Skip to main content

MINI REVIEW article

Front. Neurol., 05 June 2018
Sec. Stroke

Therapeutic Values of Human Urinary Kallidinogenase on Cerebrovascular Diseases

\r\nZhenyu Wei&#x;Zhenyu Wei1Yi Lyu&#x;Yi Lyu2XiaoLi Yang&#x;XiaoLi Yang1Xin ChenXin Chen1Ping ZhongPing Zhong3Danhong Wu*Danhong Wu1*
  • 1Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
  • 2Department of Medical Affairs, Techpool Bio-Pharma Co., Ltd., Guangzhou, China
  • 3Department of Neurology, Shanghai TCM Integrated Hospital affiliated to Shanghai University of Chinese Medicine, Shanghai, China

The term “tissue kallikrein” is used to describe a group of serine proteases shared considerable sequence homology and colocalize in the same chromosomal locus 19q13. 2–q13.4. It has been widely discovered in various tissues and has been proved to be involved in kinds of pathophysiological processes, such as inhibiting oxidative stress, inflammation, apoptosis, fibrosis and promoting angiogenesis, and neurogenesis. Human Urinary Kallidinogenase (HUK) extracted from human urine is a member of tissue kallikrein which could convert kininogen to kinin and hence improve the plasma kinin level. Medical value of HUK has been widely investigated in China, especially on acute ischemic stroke. In this review, we will summarize the therapeutic values of Human Urinary Kallidinogenase on acute ischemic stroke and its potential mechanisms.

Introduction

The term “tissue kallikrein” is used to describe a series of acid glycoprotein with highly similar gene and protein structure, which also share considerable sequence homology and colocalize in the same chromosomal locus 19q13.2–q13.4 (1, 2). Tissue Kallikrein is encoded by one of the kallikrein gene family, KLK1 gene. Originally, it was thought that the gene family included three genes, namely prostate-specific antigen, tissue kallikrein, and glandular kallikrein 2 (3). In 2000, other 12 kallikrein-related genes were found, and, until now, 15 tissue kallikrein genes have been discovered (4).

Tissue kallikreins release vasoactive peptides from low molecular weight kininogen. The peptides, mainly including kinin, trigger multiple biological functions by activating two types of G protein-coupled receptors named as B1 receptor and B2 receptor respectively (5). Tissue kallikrein is widely distributed such as in kidney, blood vessels, central nervous system, pancreas, gut, neutrophils, and so on (6). HUK is one of the tissue kallikreins that has been found in human urine. Urine product has been believed to have medical value for centuries. Galen, Greek physicians used urine product to cure inflammation, burns and skin disease, and in Japan, urine product was used to cure asthma, hypertension, and diabetes (7). The potential therapeutic value of urine reminds us that tissue kallikrein may also be expected to have therapeutic effects. But previous studies found that protective role of tissue kallikrein does not work on all types of hypertension, and that tissue kallikrein is expected to be useful for treatment of salt-sensitive hypertension (8, 9). Now in China, HUK has been used for acute ischemic stroke and recent studies has found that HUK also exert multiple functions such as attenuating brain injury by inhibiting oxidative stress and promoting angiogenesis and neurogenesis. HUK also has clinical application on cardiac and renal damage, vascular restenosis, ischemic stroke, skin wound injury, and hypertension. In this review, we summarized therapeutic values of HUK on cerebrovascular diseases by the results from clinical trials and animal experiments.

Result From Clinical Trials

HUK Improves Neurological Function of Patients With Ischemic Stroke

In a case–control study with 1,268 stroke patients, plasma tissue kallikrein levels were negatively related to the risk of first-ever stroke and stroke recurrence, which suggested that lower plasma tissue kallikrein levels are independently correlated to first-ever stroke and are an independent predictive factor of stroke recurrence (10). Tissue kallikrein is more likely involved to the pathological process of stroke. In ischemic stroke, HUK application significantly decreases National Institute of Health Stroke Scale (NIHSS) and modified Rankin Scale (mRS) scores and improves long-term outcomes (11, 12). It was discovered that HUK was able to significantly improve the NIHSS scores of cerebral ischemia resulted from large-artery atherosclerosis and small-artery atherosclerosis, the results from the study on the clinical efficacy of HUK in the treatment of ischemic stroke according to TOAST classification (13). In the studies of combined medicine, HUK may improve the curative effect of rt-PA in patients with acute ischemic stroke (AIS) (14), and improve therapeutic effect of massive cerebral infarction when combined with edaravone (15).

