1
|
Zhang T, Wang Y, Gu Y, Wu J, Zhan X, Gong P. Gellan gum-sialoglycan conjugates: a mucin mimic for alleviating inflammation in Caco-2 cells and modulating gut microbiota in the elderly. Int J Biol Macromol 2025; 310:143478. [PMID: 40286969 DOI: 10.1016/j.ijbiomac.2025.143478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/13/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
The sialylation of intestinal mucins plays a crucial role in maintaining intestinal homeostasis and shaping the gut microbiota. This study aims to develop a mucin-like polymer to modulate the intestinal microbiota in the elderly individuals. We synthesized functionalized conjugates using sialic acid monomer, 3'-sialyllactose, and Gellan gum, resulting in a series of GG-sialoglycan conjugates with diverse glycan chain lengths and terminal structures. The sialic acid contents of GG-EDA-Sia, GG-EDA-SL, GG-HAD-Sia, and GG-HAD-SL were 28.48 %, 29.78 %, 23.45 and 41.58 %, respectively. These conjugates exhibited anti-digestion effects. In addition, they demonstrated favorable biocompatibility and exhibited notable anti-inflammatory properties. In vitro fermentation experiments using fecal bacteria from elderly individuals revealed that GG-sialoglycan conjugates enhanced the proliferation of beneficial bacteria such as Lactobacillus, Bifidobacterium, and Blautia, while simultaneously suppressing pathogens like Escherichia-Shigella. It is worth noting that GG-sialoglycan conjugates containing 3'-sialyllactose exhibited superior prebiotic activity compared to those with sialic acid monomer. In summary, our findings strongly support the promising potential of functional sialic acid-based macromolecular glycan conjugates as a novel strategy to improve aging-related intestinal health.
Collapse
Affiliation(s)
- Tiantian Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yuying Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yiqun Gu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Jianrong Wu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Xiaobei Zhan
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Ping Gong
- Department of Obstetrical, Affiliated Hospital of Jiangnan University, Wuxi 214123, China
| |
Collapse
|
2
|
Skinner AG, Malik A, Siddiqui MR, Singh V, Akhtar S. Inulin Protects Caco-2 Cells Against Lipopolysaccharide-Induced Epithelial Barrier Dysfunction. Food Sci Nutr 2025; 13:e70046. [PMID: 40129999 PMCID: PMC11932052 DOI: 10.1002/fsn3.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/13/2024] [Accepted: 02/01/2025] [Indexed: 03/26/2025] Open
Abstract
Lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria, triggers inflammatory responses in intestinal epithelial cells. This activation leads to the production of pro-inflammatory cytokines which disrupt cellular homeostasis. LPS also impairs the integrity of the intestinal epithelial barrier by downregulating tight junction proteins, resulting in increased intestinal permeability. This compromised barrier function can allow further translocation of luminal antigens, perpetuating inflammation and contributing to gut-related disorders such as inflammatory bowel disease (IBD) and metabolic endotoxemia. This study investigated the therapeutic effects of inulin, a prebiotic dietary fiber, in attenuating LPS-induced intestinal epithelial barrier dysfunction. Caco-2 cells were treated with 100 ng/mL of LPS for 12 h, resulting in increased gene expression of pro-inflammatory cytokines (IL-1β, TNF-α, and IL-18) and a significant downregulation of tight junction proteins claudin-1 and claudin-2, while occludin gene expression remained unaffected. Pretreatment with 2% inulin for 24 h before LPS exposure prevented the downregulation of claudin-1 and claudin-2 and significantly upregulated occludin gene expression. These molecular findings were supported by functional assays using transwell systems. LPS treatment increased paracellular permeability of the Caco-2 monolayer, indicating barrier dysfunction, while inulin pretreatment mitigated this effect. These results demonstrate that inulin can modulate tight junction protein expression and maintain gut barrier integrity under inflammatory conditions.
