Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1417 word(s) 1417 2021-08-03 08:13:32 |
2 update references and layout Meta information modification 1417 2021-08-20 06:22:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wang, J. Chelerythrine Chloride and Urease inhibitor. Encyclopedia. Available online: https://encyclopedia.pub/entry/13387 (accessed on 20 April 2024).
Wang J. Chelerythrine Chloride and Urease inhibitor. Encyclopedia. Available at: https://encyclopedia.pub/entry/13387. Accessed April 20, 2024.
Wang, Jiaqi. "Chelerythrine Chloride and Urease inhibitor" Encyclopedia, https://encyclopedia.pub/entry/13387 (accessed April 20, 2024).
Wang, J. (2021, August 20). Chelerythrine Chloride and Urease inhibitor. In Encyclopedia. https://encyclopedia.pub/entry/13387
Wang, Jiaqi. "Chelerythrine Chloride and Urease inhibitor." Encyclopedia. Web. 20 August, 2021.
Chelerythrine Chloride and Urease inhibitor
Edit

 Inhibition of ruminal microbial urease is of particular interest due to its crucial role in regulating urea-N utilization efficiency and nitrogen pollution in the livestock industry. Chelerythrine chloride was selected as a potential urease inhibitor by inhibiting the GTPase activity of UreG and interaction between UreG and nickel, which were the key steps in disrupting urease maturation. The inhibition potency of chelerythrine chloride against UreG provided new evidence for strategies to develop novel urease inhibitors targeting UreG to reduce nitrogen excretion from ruminants.


urease inhibitor UreG chelerythrine chloride ammonia production rumen

1. Introduction

Urea is commonly used as a cost-efficient replacement of feed proteins to provide the sole nitrogen source for urease [1][2][3]. In rumens, urease catalyzes the hydrolysis of urea into ammonia, which can be synthesized into microbial protein to support the requirements for animal growth, meat or milk production [4]. However, the high urease activity leads to excessive ammonia production, which not only results in the explosion of toxic ammonia in the blood, but is also converted to volatile ammonia and escapes into the environment [5][6]. Emissions of nitrogen have been recognized as one of the major drivers in the production of greenhouse gases and wastewater and soil pollution. The livestock industry contributes approximately 65 Tg·N·yr−1, which is roughly one-third of the global human-induced nitrogen emissions [7]. Ruminant nitrogen is the major source of livestock pollution, with approximately 46 Tg·N·yr−1, equivalent to 71% of the total nitrogen emissions from livestock [7]. For ruminants, the dietary nitrogen utilization efficiency is only about 25% [8][9], with as much as 60–90% of the feed nitrogen being excreted [10]. Taken together, regulation of urease activity is crucial to reduce ammonia emissions and improve the efficiency of urea-N utilization in ruminants, and urease inhibitors have been recognized as one of the most effective strategies.
There are many kinds of urease inhibitors, including those that attack the active center of urease, such as urea analogues [11], hydroxamic acids [12], phosphoramide and their derivatives [13], as well as other inhibitors that interact with the flap regions near active centers, such as 1, 4-benzoquinone [14], catechol [15], and some heavy metal ions [16]. However, only acetohydroxamic acid (AHA) is a commercially available urease inhibitor approved by the Food and Drug Administration for the treatment of humans and animals [17]. Unfortunately, significant adverse effects, including deep-vein phlebothrombosis, lower-extremity phlebitis, and malformation of embryos, have been reported [18]. In rumens, AHA was found to degrade rapidly, and inhibit the growth of rumen microorganisms other than urease-producing bacteria [19]. In recent years, plant-derived natural compounds (including that shown in Cover picture [20][21][22][23]) have been preferred for their low toxicity, chemically stability, eco-friendliness and high efficiency at low concentrations [24]. Gnetegha Ayemele et al. [25] screened giant milkweed as an alternative plant-derived feed additive, which could improve nitrogen utilization efficiency by inhibiting protozoa in ruminants without impairing fermentation. Limited studies have found that natural compounds such as terpenoids, phenolic compounds, alkaloids, and other substances were inhibitory towards the activities of plant and microbial ureases [26]. These findings suggest that natural compounds have great potential as urease inhibitors.
The active center of urease is deeply buried in apo-urease, with an unobserved wide-open state [27]. Additionally, urease is a highly specific substrate of urea [28], which makes it challenging to design new urease inhibitors based on the active center. Accessory protein UreG, which is a GTPase involved in transferring nickel to the active center and activating urease upon GTP hydrolysis, plays an important role in urease maturation [29]. It has been reported [30][31] that UreG receives nickel from the accessory protein UreE through the formation of an UreE-UreG complex. Subsequently, nickel-charged UreG delivered nickel to the active center of apo-urease by forming an UreG-UreFH complex and apo-urease/UreGFH super-complex, in which GTP hydrolysis induced the conformational changes of UreG, and promoted the insertion of nickel into the active center. Moreover, UreG and urease maturation are relatively conserved among different urease-producing bacteria [32]. Disruption of the urease maturation process by abolishing UreG function has been proposed as a new strategy to develop urease inhibitors.

