Skip to main content
Top
Published in: Gut Pathogens 1/2022

Open Access 01-12-2022 | Ulcerative Colitis | Research

Jatrorrhizine alleviates ulcerative colitis via regulating gut microbiota and NOS2 expression

Authors: Jia Ling Zhang, Min Na Zhang, Hong Gang Wang, Xiao Zhong Yang, Cheng Gong Yu

Published in: Gut Pathogens | Issue 1/2022

Login to get access

Abstract

Background

The natural protoberberine jatrorrhizine (JA) is reported to have several medicinal properties and a significant effect on the gut microbiota of mice. The regulation of gut microbiota is generally known to play an important role in the intestinal mucosal immune response to ulcerative colitis (UC). However, whether JA can be used in the treatment of UC is still unclear. Our study aimed to investigate the underlying therapeutic effects and mechanisms of JA in treating colitis.

Results

Compared with the DSS-induced colitis model group, the JA + DSS treated group had more significant improvements in weight loss, disease activity index score, colon length shortening, and pathological inflammation. 16s rRNA sequencing analysis showed that JA treatment protected colitis mice against DSS-induced disturbance of gut microbiota. At the phylum level, reductions in Deferribacteres and Proteobacteria were observed in the JA-treated group; At the genus level, the JA-treated group showed an increased relative abundance of Akkermansia and decreased abundance of Escherichia-Shigella, Desulfovibrio, Mucispirillum, etc. Network pharmacology was then used to screen out five drug-disease target genes (NOS2, ESR1, CALM1, CALM2, CALM3). Transcriptomics analysis further validated that the NOS2 expression was significantly reduced in colon tissue of JA-administered mice compared with DSS control mice. Additionally, analysis of correlation suggested that NOS2 expression was negatively correlated with the relative abundance of AKKermansia and positively correlated with Desulfovibrio, Rikenella.