After treatment with HUK, AIS patents' cerebral blood perfusion was significantly enhanced and mean transit time of perfusion was remarkably shorten, which are in accordance with increasing serum levels of VEGF and apelin (16). These two substances are all involved in vascular formation and maturation (17, 18). In a small sample clinical research performed in acute ischemic stroke patients treated with HUK or not, microcirculation was evaluated by magnetic resonance perfusion weighted imaging (MRP). It showed HUK could increase cerebral blood flow and ameliorate neurological deficits (19). Moreover, Wang et al found that HUK remarkably increased ipsilesional sensorimotor cortex (SMC) blood volume in AIS patients (11). The effect of HUK on promoting cerebral reorganization might be an critical mechanism in the treatment of acute cerebral infarction. In a small sample research, Song et al. discovered that Human Urinary Kallidinogenase improved symptoms of neurological dysfunction by promoting remodeling of long-term cortical motor function in patients with ischemic stroke as well (20).

HUK Prevent Cerebrovascular Restenosis After Endovascular Therapy

The endovascular therapy study enrolled two groups of patients with symptomatic middle cerebral artery stenosis. Among them, HUK intervention was administered in one group and the other group as control. Carefully follow-up and evaluation for restenosis showed HUK could prevent restenosis after in-stent in middle cerebral artery (21, 22). For patients with cerebral arterial stenosis and without stroke treated by HUK, cognitive status could be improved accompanied by Aβ1-40 serum levels decreasing at 4 weeks after treatment and Aβ1-42 serum levels decreasing remarkably at 8 weeks after treatment (23). The potential mechanism of preventing restenosis was to inhibit atherosclerosis formation and to attenuate intimal hyperplasia by downregulating Smad2/3 phosphorylation and TGF-β1 expression, promoting eNOS activity (24).

Safety Assessment of Human Urinary Kallidinogenase

To assess the safety of HUK in treating patients with acute ischemic stroke, a systematic review on randomized controlled clinic trials about HUK has been performed, and all the data has been gotten from the Chinese Stroke Trials Register, the Cochrane Stroke Group Trials Register, CENTRAL, Medline, EMBASE, the China Biological Medicine Database (CBM), and the China National Knowledge Infrastructure (CNKI) (12). The research included 24 trials involving 2433 patients, in which 15 trials reported that the common adverse event was transient hypotension, and there was no difference between the treatment group and control group on non-fatal intracerebral hemorrhage. A multi-center, open label, single group study has been designed and registered in ClinicalTrials.gov to reevaluate the safety and efficacy of HUK on acute ischemic stroke, and the study expects to enroll 60 sites and 2186 subjects (25).

Results From Animal Experiments

Angiogenesis and Enhanced Neurogenesis

It has been proved that HUK promoted angiogenesis and enhanced neurogenesis after ischemia/reperfusion (I/R) induced injury (26). Another animal experiment demonstrates that HUK treatment significantly increased the number of BrdU(+) cells in the subventricular zone (SVZ) and the peri-infarction region at 3 d after middle cerebral artery occlusion (MCAO), noteworthily, the number of BrdU(+)/DCX(+) cells and BrdU(+)/nestin(+) cells in the SVZ as well as vascular density in the peri-infarction region increased more significantly than vehicle group at 3 d, peaked at 7–14 d. HUK increased the number of BrdU(+)/NeuN(+) cells in the peri-infarction region at 14 and 28 d after MCAO compared with the vehicle group (27). However,in an animal experiment study about choroidal neovascularization (CNV), pretreatment with HUK could significantly decrease laser-induced CNV size (28).

