Journal Home > Volume 1 , Issue 2–4

Stroke is a serious acute cerebrovascular disease attributable to disruptions in the blood supply to the brain tissue as a result of vascular obstruction or sudden rupture of blood vessels in the brain, which further result in hypoxia of the brain and reduction of necessary nutrients, apoptosis of neurons, and damage to brain tissue. The majority of stroke patients are ischemic stroke. The main clinical treatments for ischemic stroke include medical thrombolysis in the early stage of onset and surgical thrombectomy or stent implantation in the late stage of onset, all of which have their own indications, advantages, and disadvantages, and show limited clinical application. For cerebral ischemia-reperfusion injury with an extremely poor prognosis, there is currently no effective prevention and treatment method in the clinic. Therefore, timely and effective treatment is needed to treat cerebral ischemia-reperfusion injury. An increasing number of studies have shown that natural products have a good curative effect on cerebral ischemia-reperfusion injury. However, due to their low solubility, low bioavailability, and short half-life, many natural products cannot optimally exert their curative effects on cerebral ischemia-reperfusion injury. Natural products-based nanoparticles modified with specific ligands have attracted much attention because of their high-efficiency permeation through the blood–brain barrier, targeted delivery abilities, and the protection of the active components from degradation. Therefore, this review focused on the prevention and treatment of cerebral ischemia-reperfusion injury in the natural product-based nanoparticles.


menu
Abstract
Full text
Outline
About this article

Advances in the therapy of cerebral ischemia-reperfusion injury with natural product-based nanoparticles

Show Author's information Qingchun Mu1,§Yuhao Xue2,§Yuting Lu3Yufei Zhang4Qian Cheng5Jinlong Wan6Peng Liu7Jingjing Liu8yikun Qu9Chunming Huang6( )Longguang Tang3( )
Department of Neurosurgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570100, China
College of Life Science, Mudanjiang Medical University, Mudanjiang 157009, China
International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of medicine, Yiwu 322000, China
Department of Physiology, Guilin Medical University, Guilin 541199, China
College of Pharmacy, Guilin Medical University, Guilin 541199, China
The People's Hospital of Gaozhou, Maoming 525200, China
Department of Clinical Medicine, Jiamusi University, Jiamusi 154007, China
Department of Basic Medicine, Jiamusi University, Jiamusi 154007, China
Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi 154002, China

§ Qingchun Mu and Yuhao Xue contributed equally to this work.

Abstract

Stroke is a serious acute cerebrovascular disease attributable to disruptions in the blood supply to the brain tissue as a result of vascular obstruction or sudden rupture of blood vessels in the brain, which further result in hypoxia of the brain and reduction of necessary nutrients, apoptosis of neurons, and damage to brain tissue. The majority of stroke patients are ischemic stroke. The main clinical treatments for ischemic stroke include medical thrombolysis in the early stage of onset and surgical thrombectomy or stent implantation in the late stage of onset, all of which have their own indications, advantages, and disadvantages, and show limited clinical application. For cerebral ischemia-reperfusion injury with an extremely poor prognosis, there is currently no effective prevention and treatment method in the clinic. Therefore, timely and effective treatment is needed to treat cerebral ischemia-reperfusion injury. An increasing number of studies have shown that natural products have a good curative effect on cerebral ischemia-reperfusion injury. However, due to their low solubility, low bioavailability, and short half-life, many natural products cannot optimally exert their curative effects on cerebral ischemia-reperfusion injury. Natural products-based nanoparticles modified with specific ligands have attracted much attention because of their high-efficiency permeation through the blood–brain barrier, targeted delivery abilities, and the protection of the active components from degradation. Therefore, this review focused on the prevention and treatment of cerebral ischemia-reperfusion injury in the natural product-based nanoparticles.

Keywords: stroke, nanoparticles, drug delivery, cerebral ischemia-reperfusion injury, natural products

References(83)

[1]

Zhou, Z. Y.; Cai, G. Internal Medicine of Traditional Chinese Medicine; 2nd ed. People's Medical Publishing House: Beijing, 2008.

[2]

Jiang, B. Suggestions for domestic primary stroke care arising from epidemiological characteristics, prevention and treatment of stroke in China. Chin. Gen. Pract. 2017, 22, 3653–3661. (in Chinese)

[3]
Wei, Y. Stroke in Chinese aged 40 years and older. 2016.
[4]

Yi, D. R.; Wang, P., Qian, Y. G.; Xia, Y. J. Epidemiological characteristics of 7472 cases of stroke in Inner Mongolia autonomous region. Dis. Surveill. 2022, 37, 464–468. (in Chinese)

[5]

Knight-Greenfield, A.; Nario, J. J. Q.; Gupta, A. Causes of acute stroke: A patterned approach. Radiol. Clin. North Am. 2019, 57, 1093–1108.