Conclusion

JA alleviates ulcerative colitis via regulating gut microbiota and NOS2 expression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Molodecky NA, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46–54. e42; quiz e30.PubMedCrossRef Molodecky NA, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46–54. e42; quiz e30.PubMedCrossRef
2.
go back to reference De Souza HS, P, et al. The IBD interactome: an integrated view of aetiology, pathogenesis and therapy. Nat Rev Gastroenterol Hepatol. 2017;14(12):739–49.PubMedCrossRef De Souza HS, P, et al. The IBD interactome: an integrated view of aetiology, pathogenesis and therapy. Nat Rev Gastroenterol Hepatol. 2017;14(12):739–49.PubMedCrossRef
3.
go back to reference Xavier RJ, et al. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448(7152):427–34.PubMedCrossRef Xavier RJ, et al. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448(7152):427–34.PubMedCrossRef
4.
go back to reference Maloy KJ, et al. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474(7351):298–306.PubMedCrossRef Maloy KJ, et al. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474(7351):298–306.PubMedCrossRef
6.
go back to reference Baumgart DC, et al. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet. 2007;369(9573):1641–57.PubMedCrossRef Baumgart DC, et al. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet. 2007;369(9573):1641–57.PubMedCrossRef
7.
go back to reference Samuel S, et al. Cumulative incidence and risk factors for hospitalization and surgery in a population-based cohort of ulcerative colitis. Inflamm Bowel Dis. 2013;19(9):1858–66.PubMed Samuel S, et al. Cumulative incidence and risk factors for hospitalization and surgery in a population-based cohort of ulcerative colitis. Inflamm Bowel Dis. 2013;19(9):1858–66.PubMed
8.
go back to reference Zhong F, et al. Jatrorrhizine: A Review of Sources, Pharmacology, Pharmacokinetics and Toxicity. Front Pharmacol. 2021;12:783127.PubMedCrossRef Zhong F, et al. Jatrorrhizine: A Review of Sources, Pharmacology, Pharmacokinetics and Toxicity. Front Pharmacol. 2021;12:783127.PubMedCrossRef
9.
go back to reference Yu H, et al. Jatrorrhizine suppresses the antimicrobial resistance of methicillin-resistant Staphylococcus aureus. Exp Ther Med. 2019;18(5):3715–22.PubMedPubMedCentral Yu H, et al. Jatrorrhizine suppresses the antimicrobial resistance of methicillin-resistant Staphylococcus aureus. Exp Ther Med. 2019;18(5):3715–22.PubMedPubMedCentral
10.
go back to reference Sun Y, et al. Jatrorrhizine inhibits mammary carcinoma cells by targeting TNIK mediated Wnt/beta-catenin signalling and epithelial-mesenchymal transition (EMT). Phytomedicine. 2019;63:153015.PubMedCrossRef Sun Y, et al. Jatrorrhizine inhibits mammary carcinoma cells by targeting TNIK mediated Wnt/beta-catenin signalling and epithelial-mesenchymal transition (EMT). Phytomedicine. 2019;63:153015.PubMedCrossRef
11.
go back to reference Fu Y, et al. Hypoglycemic activity of jatrorrhizine. J Huazhong Univ Sci Technolog Med Sci. 2005;25(5):491–3.PubMedCrossRef Fu Y, et al. Hypoglycemic activity of jatrorrhizine. J Huazhong Univ Sci Technolog Med Sci. 2005;25(5):491–3.PubMedCrossRef
12.
go back to reference Wang P, et al. Jatrorrhizine inhibits colorectal carcinoma proliferation and metastasis through Wnt/beta-catenin signaling pathway and epithelial-mesenchymal transition. Drug Des Devel Ther. 2019;13:2235–47.PubMedPubMedCentralCrossRef Wang P, et al. Jatrorrhizine inhibits colorectal carcinoma proliferation and metastasis through Wnt/beta-catenin signaling pathway and epithelial-mesenchymal transition. Drug Des Devel Ther. 2019;13:2235–47.PubMedPubMedCentralCrossRef
13.
go back to reference Duan W, et al. Jatrorrhizine can improve nerve cell injury induced by Abeta 25–35, acting through miR-223-3p/HDAC4 axis. Am J Transl Res. 2021;13(5):4644–55.PubMedPubMedCentral Duan W, et al. Jatrorrhizine can improve nerve cell injury induced by Abeta 25–35, acting through miR-223-3p/HDAC4 axis. Am J Transl Res. 2021;13(5):4644–55.PubMedPubMedCentral
14.