Inflammation and Apoptosis

A continuous infusion of a sub-depressor dose of HUK by minipump decreased I/R-induced neurological disorders and brain infarction, inflammation, and oxidative stress, which suggested that HUK also acted as an anti-oxidative and anti-inflammatory agent in protecting the brain from ischemic stroke induced damages (29). Yang et al. found that HUK improves neurological deficits, reduces the infarct volume, and decreases inflammatory responses in rats following cerebral ischemia. Meanwhile, they also found that the levels of TLR4 and NF-kB in ischemic brains increased following MCAO by western blot analysis, and this effect was significantly inhibited by immediate treatment of HUK (30). It concluded that tissue kallikrein protects against ischemic stroke through antioxidation and anti-inflammation by suppressing TLR4/NF-κB signaling pathway in rats.

Transforming growth factor beta 1 (TGF-β1), as an immunomodulator, can attenuate inflammation definitely, and previous studies confirmed that TGF-β1 limited neuroinflammation and that activated the expression of Bcl-2 to suppress the apoptosis of neurons in acute ischemic stroke rats (31, 32). HUK treatment can highly increase the expression of TGF-β1 in rats with focal ischemic injury, meanwhile, decrease high-sensitivity c-reactive protein expression, a systemic inflammation indicator (33). Glutamate-induced neurotoxicity induced oxidative injury and was involved in the pathogenesis of a series of central nervous system disorders. Previous studies found that HUK could largely inhibit glutamate-induced cell death and morphological changes of cultured cortical neurons by inhibiting of ROS production and attenuating nNOS activity to inhibit glutamate-induced nitrosative stress through an intracellular signaling pathway including activation of B2R, ERK1/2, and NFkB, and up-regulation of BDNF and Bcl-2 expression (3436). Jingjing Su et al. demonstrated that HUK could activate ERK1/2 signaling cascades through Homer1b/c (37). Xia et al. proved that local or systemic delivery of the tissue kallikrein gene attenuates cerebral I/R injury in mouse models by inhibiting oxidative stress and apoptosis through activation of B2R (26). Besides, B2R-dependent regulation of autophagy is involved in inhibiting oxygen and glucose deprivation-induced neuronal cells injury (38).

Except for neuroprotective effects, it has been demonstrated that HUK might prevent and ameliorate diabetes-induced nephropathy and reduced renal inflammation, oxidative stress and renal fibrosis (39). The activation of the kallikrein-kinin system was also found in the experiment. However, some evidences suggested that kinins may play an important role in the development of diabetic retinopathy by enhancing vascular permeability, leukocytes infiltration, and inflammatory response through mediating kinin B1 and B2 receptors (40).

Conclusions

Recent animal experiments and clinic trials proved that HUK has enormous therapeutic value in acute ischemic stroke. In patients treated with HUK, the enhanced microcirculation and cerebral blood flow may promote angiogenesis and neurogenesis and inhibit inflammation and apoptosis. However, recent optimal results were from small sample clinical trials, the mechanism of HUK has not been clear enough, evidences of the safety with HUK are still absence, more animal experiments and multicenter-clinical trials are necessary in the future.

Author Contributions

ZW and YL prepared the references and designed the structure of this paper. ZW, XY, and XC prepared the manuscript. PZ and DW reviewed and revised this article.

Conflict of Interest Statement

YL was employed by company Techpool Bio-Pharma Co.,Ltd.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This work was supported by Study of the Relationship between Biomarkers and the Severity and Prognosis of Acute Ischemic Stroke 17411967700 (for DW). Shanghai Municipal Committee of Science and Technology.