[6]
Shan, G. Etiology diagnosis of acute phase of ischemic brain. In The 13th National Neurology Academic Conference of Chinese Medical Association, Chengdu, China, 2010, pp 1. (in Chinese)
[7]

Li, Z. J. Surgical treatment of hypertensive cerebral hemorrhage. Chin. J. Med. 2020, 55, 354–356.

[8]

Wu, L. R.; Zhao, Z. H.; Ying, S. Y.; Zhou, X. D. Effect of alteplase in thrombolytic therapy for treating ischemic stroke of different etiologies. Health Res. 2018, 38, 447–449.

[9]

Gu, J. X.; An, Z. X.; Wang, Y.; Zhao, L.; Lü, J. P.; Zhu, J. G. Observation on the effect of alteplase combined with agatroban in the treatment of acute ischemic stroke and its influence on neurological function and inflammatory factors. Clin. Misdiagn. Misther. 2019, 32, 44–49.

[10]

Cheng, C. C.; Tu, H. M.; Ying, A. J.; Xu, G. Q.; Hu, L. Z.; Pan, J. Z. Clinical efficacy and mechanism of alteplase combined with butylphthalide sodium chloride on patients with acute cerebral infarction. Chin. J. Clin. Pharmacol. 2015, 31, 2293–2296. (in Chinese)

[11]

Rabinstein, A. A. Treatment of acute ischemic stroke. Continuum 2017, 23, 62–81.

[12]

Sun, Y. Y.; Peng, Z. Y.; Zhao, Y. X.; Kong, F. B.; Wang, H. Research progress on anti-inflammatory effect of curcumin in disease treatment. Med. Innovat. China 2021, 18, 181–184. (in Chinese)

[13]

Hu, J. Study on biomedical function of curcumin extracted from turmeric (Curcuma longa L. ). Mol. Plant Breed. 2022, 20, 683–688. (in Chinese)

[14]

Chen, J. P.; LI, L.; Su, J. Y. Antioxidant and antitumor activities of curcumin. Mod. Food Sci. Technol. 2014, 30, 11–15, 6. (in Chinese)

[15]

Bu, Y. H.; Lu, T.; Wu, H.; Sun, M. H.; Zhang, H.; Deng, R.; Wang, Y. Research progress on chemical constituents and pharmacological action of cabinet. J. Anhui Univ. Chin. Med. 2020, 39, 89–93. (in Chinese)

[16]
Dong, L. Brain Protective Effect and mechanism of Auriculturin on focal cerebral ischemia in mice. 2015.
[17]

Long, Y.; Zhang, Y. L.; Wan, J. Y.; Liu, S. Y.; Ni, L.; Li, N.; Yang, M. Research progress on traditional Chinese medicine in treatment of multiple organs injury caused by cerebral ischemic stroke. Chin. Tradit. Herb. Drugs 2021, 52, 2106–2116. (in Chinese)

[18]

Siesjö, B. K.; Katsura, K.; Mellergård, P.; Ekholm, A.; Lundgren, J.; Smith, M. L. Acidosis-related brain damage. Prog. Brain Res. 1993, 96, 23–48.

[19]

Zhang, A. J.; Wang, S.; Wang, P.; Wang, Y. R. Progress in pathological mechanism of ischemic stroke and prevention and treatment of traditional Chinese medicine. Chin. J. Exp. Tradit. Med. Form. 2020, 26, 227–240. (in Chinese)

[20]

Jokivarsi, K. T.; Gröhn, H. I.; Gröhn, O. H.; Kauppinen, R. A. Proton transfer ratio, lactate, and intracellular pH in acute cerebral ischemia. Magn. Reson. Med. 2007, 57, 647–653.

[21]

Wang, R. R.; W. S., Du Guanhua. The role of endoplasmic reticulum in ischemic brain. Chin. Pharmacol. Bull. 2019, 35, 761–765. (in Chinese)

[22]
Zhao, H. Effects and mechanisms of the immunity and inflammations in stroke animal models. Ph. D. Dissertation, Shanxi Medical University, Shanxi, 2020.
[23]

Kaviarasi, S.; Yuba, E.; Harada, A.; Krishnan, U. M. Emerging paradigms in nanotechnology for imaging and treatment of cerebral ischemia. J. Control. Release 2019, 300, 22–45.