2. Screening of urease inhibitor

Given the importance of UreG GTPase activity and the simplicity of GTPase activity detection, we set the GTPase activity of UreG as an indicator to preliminary screen for potential inhibitors from 1130 natural compounds. Most of the top 20 compounds with better inhibition potency were alkaloids, among which nitidine chloride, chelerythrine chloride, chelerythrine, sanguinarine chloride, sanguinarine and dihydrochelerythrine were the major bioactive ingredients of Zanthoxylum nitidum. Some studies [43,44] found that Z. nitidum and its bioactive ingredients were used to treat gastritis diseases, and inhibited the growth of H. pylori. The mechanism of anti-gastritis and inhibiting H. pylori activity remains unclear, which may be related to the decrease in UreG GTP activity.
To better identify potent inhibitors of urease, the IC50 of compounds with stronger inhibition potency towards UreG GTP activity, as well as the binding energy and inhibitor constant obtained using molecular docking, were used for further screening. Chelerythrine chloride is considered to be the most potential inhibitor of urease, with the lowest IC50 and Ki values and the highest binding energy. Li et al. [33] demonstrated that when the final concentration of H. pylori UreG was 5 μM, the IC50 value of coptisine was 89.86 μM. In this study, the IC50 value of coptisine chloride was 86.62 μM, with a final concentration of 3 μM UreG, and the IC50 value of 18.13 μM for chelerythrine chloride was significantly lower than that of coptisine chloride. Additionally, chelerythrine chloride and coptisine chloride, two alkaloids, had similar binding energy and Ki, which were different from that of the caffeoylquinic acid isochlorogenic acid C. Notably, when the final concentration of natural compounds was 100 µM, the inhibition rate of isochlorogenic acid C against UreG activity was stronger than that of chelerythrine chloride. However, when their final concentration was lower than 100 µM, the fitting curve of chelerythrine chloride was steeper with weaker GTPase activity, suggesting that it is better to evaluate the inhibition potency of compounds using multiple additive concentrations.