References

1. Yousef GM, Diamandis EP. The new human tissue kallikrein gene family: structure, function, and association to disease. Endocr Rev. (2001) 22:184–204. doi: 10.1210/er.22.2.184

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Pathak M, Wong SS, Dreveny I, Emsley J. Structure of plasma and tissue kallikreins. Thromb Haemost. (2013) 110:423–33. doi: 10.1160/TH12-11-0840

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Murray SR, Chao J, Lin FK, Chao L. Kallikrein multigene families and the regulation of their expression. J Cardiovasc Pharmacol. (1990) 15 (Suppl. 6):S7–16.

PubMed Abstract | Google Scholar

4. Yousef GM, Chang A, Scorilas A, Diamandis EP. Genomic organization of the human kallikrein gene family on chromosome 19q13.3-q13.4. Biochem Biophys Res Commun. (2000) 276:125–33. doi: 10.1006/bbrc.2000.3448

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Whalley ET, Figueroa CD, Gera L, Bhoola KD. Discovery and therapeutic potential of kinin receptor antagonists. Expert Opin Drug Discov. (2012) 7:1129–48. doi: 10.1517/17460441.2012.729038

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Igic R. Four decades of ocular renin-angiotensin and kallikrein-kinin systems (1977-2017). Exp Eye Res. (2017) 166:74–83. doi: 10.1016/j.exer.2017.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Savica V, Calo LA, Santoro D, Monardo P, Mallamace A, Bellinghieri G. Urine therapy through the centuries. J Nephrol. (2011) 24 (Suppl. 17):S123–5. doi: 10.5301/JN.2011.6463

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Potier L, Waeckel L, Richer C, Roussel R, Bouby N, Alhenc-Gelas F. Tissue kallikrein, blood pressure regulation, and hypertension: insight from genetic kallikrein deficiency. Biol Chem. (2013) 394:329–33. doi: 10.1515/hsz-2012-0332

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Katori M, Majima M. Renal (tissue) kallikrein-kinin system in the kidney and novel potential drugs for salt-sensitive hypertension. Prog Drug Res. (2014) 69:59–109. doi: 10.1007/978-3-319-06683-7_4

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Zhang Q, Ding H, Yan J, Wang W, Ma A, Zhu Z, et al. Plasma tissue kallikrein level is negatively associated with incident and recurrent stroke: a multicenter case-control study in China. Ann Neurol. (2011) 70:265–73. doi: 10.1002/ana.22404

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Wang YD, Lu RY, Huang XX, Yuan F, Hu T, Peng Y, et al. Human tissue kallikrein promoted activation of the ipsilesional sensorimotor cortex after acute cerebral infarction. Eur Neurol. (2011) 65:208–14. doi: 10.1159/000325735

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhang C, Tao W, Liu M, Wang D. Efficacy and safety of human urinary kallidinogenase injection for acute ischemic stroke: a systematic review. J Evid Based Med. (2012) 5:31–9. doi: 10.1111/j.1756-5391.2012.01167.x

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Li C, Zha OG, He QY, Wu YZ, Wang TS, Teng JF. Study on the clinical efficacy of Human Urinary Kalllikrein in the treatment of acute cerebral infarction according to TOAST classification. Pak J Pharm Sci. (2015) 28(Suppl. 4):1505–10.

PubMed Abstract | Google Scholar

14. Wang YX, Chen Y, Zhang CH, Li CH, Dong Z, Zhao SN, et al. Study on the effect of urinary kallidinogenase after thrombolytic treatment for acute cerebral infarction. Eur Rev Med Pharmacol Sci. (2015) 19:1009–12.

PubMed Abstract | Google Scholar

15. Ke J, Jing M. Analysis of treatment effect of urinary kallidinogenase combined with edaravone on massive cerebral infarction. Biomed Rep. (2016) 5:155–8. doi: 10.3892/br.2016.692

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Li J, Chen Y, Zhang X, Zhang B, Zhang M, Xu Y. Human urinary kallidinogenase improves outcome of stroke patients by shortening mean transit time of perfusion magnetic resonance imaging. J Stroke Cerebrovasc Dis. (2015) 24:1730–7. doi: 10.1016/j.jstrokecerebrovasdis.2015.03.032