Collapse
Affiliation(s)
- A. Georgetta Skinner
- Department of Biochemistry, Kirksville College of Osteopathic MedicineA.T. Still UniversityKirksvilleMissouriUSA
| | - Abdul Malik
- Department of Pharmaceutics, College of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - M. Rizwan Siddiqui
- Department of Biological SciencesMurray State UniversityMurrayKentuckyUSA
| | - Vineet Singh
- Thomas F. Frist, JR. College of MedicineBelmont UniversityNashvilleTennesseeUSA
| | - Suhail Akhtar
- Department of Biochemistry, Kirksville College of Osteopathic MedicineA.T. Still UniversityKirksvilleMissouriUSA
| |
Collapse
|
3
|
Liu L, Chen Y, Han Y, Zhang X, Wu Y, Lin J, Cao L, Wu M, Zheng H, Fang Y, Wei L, Sferra TJ, Jafri A, Ke X, Peng J, Shen A. Qing Hua Chang Yin ameliorates chronic colitis in mice by inhibiting PERK-ATF4-CHOP pathway of ER stress and the NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2024; 62:607-620. [PMID: 39034914 PMCID: PMC11265301 DOI: 10.1080/13880209.2024.2378012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 07/02/2024] [Indexed: 07/23/2024]
Abstract
CONTEXT Ulcerative colitis has been clinically treated with Qing Hua Chang Yin (QHCY), a traditional Chinese medicine formula. However, its precise mechanisms in mitigating chronic colitis are largely uncharted. OBJECTIVE To elucidate the therapeutic efficiency of QHCY on chronic colitis and explore its underlying molecular mechanisms. MATERIALS AND METHODS A total ion chromatogram fingerprint of QHCY was analysed. Chronic colitis was induced in male C57BL/6 mice using 2% dextran sodium sulphate (DSS) over 49 days. Mice were divided into control, DSS, DSS + QHCY (0.8, 1.6 and 3.2 g/kg/d dose, respectively) and DSS + mesalazine (0.2 g/kg/d) groups (n = 6). Mice were intragastrically administered QHCY or mesalazine for 49 days. The changes of disease activity index (DAI), colon length, colon histomorphology and serum pro-inflammatory factors in mice were observed. RNA sequencing was utilized to identify the differentially expressed transcripts (DETs) in colonic tissues and the associated signalling pathways. The expression of endoplasmic reticulum (ER) stress-related protein and NF-κB signalling pathway-related proteins in colonic tissues was detected by immunohistochemistry staining. RESULTS Forty-seven compounds were identified in QHCY. Compared with the DSS group, QHCY significantly improved symptoms of chronic colitis like DAI increase, weight loss, colon shortening and histological damage. It notably reduced serum levels of IL-6, IL-1β and TNF-α. QHCY suppressed the activation of PERK-ATF4-CHOP pathway of ER stress and NF-κB signalling pathways in colonic tissues. DISCUSSION AND CONCLUSIONS The findings in this study provide novel insights into the potential of QHCY in treating chronic colitis patients.
Collapse
Affiliation(s)
- Liya Liu
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children’s Hospital, Cleveland, OH, USA
| | - Yuying Han
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Xinran Zhang
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Yulun Wu
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Jing Lin
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Liujing Cao
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Meizhu Wu
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Huifang Zheng
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Yi Fang
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lihui Wei
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Thomas J. Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children’s Hospital, Cleveland, OH, USA
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, Histology Core, Case Western Reserve University, Cleveland, OH, USA
| | - Xiao Ke
- Department of Gastroenterology, The Second People’s Hospital affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Clinical Medical Research Centre of Chinese Medicine for Spleen and Stomach, Fuzhou, China
| | - Jun Peng
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Aling Shen
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Clinical Research Institute, The Second Affiliated Hospital & Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| |
Collapse
|
4
|
Reale M, Costantini E, Aielli L, Rienzo AD, Biase GD, Stefano AD, Cacciatore I. Exploring the therapeutic potential of cinnamoyl derivatives in attenuating inflammation in lipopolysaccharide-induced Caco-2 cells. Future Med Chem 2024; 16:1395-1411. [PMID: 39190472 DOI: 10.1080/17568919.2024.2351293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 08/28/2024] Open
Abstract
Aim: In gastrointestinal (GI) diseases, lipopolysaccharide (LPS) exacerbates gut-barrier dysfunction and inflammation. Cinnamoyl derivatives show potential in mitigating LPS-induced inflammation.Materials & methods: We assessed intestinal epithelial barrier function using Trans-epithelial electrical resistance values and measured inflammatory mediators through real-time PCR and ELISA in Caco-2 cells.Results: LPS treatment increased IL-6, IL-1β, TNF-α, PGE2 and TRL4 expression in Caco-2 cells. Pre-treatment with DM1 (1 or 10 μM) effectively countered LPS-induced TLR4 overexpression and reduced IL-6, IL-1β, TNF-α and PGE2 levels.Conclusion: DM1 holds promise in regulating inflammation and maintaining intestinal integrity by suppressing TLR4 and inflammatory mediators in Caco-2 cells. These findings suggest a potential therapeutic avenue for GI diseases.