3.  The mechanism of chelerythrine chloride as a  urease inhibitor of UreG

UreG is thought to act as a bridge that transfers nickel from UreE to apo-urease upon the formation of the UreE-UreG and UreDFG complexes. The key to UreG serving as a target to design urease inhibitors is to disrupt the urease activation process by blocking nickel delivery to UreG. Here, we further explored whether nickel binding to UreG was suppressed by chelerythrine chloride as well as its underlying mechanism. The ITC results show that chelerythrine chloride reduced the molar ratio, binding affinity and −ΔH value in between nickel and UreG, inhibiting nickel binding to UreG. Furthermore, chelerythrine chloride induced a secondary structure change of UreG, and formed a pi-anion interaction with Asp41. The crystal structure of the UreGFH complex [31] revealed that the Asp37 residue (Asp41 residue in the study) generated charge-charge repulsion with γ-phosphate upon GTP binding, which induced the formation of a salt bridge between Glu42 and Arg130, and the motion of “zip-up” between β2 strand and β3 strand, subsequently propagating the conformational changes of UreG in the CPH motif, which was the key site for nickel binding. Chelerythrine chloride may disrupt the biological function of CPH motif through the induced structure change of UreG and interaction with Asp41 residue. Our previous study [34] also found that the Asp41 residue was the key residue affecting the binding of nickel and UreG. A recent study [35] demonstrated that the cmpd4 compound formed a hydrogen bond with Thy15 residue of H. pylori UreG, and inhibited the activities of UreG and H. pylori. The results of Thy15 are consistent with those of our Thy19.
Yang et al. [35] first proposed and confirmed that targeting the metallochaperone UreG to design urease inhibitors could disrupt the urease maturation process, and provided two effective urease inhibitors of colloidal bismuth against H. pylori activity only through UreG, not apo-urease. Li et al. [33] demonstrated that coptisine inactivation of H. pylori urease involved binding to the urease active site sulfhydryl group and accessory protein UreG. These results show the potential of UreG serving as a target for developing urease inhibitors. Chelerythrine chloride not only inhibited the GTPase activity of UreG from a predominant ruminal microbial urease, but also suppressed nickel binding to UreG, which would interfere with urease maturation. Moreover, in clinical practice, the antimicrobial, antibacterial, anti-inflammatory and antiplatelet activities of chelerythrine have been extensively studied [36][37]. In the livestock industry, pharmacokinetic analysis concluded that the addition of chelerythrine to feed was safe due to the first pass effect after intestinal and liver metabolism [38]. These characteristics of chelerythrine chloride provide a solid foundation for the development of chelerythrine chloride as a natural feed additive in ruminants.