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Fagan SC, Hess DC, Hohnadel EJ, Pollock DM, Ergul A. Targets for vascular protection after acute ischemic stroke. Stroke (2004) 35:2220–5. doi: 10.1161/01.STR.0000138023.60272.9e

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Kidoya H, Takakura N. Biology of the apelin-APJ axis in vascular formation. J Biochem. (2012) 152:125–31. doi: 10.1093/jb/mvs071

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Miao J, Deng F, Zhang Y, Xie HY. Exogenous human urinary kallidinogenase increases cerebral blood flow in patients with acute ischemic stroke. Neurosciences (2016) 21:126–30. doi: 10.17712/nsj.2016.2.20150581

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Song X, Han L, Liu Y. Remodeling of motor cortex function in acute cerebral infarction patients following human urinary kallidinogenase: a functional magnetic resonance imaging evaluation after 6 months. Neural Regen Res. (2012) 7:867–73. doi: 10.3969/j.issn.1673-5374.2012.11.012

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Shi R, Zhang R, Yang F, Lin M, Li M, Liu L, et al. Tissue kallikrein prevents restenosis after stenting of severe atherosclerotic stenosis of the middle cerebral artery: a randomized controlled trial. Medicine (2016) 95:e2809. doi: 10.1097/MD.0000000000002809

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Lan W, Yang F, Liu L, Yin Q, Li M, Li Z, et al. Tissue kallikrein preventing the restenosis after stenting of symptomatic MCA atherosclerotic stenosis (KPRASS). Int J Stroke (2014) 9:533–5. doi: 10.1111/ijs.12229

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Zhao L, Zhao Y, Wan Q, Zhang H. Urinary kallidinogenase for the treatment of cerebral arterial stenosis. Drug Des Devel Ther. (2015) 9:5595–600. doi: 10.2147/DDDT.S93150

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Lan W, Yang F, Li Z, Liu L, Sang H, Jiang Y, et al. Human urine kininogenase attenuates balloon-induced intimal hyperplasia in rabbit carotid artery through transforming growth factor beta1/Smad2/3 signaling pathway. J Vasc Surg. (2016) 64:1074–83. doi: 10.1016/j.jvs.2015.04.433

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Ni J, Qu J, Yao M, Zhang Z, Zhong X, Cui L. Re-evaluate the Efficacy and Safety of Human Urinary Kallidinogenase (RESK): protocol for an open-label, single-arm, multicenter phase IV trial for the treatment of acute ischemic stroke in Chinese patients. Transl Stroke Res. (2017) 8:341–6. doi: 10.1007/s12975-017-0527-5

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Xia CF, Yin H, Yao YY, Borlongan CV, Chao L, Chao J. Kallikrein protects against ischemic stroke by inhibiting apoptosis and inflammation and promoting angiogenesis and neurogenesis. Hum Gene Ther. (2006) 17:206–19. doi: 10.1089/hum.2006.17.206

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Ling L, Hou Q, Xing S, Yu J, Pei Z, Zeng J. Exogenous kallikrein enhances neurogenesis and angiogenesis in the subventricular zone and the peri-infarction region and improves neurological function after focal cortical infarction in hypertensive rats. Brain Res. (2008) 1206:89–97. doi: 10.1016/j.brainres.2008.01.099

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Fukuhara J, Noda K, Murata M, Namba S, Kinoshita S, Dong Z, et al. Tissue kallikrein attenuates choroidal neovascularization via cleavage of vascular endothelial growth factor. Invest Ophthalmol Vis Sci. (2013) 54:274–9. doi: 10.1167/iovs.12-10512

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Chao J, Chao L. Experimental therapy with tissue kallikrein against cerebral ischemia. Front Biosci. (2006) 11:1323–7. doi: 10.2741/1886

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Yang J, Su J, Wan F, Yang N, Jiang H, Fang M, et al. Tissue kallikrein protects against ischemic stroke by suppressing TLR4/NF-kappaB and activating Nrf2 signaling pathway in rats. Exp Ther Med. (2017) 14:1163–70. doi: 10.3892/etm.2017.4614