Collapse
Affiliation(s)
- Marcella Reale
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| | - Erica Costantini
- Department of Medicine & Aging Sciences, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| | - Lisa Aielli
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| | - Annalisa Di Rienzo
- Department of Pharmacy, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| | - Giuseppe Di Biase
- Department of Pharmacy, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| | - Antonio Di Stefano
- Department of Pharmacy, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy
| |
Collapse
|
5
|
Sardelli L, Campanile M, Boeri L, Donnaloja F, Fanizza F, Perottoni S, Petrini P, Albani D, Giordano C. A novel on-a-chip system with a 3D-bioinspired gut mucus suitable to investigate bacterial endotoxins dynamics. Mater Today Bio 2024; 24:100898. [PMID: 38204482 PMCID: PMC10776420 DOI: 10.1016/j.mtbio.2023.100898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024] Open
Abstract
The possible pathogenic impact of pro-inflammatory molecules produced by the gut microbiota is one of the hypotheses considered at the basis of the biomolecular dialogue governing the microbiota-gut-brain axis. Among these molecules, lipopolysaccharides (LPS) produced by Gram-negative gut microbiota strains may have a potential key role due to their toxic effects in both the gut and the brain. In this work, we engineered a new dynamic fluidic system, the MINERVA device (MI-device), with the potential to advance the current knowledge of the biological mechanisms regulating the microbiota-gut molecular crosstalk. The MI-device supported the growth of bacteria that are part of the intestinal microbiota under dynamic conditions within a 3D moving mucus model, with features comparable to the physiological conditions (storage modulus of 80 ± 19 Pa, network mesh size of 41 ± 3 nm), without affecting their viability (∼ 109 bacteria/mL). The integration of a fluidically optimized and user-friendly design with a bioinspired microenvironment enabled the sterile extraction and quantification of the LPS produced within the mucus by bacteria (from 423 ± 34 ng/mL to 1785 ± 91 ng/mL). Compatibility with commercially available Transwell-like inserts allows the user to precisely control the transport phenomena that occur between the two chambers by selecting the pore density of the insert membrane without changing the design of the system. The MI-device is able to provide the flow of sterile medium enriched with LPS directly produced by bacteria, opening up the possibility of studying the effects of bacteria-derived molecules on cells in depth, as well as the assessment and characterization of their effects in a physiological or pathological scenario.
Collapse
Affiliation(s)
- L. Sardelli
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - M. Campanile
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - L. Boeri
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - F. Donnaloja
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - F. Fanizza
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - S. Perottoni
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - P. Petrini
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - D. Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - C. Giordano
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| |
Collapse
|
6
|
Garcia BREV, Makiyama EN, Sampaio GR, Soares-Freitas RAM, Bonvini A, Amaral AG, Bordin S, Fock RA, Rogero MM. Effects of Branched-Chain Amino Acids on the Inflammatory Response Induced by LPS in Caco-2 Cells. Metabolites 2024; 14:76. [PMID: 38276311 PMCID: PMC10821323 DOI: 10.3390/metabo14010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/09/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
Branched-chain amino acids (BCAA) are essential for maintaining intestinal mucosal integrity. However, only a few studies have explored the role of BCAA in the modulation of intestinal inflammation. In this study, we investigated in vitro effects of BCAA on the inflammatory response induced by lipopolysaccharide (LPS) (1 µg/mL) in Caco-2 cells. Caco-2 cells were assigned to six groups: control without BCAA (CTL0), normal BCAA (CTL; 0.8 mM leucine, 0.8 mM isoleucine, and 0.8 mM valine); leucine (LEU; 2 mM leucine), isoleucine (ISO; 2 mM isoleucine), valine (VAL; 2 mM valine), and high BCAA (LIV; 2 mM leucine, 2 mM isoleucine, and 2 mM valine). BCAA was added to the culture medium 24 h before LPS stimulation. Our results indicated that BCAA supplementation did not impair cell viability. The amino acids leucine and isoleucine attenuated the synthesis of IL-8 and JNK and NF-kB phosphorylation induced by LPS. Furthermore, neither BCAA supplementation nor LPS treatment modulated the activity of glutathione peroxidase or the intracellular reduced glutathione/oxidized glutathione ratio. Therefore, leucine and isoleucine exert anti-inflammatory effects in Caco-2 cells exposed to LPS by modulating JNK and NF-kB phosphorylation and IL-8 production. Further in vivo studies are required to validate these findings and gather valuable information for potential therapeutic or dietary interventions.