References

  1. Matthews, C.; Crispie, F.; Lewis, E.; Reid, M.; O’Toole, P.W.; Cotter, P.D. The rumen microbiome: A crucial consideration when optimising milk and meat production and nitrogen utilisation efficiency. Gut Microbes 2019, 10, 115–132.
  2. Kertz, A.F. Review: Urea feeding to dairy cattle: A historical perspective and review. Prof. Anim. Sci. 2010, 26, 257–272.
  3. Lin, W.; Mathys, V.; Ang, E.L.; Koh, V.H.; Martínez Gómez, J.M.; Ang, M.L.; Zainul Rahim, S.Z.; Tan, M.P.; Pethe, K.; Alonso, S. Urease activity represents an alternative pathway for Mycobacterium tuberculosis nitrogen metabolism. Infect. Immun. 2012, 80, 2771–2779.
  4. Reynolds, C.K.; Kristensen, N.B. Nitrogen recycling through the gut and the nitrogen economy of ruminants: An asynchronous symbiosis. J. Anim. Sci. 2008, 86, E293–E305.
  5. Powell, J.M.; Wattiaux, M.A.; Broderick, G.A. Short communication: Evaluation of milk urea nitrogen as a management tool to reduce ammonia emissions from dairy farms. J. Dairy Sci. 2011, 94, 4690–4694.
  6. Patra, A.K. Urea/Ammonia Metabolism in the Rumen and Toxicity in Ruminants. In Rumen Microbiology: From Evolution to Revolution; Puniya, A.K., Singh, R., Eds.; Springer: New Delhi, India, 2015; pp. 329–341.
  7. Uwizeye, A.; de Boer, I.J.M.; Opio, C.I.; Schulte, R.P.O.; Falcucci, A.; Tempio, G.; Teillard, F.; Casu, F.; Rulli, M.; Galloway, J.N.; et al. Nitrogen emissions along global livestock supply chains. Nat. Food 2020, 1, 436–437.
  8. Kohn, A.; Dinneen, M.; Russek-Cohen, E. Using blood urea nitrogen to predict nitrogen excretion and efficiency of nitrogen utilization in cattle, sheep, goats, horses, pigs, and rats. J. Anim. Sci. Technol. 2005, 83, 879–889.
  9. Huhtanen, P.; Hristov, N. A meta-analysis of the effects of dietary protein concentration and degradability on milk protein yield and milk N efficiency in dairy cows. J. Dairy Sci. 2009, 92, 3222–3232.
  10. Flachowsky, G.; Lebzien, P. Possibilities for reduction of nitrogen (N) excretion from ruminants and the need for further research—A review. Landbauforsch. Volkenrode 2006, 56, 19–30.
  11. Estiu, G.; Merz, K.M. Enzymatic catalysis of urea decomposition: Elimination or hydrolysis? J. Am. Chem. Soc. 2004, 126, 11832–11842.
  12. Krajewska, B.; Zaborska, W.; Leszko, M. Inhibition of chitosan-immobilized urease by slow-binding inhibitors: Ni2+, F− and acetohydroxamic acid. J. Mol. Catal. B Enzym. 2001, 14, 101–109.
  13. Domínguez, M.J.; Sanmartín, C.; Font, M.; Palop, J.A.; San Francisco, S.; Urrutia, O.; Houdusse, F.; García-Mina, J. Design, synthesis, and biological evaluation of phosphoramide derivatives as urease inhibitors. J. Agric. Food Chem. 2008, 56, 3721–3731.
  14. Mazzei, L.; Cianci, M.; Musiani, F.; Ciurli, S. Inactivation of urease by 1,4-benzoquinone: Chemistry at the protein surface. Dalton Trans. 2016, 45, 5455–5459.
  15. Mazzei, L.; Cianci, M.; Musiani, F.; Lente, G.; Palombo, M.; Ciurli, S. Inactivation of urease by catechol: Kinetics and structure. J. Inorg. Biochem. 2017, 166, 182–189.
  16. Shaw, W.H.R.; Raval, D.N. The inhibition of urease by metal ions at pH 8. 9. J. Am. Chem. Soc. 1961, 83, 3184–3187.
  17. Kosikowska, P.; Berlicki, L. Urease inhibitors as potential drugs for gastric and urinary tract infections: A patent review. Expert Opin. Ther. Pat. 2011, 21, 945–957.
  18. Williams, J.J.; Rodman, J.S.; Peterson, C.M. A randomized double-blind study of acetohydroxamic acid in struvite nephrolithiasis. N. Engl. J. Med. 1984, 311, 760–764.
  19. Patra, A.K.; Aschenbach, J.R. Ureases in the gastrointestinal tracts of ruminant and monogastric animals and their implication in urea-N/ammonia metabolism: A review. J. Adv. Res. 2018, 13, 39–50.
  20. Shang, X.; Pan, H.; Li, M.; Miao, X.; Ding, H. Lonicera japonica Thunb.: Ethnopharmacology, phytochemistry and pharmacology of an important traditional Chinese medicine. J. Ethnopharmacol. 2011, 138, 1–21.
  21. Zhang, M.; Wang, J.; Zhu, L.; Li, T.; Jiang, W.; Zhou, J.; Peng, W.; Wu, C. Zanthoxylum bungeanum maxim. (rutaceae): A systematic review of its traditional uses, botany, phytochemistry, pharmacology, pharmacokinetics, and toxicology. Int. J. Mol. Sci. 2017, 18, 2172.