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Cekanaviciute E, Fathali N, Doyle KP, Williams AM, Han J, Buckwalter MS. Astrocytic transforming growth factor-beta signaling reduces subacute neuroinflammation after stroke in mice. Glia (2014) 62:1227–40. doi: 10.1002/glia.22675

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Zhu H, Gui Q, Hui X, Wang X, Jiang J, Ding L, et al. TGF-beta1/Smad3 signaling pathway suppresses cell apoptosis in cerebral ischemic stroke rats. Med Sci Monit. (2017) 23:366–76. doi: 10.12659/MSM.899195

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Dong TF, Lv HX, Niu XL, Gui YK, Zhang P, Yan HQ, et al. Effect of urinary kallidinogenase on transforming growth factor-beta1 and high-sensitivity C-reactive protein expression in rat focal cerebral ischemic injury. Med Sci Monit. (2016) 22:2852–8. doi: 10.12659/MSM.892724

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Liu L, Zhang R, Liu K, Zhou H, Tang Y, Su J, et al. Tissue kallikrein alleviates glutamate-induced neurotoxicity by activating ERK1. J Neurosci Res. (2009) 87:3576–90. doi: 10.1002/jnr.22151

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Liu L, Zhang R, Liu K, Zhou H, Yang X, Liu X, et al. Tissue kallikrein protects cortical neurons against in vitro ischemia-acidosis/reperfusion-induced injury through the ERK1/2 pathway. Exp Neurol. (2009) 219:453–65. doi: 10.1016/j.expneurol.2009.06.021

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Liu L, Liu H, Yang F, Chen G, Zhou H, Tang M, et al. Tissue kallikrein protects cortical neurons against hypoxia/reoxygenation injury via the ERK1/2 pathway. Biochem Biophys Res Commun. (2011) 407:283–7. doi: 10.1016/j.bbrc.2011.02.112

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Su J, Tang Y, Zhou H, Liu L, Dong Q. Tissue kallikrein protects neurons from hypoxia/reoxygenation-induced cell injury through Homer1b/c. Cell Signal. (2012) 24:2205–15. doi: 10.1016/j.cellsig.2012.04.021

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Liu Y, Lu Z, Cui M, Yang Q, Tang Y, Dong Q. Tissue kallikrein protects SH-SY5Y neuronal cells against oxygen and glucose deprivation-induced injury through bradykinin B2 receptor-dependent regulation of autophagy induction. J Neurochem. (2016) 139:208–20. doi: 10.1111/jnc.13690

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Liu W, Yang Y, Liu Y, Lu X, Guo S, Wu M, et al. Exogenous kallikrein protects against diabetic nephropathy. Kidney Int. (2016) 90:1023–36. doi: 10.1016/j.kint.2016.06.018

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Bhat M, Pouliot M, Couture R, Vaucher E. The kallikrein-kinin system in diabetic retinopathy. Prog Drug Res. (2014) 69:111–43. doi: 10.1007/978-3-319-06683-7_5

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: acute ischemic stroke, angiogenesis, neurogenesis, inflammation, human urinary kallidinogenase

Citation: Wei Z, Lyu Y, Yang X, Chen X, Zhong P and Wu D (2018) Therapeutic Values of Human Urinary Kallidinogenase on Cerebrovascular Diseases. Front. Neurol. 9:403. doi: 10.3389/fneur.2018.00403

Received: 03 December 2017; Accepted: 15 May 2018;
Published: 05 June 2018.

Edited by:

Nikolaus Plesnila, Institut für Schlaganfall- und Demenzforschung, Germany

Reviewed by:

Christiane Albert-Weissenberger, Universität Würzburg, Germany
Maxime Gauberti, INSERM U1237 Physiopathologie et Imagerie des Troubles Neurologiques (PhIND), France

Copyright © 2018 Wei, Lyu, Yang, Chen, Zhong and Wu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Danhong Wu, wudanhong@5thhospital.com

Co-first authors.

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.