[24]

Tianli, G. The clinical application of different drugs in patients with episodic ischemic stroke and intravenous thrombolytic therapy. Ya Jian: European Journal of Acute Ischemic Stroke, 2021 Guidelines for Intravenous Thrombolysis read 2021. Pract. J. Cardiac Cereb. Pneum. Vasc. Dis. 2021, 29, 1–8. (in Chinese)

[25]

Salman, M.; Tabassum, H.; Parvez, S. Nrf2/HO-1 mediates the neuroprotective effects of pramipexole by attenuating oxidative damage and mitochondrial perturbation after traumatic brain injury in rats. Dis. Model. Mech. 2020, 13, dmm045021.

[26]

Andrabi, S. S.; Ali, M.; Tabassum, H.; Parveen, S.; Parvez, S. Pramipexole prevents ischemic cell death via mitochondrial pathways in ischemic stroke. Dis. Model. Mech. 2019, 12, dmm033860.

[27]

Iadecola, C.; Anrather, J. The immunology of stroke: From mechanisms to translation. Nat. Med. 2011, 17, 796–808.

[28]

Wang, X. P.; Ni, J. M. Advance in research of therapeutic medicines for cerebral ischemia-reperfusion injury. Chin. J. New Drugs 2016, 25, 659–663, 691. (in Chinese)

[29]

Gao, L.; Wu, L. P.; Shi, Z. G.; Liu, L. N.; Shang, J. Research progress on effect of traditional Chinese medicine on blood-brain barrier permeability. Chin. J. Exp. Tradit. Med. Form. 2019, 25, 200–207. (in Chinese)

[30]

Long, L. L.; Xiao, B. Nanoparticles: A targeted vector for drug delivery across the blood-brain barrier. J. Int. Neurol. Neuros. 2006, 33, 308–311.

[31]

Zhao, Y.; Cao, W. Q.; Liu, Y. Recent advances in polymeric nano-sized carrier systems. Chem. J. Chin. Univ. 2020, 41, 909–923. (in Chinese)

[32]

Liu, Y.; Jiang, C. Brain targeting by nanodrug delivery system. Acta Pharm. Sin. 2013, 48, 1532–1543.

[33]

Zhang, C.; Ling, C. L.; Pang, L.; Wang, Q.; Liu, J. X.; Wang, B. S.; Liang, J. M.; Guo, Y. Z.; Qin, J.; Wang, J. X. Direct macromolecular drug delivery to cerebral ischemia area using neutrophil-mediated nanoparticles. Theranostics 2017, 7, 3260–3275.

[34]

Shi, K. B.; Tian, D. C.; Li, Z. G.; Ducruet, A. F.; Lawton, M. T.; Shi, F. D. Global brain inflammation in stroke. Lancet Neurol. 2019, 18, 1058–1066.

[35]

Dong, X. Y.; Gao, J.; Zhang, C. Y.; Hayworth, C.; Frank, M.; Wang, Z. J. Neutrophil membrane-derived nanovesicles alleviate inflammation to protect mouse brain injury from ischemic stroke. ACS Nano 2019, 13, 1272–1283.

[36]
Zhao, T. Construction of a Lactoferrin-Astaxanthin emulsion delivery system and its intervention effect on neuroinflammation and cognitive dysfunction. Ph. D. Dissertation, Northwest A & F University, Yangling, 2021.
[37]

Meng, H. L.; Zhao, H. R.; Cao, X.; Hao, J. W.; Zhang, H.; Liu, Y.; Zhu, M. S.; Fan, L. Z.; Weng, L. H.; Qian, L. et al. Double-negative T cells remarkably promote neuroinflammation after ischemic stroke. Proc. Natl. Acad. Sci. USA 2019, 116, 5558–5563.

[38]

Sarker, K. P.; Biswas, K. K.; Yamakuchi, M.; Lee, K. Y.; Hahiguchi, T.; Kracht, M.; Kitajima, I.; Maruyama, I. ASK1-p38 MAPK/JNK signaling cascade mediates anandamide-induced PC12 cell death. J. Neurochem. 2003, 85, 50–61.

[39]

An, S.; Kuang, Y. Y.; Shen, T.; Li, J. F.; Ma, H. J.; Guo, Y. B.; He, X.; Jiang, C. Brain-targeting delivery for RNAi neuroprotection against cerebral ischemia reperfusion injury. Biomaterials 2013, 34, 8949–8959.