go back to reference Qiu H, et al. Jatrorrhizine Hydrochloride Suppresses Proliferation, Migration, and Secretion of Synoviocytes In Vitro and Ameliorates Rat Models of Rheumatoid Arthritis In Vivo. Int J Mol Sci. 2018;19(5). Qiu H, et al. Jatrorrhizine Hydrochloride Suppresses Proliferation, Migration, and Secretion of Synoviocytes In Vitro and Ameliorates Rat Models of Rheumatoid Arthritis In Vivo. Int J Mol Sci. 2018;19(5).
15.
go back to reference Wu G, et al. Jatrorrhizine Hydrochloride alleviates tert-butyl hydroperoxide-induced endothelial cell injury through its anti-inflammatory activity and PPAR-gamma activation. Cell Mol Biol (Noisy-le-grand). 2020;66(2):125–9.CrossRef Wu G, et al. Jatrorrhizine Hydrochloride alleviates tert-butyl hydroperoxide-induced endothelial cell injury through its anti-inflammatory activity and PPAR-gamma activation. Cell Mol Biol (Noisy-le-grand). 2020;66(2):125–9.CrossRef
16.
go back to reference Wang S, et al. Jatrorrhizine Balances the Gut Microbiota and Reverses Learning and Memory Deficits in APP/PS1 transgenic mice. Sci Rep. 2019;9(1):19575.PubMedPubMedCentralCrossRef Wang S, et al. Jatrorrhizine Balances the Gut Microbiota and Reverses Learning and Memory Deficits in APP/PS1 transgenic mice. Sci Rep. 2019;9(1):19575.PubMedPubMedCentralCrossRef
17.
go back to reference Argollo M, et al. Novel therapeutic targets for inflammatory bowel disease. J Autoimmun. 2017;85:103–16.PubMedCrossRef Argollo M, et al. Novel therapeutic targets for inflammatory bowel disease. J Autoimmun. 2017;85:103–16.PubMedCrossRef
19.
go back to reference Feng W, et al. Gut microbiota, a new frontier to understand traditional Chinese medicines. Pharmacol Res. 2019;142:176–91.PubMedCrossRef Feng W, et al. Gut microbiota, a new frontier to understand traditional Chinese medicines. Pharmacol Res. 2019;142:176–91.PubMedCrossRef
20.
21.
go back to reference Li C, et al. Oxyberberine, a novel gut microbiota-mediated metabolite of berberine, possesses superior anti-colitis effect: Impact on intestinal epithelial barrier, gut microbiota profile and TLR4-MyD88-NF-kappaB pathway. Pharmacol Res. 2020;152:104603.PubMedCrossRef Li C, et al. Oxyberberine, a novel gut microbiota-mediated metabolite of berberine, possesses superior anti-colitis effect: Impact on intestinal epithelial barrier, gut microbiota profile and TLR4-MyD88-NF-kappaB pathway. Pharmacol Res. 2020;152:104603.PubMedCrossRef
22.
go back to reference Liu C, et al. Dynamic Alterations in Yak Rumen Bacteria Community and Metabolome Characteristics in Response to Feed Type. Front Microbiol. 2019;10:1116.PubMedPubMedCentralCrossRef Liu C, et al. Dynamic Alterations in Yak Rumen Bacteria Community and Metabolome Characteristics in Response to Feed Type. Front Microbiol. 2019;10:1116.PubMedPubMedCentralCrossRef
25.
go back to reference Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10(10):996–8.PubMedCrossRef Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10(10):996–8.PubMedCrossRef
26.
go back to reference Schloss PD, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75(23):7537–41.PubMedPubMedCentralCrossRef Schloss PD, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75(23):7537–41.PubMedPubMedCentralCrossRef
27.
28.
go back to reference Mortazavi A, et al. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008;5(7):621–8.PubMedCrossRef Mortazavi A, et al. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008;5(7):621–8.PubMedCrossRef
31.
go back to reference Kanehisa M, et al. KEGG for linking genomes to life and the environment. Nucleic Acids Res. 2008;36(Database issue):D480-4.PubMed Kanehisa M, et al. KEGG for linking genomes to life and the environment. Nucleic Acids Res. 2008;36(Database issue):D480-4.PubMed
32.
go back to reference Ohkawara T, et al. DNA vaccination targeting macrophage migration inhibitory factor prevents murine experimental colitis. Clin Exp Immunol. 2011;163(1):113–22.PubMedPubMedCentralCrossRef Ohkawara T, et al. DNA vaccination targeting macrophage migration inhibitory factor prevents murine experimental colitis. Clin Exp Immunol. 2011;163(1):113–22.PubMedPubMedCentralCrossRef
33.