  22. Wu, J.; Luo, Y.; Deng, D.; Su, S.; Li, S.; Xiang, L.; Hu, Y.; Wang, P.; Meng, X. Coptisine from coptis chinensis exerts diverse beneficial properties: A concise review. J. Cell. Mol. Med. 2019, 23, 946–7960.
  23. Pérard-Viret, J.; Quteishat, L.; Alsalim, R.; Royer, J.; Dumas, F. Cephalotaxus alkaloids. Alkaloids Chem. Biol. 2017, 78, 205–352.
  24. Modolo, L.V.; de Souza, A.X.; Horta, L.P.; Araujo, D.P.; de Fátima, Â. An overview on the potential of natural products as ureases inhibitors: A review. J. Adv. Res. 2015, 6, 35–44.
  25. Gnetegha Ayemele, A.; Ma, L.; Park, T.; Xu, J.; Yu, Z.; Bu, D. Giant milkweed (Calotropis gigantea): A new plant resource to inhibit protozoa and decrease ammoniagenesis of rumen microbiota in vitro without impairing fermentation. Sci. Total Environ. 2020, 743, 140665.
  26. Hassan, S.T.; Žemlička, M. Plant-Derived Urease Inhibitors as Alternative Chemotherapeutic Agents. Arch. Pharm. (Weinheim) 2016, 349, 507–522.
  27. Jabri, E.; Carr, M.B.; Hausinger, R.P.; Karplus, P.A. The crystal structure of urease from Klebsiella aerogenes. Science 1995, 268, 998–1004.
  28. Ha, N.C.; Oh, S.T.; Sung, J.Y.; Cha, K.A.; Lee, M.H.; Oh, B.H. Supramolecular assembly and acid resistance of Helicobacter pylori urease. Nat. Struct. Biol. 2001, 8, 505–509.
  29. Leipe, D.D.; Wolf, Y.I.; Koonin, E.V.; Aravind, L. Classification and evolution of P-loop GTPases and related ATPases. J. Mol. Biol. 2002, 317, 41–72.
  30. Yang, X.; Li, H.; Lai, T.P.; Sun, H. UreE-UreG complex facilitates nickel transfer and preactivates GTPase of UreG in Helicobacter pylori. J. Biol. Chem. 2015, 290, 12474–12485.
  31. Yuen, M.H.; Fong, Y.H.; Nim, Y.S.; Lau, P.H.; Wong, K.B. Structural insights into how GTP-dependent conformational changes in a metallochaperone UreG facilitate urease maturation. Proc. Natl. Acad. Sci. USA 2017, 114, E10890–E10898.
  32. Farrugia, M.A.; Macomber, L.; Hausinger, R.P. Biosynthesis of the urease metallocenter. J. Biol. Chem. 2013, 288, 13178–13185.
  33. Lu, Q.; Li, C.; Wu, G. Insight into the inhibitory effects of Zanthoxylum nitidum against Helicobacter pylori urease and jack bean urease: Kinetics and mechanism. J. Ethnopharmacol. 2020, 249, 112419.
  34. Mahady, G.B.; Pendland, S.L.; Stoia, A.; Chadwick, L.R. In vitro susceptibility of Helicobacter pylori to isoquinoline alkaloids from Sanguinaria canadensis and Hydrastis canadensis. Phytother. Res. 2003, 17, 217–221.
  35. Li, C.; Huang, P.; Wong, K.; Xu, Y.; Tan, L.; Chen, H.; Lu, Q.; Luo, C.; Tam, C.; Zhu, L.; et al. Coptisine-induced inhibition of Helicobacter pylori: Elucidation of specific mechanisms by probing urease active site and its maturation process. J. Enzyme Inhib. Med. Chem. 2018, 33, 1362–1375.
  36. Zhang, X.; Zhao, S.; He, Y.; Zheng, N.; Yan, X.; Wang, J. Substitution of residues in UreG to investigate UreE interactions and nickel binding in a predominant urease gene cluster from the ruminal metagenome. Int. J. Biol. Macromol. 2020, 161, 1591–1601.
  37. Yang, X.; Koohi-Moghadam, M.; Wang, R.; Chang, Y.Y.; Woo, P.C.Y.; Wang, J.; Li, H.; Sun, H. Metallochaperone UreG serves as a new target for design of urease inhibitor: A novel strategy for development of antimicrobials. PLoS Biol. 2018, 16, e2003887.
  38. Kosina, P.; Gregorova, J.; Gruz, J.; Vacek, J.; Kolar, M.; Vogel, M.; Roos, W.; Naumann, K.; Simanek, V.; Ulrichova, J. Phytochemical and antimicrobial characterization of Macleaya cordata herb. Fitoterapia 2010, 81, 1006–1012.
  39. Hu, N.X.; Chen, M.; Liu, Y.S.; Shi, Q.; Yang, B.; Zhang, H.C.; Cheng, P.; Tang, Q.; Liu, Z.Y.; Zeng, J.G. Pharmacokinetics of sanguinarine, chelerythrine, and their metabolites in broiler chickens following oral and intravenous administration. J. Vet. Pharmacol. Ther. 2019, 42, 197–206.
More
Information
Subjects: Microbiology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 385
Revisions: 2 times (View History)
Update Date: 20 Aug 2021
1000/1000