[40]

Bai, X. J.; Shi, X. B., Jiang, L.; Sun, B.; Wu, W. P. Research progress in stem cell therapy for ischemic stroke. Chin. J. Geriatr. Heart Brain Ves. Dis. 2022, 24, 553–555. (in Chinese)

[41]

Liao, Y. D.; Wang, Z. L.; Chen, G. T.; Wang, X. D.; Xu, K. Y. Research progress on stem cell transplantation for ischemic stroke. J. Pract. Med. 2021, 37, 2817–2821. (in Chinese)

[42]

Liu, W. J.; Zhang, G. L.; Wu, J. R.; Zhang, Y. L.; Liu, J.; Luo, H. Y.; Shao, L. Q. Insights into the angiogenic effects of nanomaterials: Mechanisms involved and potential applications. J. Nanobiotechnol. 2020, 18, 9.

[43]

Yao, M. H.; Shi, X. J.; Zuo, C. J.; Ma, M.; Zhang, L.; Zhang, H. B.; Li, X.; Yang, G. Y.; Tang, Y. H.; Wu, R. Engineering of SPECT/photoacoustic imaging/antioxidative stress triple-function nanoprobe for advanced mesenchymal stem cell therapy of cerebral ischemia. ACS Appl. Mater. Interfaces 2020, 12, 37885–37895.

[44]

Li, G. Q.; Yang, F. B.; Yu, Y.; Xu, Y. L.; Zhang, Y. N. Research progress of curcumin nanoparticles in central nervous system diseases. Chin. J. Mod. Appl. Pharm. 2022, 39, 403–409. (in Chinese)

[45]

Xie, J. J.; Zhang, D. S.; Jing, Y. S. Neuroprotective effect of curcumin on cerebral ischemia reperfusion injury and its mechanism. Chin. J. Pharm. Toxicol. 2021, 35, 672. (in Chinese)

[46]

Li, G.; Xia, Z. Study on the effect of ourcumin on inflammation actions and blood-brain barrier permeability in rats with cerebral ischemia/reperfusion injury. Mod. J. Integr. Tradit. Chin. Western Med. 2015, 24, 814–816, 877. (in Chinese)

[47]

Yuan, Y. H.; Guang, Q.; Zhang, S. S.; Tang, M. Preparation of curcumin TPP-PEG-PE nanomicelles with mitochondrial targeting and lysosomal escape functions and its effect on promoting breast cancer cell apoptosis. China J. Chin. Mater. Med. 2020, 45, 5495–5503. (in Chinese)

[48]

Zhang, X. L.; Li, Y. P.; Lü, S. W.; Wang, Y. H.; Li, Y. J. Research progress of curcumin nanocarriers and their applications. Contempor. Chem. Ind. 2021, 50, 2685–2688. (in Chinese)

[49]

Kakkar, V.; Muppu, S. K.; Chopra, K.; Kaur, I. P. Curcumin loaded solid lipid nanoparticles: An efficient formulation approach for cerebral ischemic reperfusion injury in rats. Eur. J. Pharm. Biopharm. 2013, 85, 339–345.

[50]

Wang, Y.; Luo, J.; Li, S. Y. Nano-curcumin simultaneously protects the blood-brain barrier and reduces M1 microglial activation during cerebral ischemia-reperfusion injury. ACS Appl. Mater. Interfaces 2019, 11, 3763–3770.

[51]

Wei, L.; Jiao, Y. P., Li, N.; Li, Y. Preparation of temperature responsive hydrogel and its controlled release of curcumin. J. Shaanxi Univ. Chin. Med. 2022, 45, 72–76. (in Chinese)

[52]

Chen, B. X., Liu, Y. N.; Li, H. Y.; Zhao, M. J.; Tian, H.; Song, J.; Zhang, Q.; Wang, M.; Liu, F. Neuroprotective effect of puerarin on ischemic brain injury in mice by antioxidant stress. Clin. Basic Bridg. Res. 2021, 37, 1355–1357, 1362.

[53]

Huang, X. F.; Wang, J. M. Research progress of neuroprotective mechanisms of puerarin. Chin. J. Exp. Tradit. Med. Form. 2015, 21, 224–230. (in Chinese)

[54]

Liu, W.; Lu, H. W.; Rao, X. Y.; Li, X.; Lu, H. D.; Li, F. F.; He, Y.; Yu, R. Y.; Zhong, R. S.; Zhang, Y. et al. Enhanced treatment for cerebral ischemia-reperfusion injury of puerarin loading liposomes through neutrophils-mediated targeted delivery. Nano Res. 2021, 14, 4634– 4643.