go back to reference Becker C, et al. The Intestinal Microbiota in Inflammatory Bowel Disease. ILAR J. 2015;56(2):192–204.PubMedCrossRef Becker C, et al. The Intestinal Microbiota in Inflammatory Bowel Disease. ILAR J. 2015;56(2):192–204.PubMedCrossRef
34.
go back to reference Hegazy AN, et al. Circulating and Tissue-Resident CD4(+) T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology. 2017;153(5):1320-37 e16.CrossRef Hegazy AN, et al. Circulating and Tissue-Resident CD4(+) T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology. 2017;153(5):1320-37 e16.CrossRef
35.
go back to reference Hu Y, et al. The Communication Between Intestinal Microbiota and Ulcerative Colitis: An Exploration of Pathogenesis, Animal Models, and Potential Therapeutic Strategies. Front Med (Lausanne). 2021;8:766126.CrossRef Hu Y, et al. The Communication Between Intestinal Microbiota and Ulcerative Colitis: An Exploration of Pathogenesis, Animal Models, and Potential Therapeutic Strategies. Front Med (Lausanne). 2021;8:766126.CrossRef
36.
go back to reference Sun Z, et al. Gut Microbiota-Mediated NLRP12 Expression Drives the Attenuation of Dextran Sulphate Sodium-Induced Ulcerative Colitis by Qingchang Wenzhong Decoction. Evid Based Complement Alternat Med. 2019;2019:9839474.PubMedPubMedCentralCrossRef Sun Z, et al. Gut Microbiota-Mediated NLRP12 Expression Drives the Attenuation of Dextran Sulphate Sodium-Induced Ulcerative Colitis by Qingchang Wenzhong Decoction. Evid Based Complement Alternat Med. 2019;2019:9839474.PubMedPubMedCentralCrossRef
37.
go back to reference Eeckhaut V, et al. Butyricicoccus pullicaecorum in inflammatory bowel disease. Gut. 2013;62(12):1745–52.PubMedCrossRef Eeckhaut V, et al. Butyricicoccus pullicaecorum in inflammatory bowel disease. Gut. 2013;62(12):1745–52.PubMedCrossRef
38.
go back to reference You Q, et al. Neutrophil Extracellular Traps Caused by Gut Leakage Trigger the Autoimmune Response in Nonobese Diabetic Mice. Front Immunol. 2021;12:711423.PubMedCrossRef You Q, et al. Neutrophil Extracellular Traps Caused by Gut Leakage Trigger the Autoimmune Response in Nonobese Diabetic Mice. Front Immunol. 2021;12:711423.PubMedCrossRef
39.
go back to reference Berry D, et al. Phylotype-level 16S rRNA analysis reveals new bacterial indicators of health state in acute murine colitis. ISME J. 2012;6(11):2091–106.PubMedPubMedCentralCrossRef Berry D, et al. Phylotype-level 16S rRNA analysis reveals new bacterial indicators of health state in acute murine colitis. ISME J. 2012;6(11):2091–106.PubMedPubMedCentralCrossRef
40.
go back to reference Berry D, et al. Intestinal Microbiota Signatures Associated with Inflammation History in Mice Experiencing Recurring Colitis. Front Microbiol. 2015;6:1408.PubMedPubMedCentralCrossRef Berry D, et al. Intestinal Microbiota Signatures Associated with Inflammation History in Mice Experiencing Recurring Colitis. Front Microbiol. 2015;6:1408.PubMedPubMedCentralCrossRef
41.
go back to reference Shin NR, et al. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015;33(9):496–503.PubMedCrossRef Shin NR, et al. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015;33(9):496–503.PubMedCrossRef
42.
go back to reference Mukhopadhya I, et al. IBD-what role do Proteobacteria play? Nat Rev Gastroenterol Hepatol. 2012;9(4):219–30.PubMedCrossRef Mukhopadhya I, et al. IBD-what role do Proteobacteria play? Nat Rev Gastroenterol Hepatol. 2012;9(4):219–30.PubMedCrossRef
43.
go back to reference Sun D, et al. Angiogenin maintains gut microbe homeostasis by balancing alpha-Proteobacteria and Lachnospiraceae. Gut. 2021;70(4):666–76.PubMedCrossRef Sun D, et al. Angiogenin maintains gut microbe homeostasis by balancing alpha-Proteobacteria and Lachnospiraceae. Gut. 2021;70(4):666–76.PubMedCrossRef
44.
go back to reference Macchione IG, et al. Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders. Eur Rev Med Pharmacol Sci. 2019;23(18):8075–83.PubMed Macchione IG, et al. Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders. Eur Rev Med Pharmacol Sci. 2019;23(18):8075–83.PubMed
45.
