendre F., Bogdanowicz P., Martin G., et al. Rhein, a diacerhein-derived metabolite, modulates the expression of matrix degrading enzymes and the cell proliferation of articular chondrocytes by inhibiting ERK and JNK-AP-1 dependent pathways. Clinical and Experimental Rheumatology. 2007;25(4):546–555.
    1. Moldovan F., Pelletier J. P., Jolicoeur F.-C., Cloutier J.-M., Martel-Pelletier J. Diacerhein and rhein reduce the ICE-induced IL-1β and IL-18 activation in human osteoarthritic cartilage. Osteoarthritis and Cartilage. 2000;8(3):186–196. doi: 10.1053/joca.1999.0289.
    1. Su J., Yin L. P., Zhang X., Li B. B., Liu L., Li H. Chronic allograft nephropathy in rats is improved by the intervention of rhein. Transplantation Proceedings. 2013;45(6):2546–2552. doi: 10.1016/j.transproceed.2013.03.030.
    1. Zhou Y., Wang Y., Wang J., Anne Stetler R., Yang Q.-W. Inflammation in intracerebral hemorrhage: From mechanisms to clinical translation. Progress in Neurobiology. 2014;115(C):25–44. doi: 10.1016/j.pneurobio.2013.11.003.
    1. Lin S., Yin Q., Zhong Q., et al. Heme activates TLR4-mediated inflammatory injury via MyD88/TRIF signaling pathway in intracerebral hemorrhage. Journal of Neuroinflammation. 2012;9, article 46 doi: 10.1186/1742-2094-9-46.
    1. Ma C. X., Yin W. N., Cai B. W., et al. Toll-like receptor 4/nuclear factor-kappa B signaling detected in brain after early subarachnoid hemorrhage. Chinese Medical Journal. 2009;122:1575–1581.
    1. Teng Weiyu, Wang Lishu, Xue Weishuang, Guan Chao. Activation of TLR4-Mediated NFκB Signaling in Hemorrhagic Brain in Rats. Mediators of Inflammation. 2009;2009:6. doi: 10.1155/2009/473276.473276
    1. Sansing L. H., Harris T. H., Welsh F. A., Kasner S. E., Hunter C. A., Kariko K. Toll-like receptor 4 contributes to poor outcome after intracerebral hemorrhage. Annals of Neurology. 2011;70(4):646–656. doi: 10.1002/ana.22528.
    1. Holmin S., Mathiesen T. Intracerebral administration of interleukin-1β and induction of inflammation, apoptosis, and vasogenic edema. Journal of Neurosurgery. 2000;92(1):108–120. doi: 10.3171/jns.2000.92.1.0108.
    1. Wang J., Doré S. Inflammation after intracerebral hemorrhage. Journal of Cerebral Blood Flow & Metabolism. 2007;27(5):894–908. doi: 10.1038/sj.jcbfm.9600403.
    1. Wu J., Yang S., Xi G., Fu G., Keep R. F., Hua Y. Minocycline reduces intracerebral hemorrhage-induced brain injury. Neurological Research. 2009;31(2):183–188. doi: 10.1179/174313209X385680.
    1. Zhao F., Hua Y., He Y., Keep R. F., Xi G. Minocycline-induced attenuation of iron overload and brain injury after experimental intracerebral hemorrhage. Stroke. 2011;42(12):3587–3593. doi: 10.1161/STROKEAHA.111.623926.
    1. Szymanska A., Biernaskie J., Laidley D., Granter-Button S., Corbett D. Minocycline and intracerebral hemorrhage: Influence of injury severity and delay to treatment. Experimental Neurology. 2006;197(1):189–196. doi: 10.1016/j.expneurol.2005.09.011.
    1. Wasserman J. K., Schlichter L. C. Neuron death and inflammation in a rat model of intracerebral hemorrhage: Effects of delayed minocycline treatment. Brain Research. 2007;1136(1):208–218. doi: 10.1016/j.brainres.2006.12.035.
    1. Bullock R., Mendelow A. D., Teasdale G. M., Graham D. I. Intracranial haemorrhage induced at arterial pressure in the rat. Part 1: Description of technique, ICP changes and neuropathological findings. Neurological Research. 1984;6(4):184–188. doi: 10.1080/01616412.1984.11739687.
    1. Xi G., Keep R. F., Hoff J. T. Pathophysiology of brain edema formation. Neurosurgery Clinics of North America. 2002;13(3):371–383. doi: 10.1016/S1042-3680(02)00007-4.
    1. Keep R. F., Hua Y., Xi G. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. The Lancet Neurology. 2012;11(8):720–731. doi: 10.1016/s1474-4422(12)70104-7.

Source: PubMed

3
Subscribe