[55]

Tao, H. Q.; Cheng, G. S.; Zhang, C. M.; Tang, S. K.; Jiang, L.; Jiang, J. Q.; Liu, X. Y.; Guo, L. Effects of brain ischemia-reperfusion injury by HP-β-CD-PLGA nanoparticles in rats. J. Harbin Med. Univ. 2021, 55, 349–354. (in Chinese)

[56]
Zhang, J. W. The effect and mechanism of puerarin hydrogel on MSCs transplantation in the treatment of myocardial infarction. Ph. D. Dissertation, Southern Medical University, Guangzhou, 2016.
[57]

Yang, G.; Qian, C.; Wang, N.; Lin, C. Y.; Wang, Y.; Wang, G. Y.; Piao, X. Tetramethylpyrazine protects against oxygen-glucose deprivation-induced brain microvascular endothelial cells injury via Rho/Rho-kinase signaling pathway. Cell Mol. Neurobiol. 2017, 37, 619–633.

[58]

Zhang, H.; Tang, W. W.; Wang, S.; Zhang, J. H.; Fan, X. Tetramethylpyrazine inhibits platelet adhesion and inflammatory response in vascular endothelial cells by inhibiting P38 MAPK and NF-κB signaling pathways. Inflammation 2020, 43, 286–297.

[59]

He, J. H.; Wen, M.; Shi, X. H.; Zhao, Q. H. Splitting and eastward withdrawal of the subtropical high belt during the onset of the South China Sea summer monsoon and their possible mechanism. J. Nanjing Univ. (Nat. Sci. ) 2002, 38, 318–330.

[60]

Hao, C. J.; M. L., Li Ang, Lu Tianyuan, Kong Qinglan. Preparation of Ligustrazine solid lipid nanoparticles for oral use. J. Tianjing Uni. Chin. Med. 2007, 96. (in Chinese)

[61]

Liu, X. Y.; Fan, Q.; Zhao, C. Y.; Shao, M. K.; Ma, H.; Cheng, L. C.; A, R. B. L. G.; Yu, Z. L. Reversal effect of peptide-modified chitosan tetramethylpyrazine nanoparticles on multidrug resistance in tumor cells. China J. Chin. Mater. Med. 2020, 45, 5487–5494. (in Chinese)

[62]

Ji, S. L.; Ni, H.; He, L. Preparation and in vitro evaluation of ligustrazine-loaded solid lipid nanoparticles. J. Shenyang Pharm. Univ. 2017, 34, 629–633, 639. (in Chinese)

[63]

Ming Xie, J. Z., Jiayuan Qiu, wenhaifeng Zeng, wengthen Zeng, Xu Liu. Effects of gastrodin on neural repair and aquaporin 4 in rats with cerebral ischemia-reperfusion injury. Chin. J. Neuroanat. 2022, 38, 65–73. (in Chinese)

[64]

Liu, B.; Li, F.; Shi, J. S.; Yang, D. L.; Deng, Y. Y.; Gong, Q. H. Gastrodin ameliorates subacute phase cerebral ischemiareperfusion injury by inhibiting inflammation and apoptosis in rats. Mol. Med. Rep. 2016, 14, 4144–4152.

[65]

Mei, H. Study on the preparation and drug release in vitro of gastrodin nanoliposomes. China Pharm. 2011, 14, 480–483. (in Chinese)

[66]

Du, X. S.; Pu, X. Y. Preparation, characterization and in vitro release properties of gastrodin tamarind gum chitosan sustained release microspheres. Gansu Agric. 2015, (18), 44–47.

[67]

Liu, R.; Li, X. D. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: An review of their molecular mechanisms. Chin. J. Med. Guide 2019, 21, 749–752. (in Chinese)

[68]

Assini, J. M.; Mulvihill, E. E.; Huff, M. W. Citrus flavonoids and lipid metabolism. Curr. Opin. Lipidol. 2013, 24, 34–40.

[69]

Benavente-García, O.; Castillo, J. Update on uses and properties of citrus flavonoids: New findings in anticancer, cardiovascular, and anti-inflammatory activity. J. Agric. Food Chem. 2008, 56, 6185–6205.