go back to reference Jialing L, et al. Changes in serum inflammatory cytokine levels and intestinal flora in a self-healing dextran sodium sulfate-induced ulcerative colitis murine model. Life Sci. 2020;263:118587.PubMedCrossRef Jialing L, et al. Changes in serum inflammatory cytokine levels and intestinal flora in a self-healing dextran sodium sulfate-induced ulcerative colitis murine model. Life Sci. 2020;263:118587.PubMedCrossRef
46.
47.
go back to reference Matsuda H, et al. Characterization of antibody responses against rectal mucosa-associated bacterial flora in patients with ulcerative colitis. J Gastroenterol Hepatol. 2000;15(1):61–8.PubMedCrossRef Matsuda H, et al. Characterization of antibody responses against rectal mucosa-associated bacterial flora in patients with ulcerative colitis. J Gastroenterol Hepatol. 2000;15(1):61–8.PubMedCrossRef
48.
go back to reference Do KH, et al. Ribosome Inactivation Leads to Attenuation of Intestinal Polymeric Ig Receptor Expression via Differential Regulation of Human Antigen R. J Immunol. 2016;197(3):847–58.PubMedCrossRef Do KH, et al. Ribosome Inactivation Leads to Attenuation of Intestinal Polymeric Ig Receptor Expression via Differential Regulation of Human Antigen R. J Immunol. 2016;197(3):847–58.PubMedCrossRef
50.
go back to reference Kanwar JR, et al. Recent advances on the roles of NO in cancer and chronic inflammatory disorders. Curr Med Chem. 2009;16(19):2373–94.PubMedCrossRef Kanwar JR, et al. Recent advances on the roles of NO in cancer and chronic inflammatory disorders. Curr Med Chem. 2009;16(19):2373–94.PubMedCrossRef
51.
go back to reference Cross RK, et al. Nitric oxide in inflammatory bowel disease. Inflamm Bowel Dis. 2003;9(3):179–89.PubMedCrossRef Cross RK, et al. Nitric oxide in inflammatory bowel disease. Inflamm Bowel Dis. 2003;9(3):179–89.PubMedCrossRef
52.
go back to reference Rachmilewitz D, et al. Enhanced colonic nitric oxide generation and nitric oxide synthase activity in ulcerative colitis and Crohn’s disease. Gut. 1995;36(5):718–23.PubMedPubMedCentralCrossRef Rachmilewitz D, et al. Enhanced colonic nitric oxide generation and nitric oxide synthase activity in ulcerative colitis and Crohn’s disease. Gut. 1995;36(5):718–23.PubMedPubMedCentralCrossRef
53.
go back to reference [53] Godkin AJ, et al. Expression of nitric oxide synthase in ulcerative colitis. Eur J Clin Invest. 1996;26(10):867–72.PubMedCrossRef [53] Godkin AJ, et al. Expression of nitric oxide synthase in ulcerative colitis. Eur J Clin Invest. 1996;26(10):867–72.PubMedCrossRef
54.
go back to reference Singer I, et al. Expression of inducible nitric oxide synthase and nitrotyrosine in colonic epithelium in inflammatory bowel disease. Gastroenterology. 1996;111(4):871–85.PubMedCrossRef Singer I, et al. Expression of inducible nitric oxide synthase and nitrotyrosine in colonic epithelium in inflammatory bowel disease. Gastroenterology. 1996;111(4):871–85.PubMedCrossRef
55.
go back to reference Bernstein H, et al. Deoxycholate-induced colitis is markedly attenuated in Nos2 knockout mice in association with modulation of gene expression profiles. Dig Dis Sci. 2007;52(3):628–42.PubMedCrossRef Bernstein H, et al. Deoxycholate-induced colitis is markedly attenuated in Nos2 knockout mice in association with modulation of gene expression profiles. Dig Dis Sci. 2007;52(3):628–42.PubMedCrossRef
56.
57.
go back to reference Lukovac S, et al. Differential modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of host peripheral lipid metabolism and histone acetylation in mouse gut organoids. mBio. 2014;5(4). Lukovac S, et al. Differential modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of host peripheral lipid metabolism and histone acetylation in mouse gut organoids. mBio. 2014;5(4).
Metadata
Title
Jatrorrhizine alleviates ulcerative colitis via regulating gut microbiota and NOS2 expression
Authors
Jia Ling Zhang
Min Na Zhang
Hong Gang Wang
Xiao Zhong Yang
Cheng Gong Yu
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Gut Pathogens / Issue 1/2022
Electronic ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-022-00514-z

Other articles of this Issue 1/2022

Gut Pathogens 1/2022 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.