[70]

Jiang, J. G.; Wu, F. F.; Tian, W. Q. Experimental study on the improvement of learning and memory ability of vascular dementia mice by hesperidin-loaded nanoliposomes. China Pharm. 2021, 24, 1641–1646. (in Chinese)

[71]

Gu, S. F.; Wang, L. Y.; Tian, Y. J.; Zhou, Z. X.; Tang, J. B.; Liu, X. R.; Jiang, H. P.; Shen, Y. Q. Enhanced water solubility, antioxidant activity, and oral absorption of hesperetin by D-α-tocopheryl polyethylene glycol 1000 succinate and phosphatidylcholine. J. Zhejiang Univ. Sci. B 2019, 20, 273–281. (in Chinese)

[72]

Moghaddam, A. H.; Pour, Y. S.; Sangdehi, S. R. M.; Hasantabar, V. Evaluation of hesperetin-loaded on multiple wall carbon nanotubes on cerebral ischemia/reperfusion injury in rats. Biomed. Pharmacother. 2021, 138, 111467.

[73]

Zhang, X.; Peng, F. Q.; He, F. L.; Liu, M. Risk control of traditional Chinese medicines containing Triptolide. Chin. Tradit. Patent Med. 2019, 41, 1667–1671. (in Chinese)

[74]

Cheng, T.; Gao, N.; Zheng, X. L.; Ying, M. Y. Molecular mechanism of triptolide-induced toxicity to Chinese hamster ovary cells based on transcriptomics. Nat. Prod. Res. Dev. 2021, 33, 1836–1844, 1968.

[75]

Bai, S.; Hu, Z. Y.; Yang, Y.; Yin, Y. F.; Li, W. Y.; Wu, L. J.; Fang, M. R. Anti-inflammatory and neuroprotective effects of triptolide via the NF-κB signaling pathway in a rat MCAO model. Anat. Rec. 2016, 299, 256–266.

[76]

Wan, Y. S.; You, Y.; Ding, Q. Y.; Xu, Y. X.; Chen, H.; Wang, R. R.; Huang, Y. W.; Chen, Z.; Hu, W. W.; Jiang, L. Triptolide protects against white matter injury induced by chronic cerebral hypoperfusion in mice. Acta Pharmacol. Sin. 2022, 43, 15–25.

[77]
Zhang, C. Studies on triptolide-loaded lipid nanoparticles. Ph. D. Dissertation, Huazhong University of Science and Technology, Wuhan, 2014.
[78]

Li, P.; Yang, X. Y.; Yang, Y.; He, H. M.; Chou, C. K.; Chen, F. Y.; Pan, H.; Liu, L. L.; Cai, L. T.; Ma, Y. F. et al. Synergistic effect of all-trans-retinal and triptolide encapsulated in an inflammation-targeted nanoparticle on collagen-induced arthritis in mice. J. Control. Release 2020, 319, 87–103.

[79]

Zheng, Y.; Zhang, W. J.; Wang, X. M. Triptolide with potential medicinal value for diseases of the central nervous system. CNS Neurosci. Ther. 2013, 19, 76–82.

[80]
al, Y. Q. e. Effect of Geniposide iridoid glycosides on angiogenesis in rats with intracerebral hemorrhage. 2010.
[81]

Cheng, C. Y.; Ho, T. Y.; Hsiang, C. Y.; Tang, N. Y.; Hsieh, C. L.; Kao, S. T.; Lee, Y. C. Angelica sinensis exerts angiogenic and anti-apoptotic effects against cerebral ischemia-reperfusion injury by activating p38MAPK/HIF-1α/VEGF-A signaling in rats. Am. J. Chin. Med. 2017, 45, 1683–1708.

[82]
Huang, S. Z. Neuroprotection of emodin against middle cerebral artery occlusion injury in rats and its mechanism. Ph. D. Dissertation, Fujian University of Traditional Chinese Medicine, Fuzhou, 2018.
[83]

Caixia Jia, J. C., Xiaohan POM, Guang Gao, Jingzhong Li, Feilong Zhang, Jinping Wang, Wei Wang, Xiaoxin Xu and Huihui Zhao. Prediction of the mechanism of action of emodin in cerebral. Pharmacotherapie 2021, 138, 111467.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 01 July 2022
Revised: 08 August 2022
Accepted: 12 August 2022
Published: 13 October 2022
Issue date: December 2022

Copyright

© The Author(s) 2022. Nano TransMed published by Tsinghua University Press.

Acknowledgements

Acknowledgements

The authors are grateful for grants received from Zhejiang Provincial Program for the Cultivation of High-Level Innovative Health Talents.

Rights and permissions

The articles published in this open access journal are distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

Return