Collapse
Affiliation(s)
- Bruna Ruschel Ewald Vega Garcia
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (B.R.E.V.G.); (G.R.S.); (R.A.M.S.-F.)
| | - Edson Naoto Makiyama
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (E.N.M.); (R.A.F.)
| | - Geni Rodrigues Sampaio
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (B.R.E.V.G.); (G.R.S.); (R.A.M.S.-F.)
| | | | - Andrea Bonvini
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo 05508-000, Brazil;
| | - Andressa Godoy Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508-000, Brazil; (A.G.A.); (S.B.)
| | - Silvana Bordin
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508-000, Brazil; (A.G.A.); (S.B.)
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (E.N.M.); (R.A.F.)
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (B.R.E.V.G.); (G.R.S.); (R.A.M.S.-F.)
- Food Research Center (FoRC), CEPID-FAPESP (Research Innovation and Dissemination Centers São Paulo Research Foundation), São Paulo 05508-000, Brazil
| |
Collapse
|
7
|
Song J, Xu X, He S, Zhang H, Wang N, Bai Y, Li B, Zhang S. Identification of the therapeutic effect and molecular mechanism of Coptis chinensis Franch. and Magnolia officinalis var. biloba on chronic gastritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116864. [PMID: 37393026 DOI: 10.1016/j.jep.2023.116864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/12/2023] [Accepted: 06/27/2023] [Indexed: 07/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) theory believes that clearing heat and promoting dampness is the main treatment method for chronic gastritis. Coptis chinensis Franch. has the effects of clearing heat, detoxifying, and anti-inflammatory; Magnolia officinalis var. biloba can be used to treat abdominal pain, cough, and asthma. Coptis chinensis Franch. and Magnolia officinalis var. biloba can regulate the balance of intestinal microbiota and inhibit inflammatory reactions. AIM This study will verify the therapeutic effect of Coptis chinensis Franch. and Magnolia officinalis var. biloba on chronic gastritis, and explore its mechanism through transcriptome sequencing. METHODS Firstly, a rat chronic gastritis model was established, and the anal temperature and body weight changes of the rats before and after modeling were observed. Next, H&E staining, TUNEL assay and ELISA assay were performed on rat gastric mucosal tissues. Subsequently, the key fractions of Coptis chinensis Franch. and Magnolia officinalis var. biloba were obtained by high performance liquid chromatography (HPLC), and a GES-1 cell inflammation model was constructed to select the optimal monomer. Finally, the mechanism of action of Coptis chinensis Franch. and Magnolia officinalis var. biloba was explored through RNA seq. RESULTS Compared with the control group, the rats in the administered group were in better condition, with higher anal temperature, reduced inflammatory response in gastric mucosal tissue and reduced apoptosis. The optimal fraction Coptisine was subsequently determined by HPLC and GES-1 cell model. RNA-seq analysis revealed that DEG was significantly enriched in ribosomes, NF-κB signaling pathway, etc. The key genes TPT1 and RPL37 were subsequently obtained. CONCLUSIONS This study verified the therapeutic effects of Coptis chinensis Franch. and Magnolia officinalis var. biloba on chronic gastritis by in vivo and in vitro experiments in rats, identified Coptisine as the optimal component, and obtained two potential target genes.
Collapse
Affiliation(s)
- Jin Song
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Huicun Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Ning Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Yunjing Bai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Bo Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China; Beijing Institute of Chinese Medicine, Beijing, 100010, China.
| | - Shengsheng Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| |
Collapse
|
8
|
Anthonymuthu S, Sabui S, Lee K, Sheikh A, Fleckenstein JM, Said HM. Bacterial lipopolysaccharide inhibits colonic carrier-mediated uptake of thiamin pyrophosphate: roles for TLR4 receptor and NF-κB/P38/JNK signaling pathway. Am J Physiol Cell Physiol 2023; 325:C758-C769. [PMID: 37519229 PMCID: PMC10635650 DOI: 10.1152/ajpcell.00272.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023]
Abstract
This study investigated the effect of the bacterial endotoxin lipopolysaccharide (LPS) on colonic uptake of thiamin pyrophosphate (TPP), the biologically active form of vitamin B1 that is generated by gut microbiota. We used three complementary models in our study: in vitro (human-derived colonic epithelial NCM460), ex vivo (human differentiated colonoid monolayers), and in vivo (mouse colonic tissue). The results showed that exposure of NCM460 cells to LPS leads to a significant inhibition of carrier-mediated TPP uptake as well as in decreased expression of the colonic TPP transporter (cTPPT) protein, mRNA, and heterologous nuclear RNA (hnRNA) compared with untreated controls. Similarly, exposure of human differentiated colonoid monolayers and mice to LPS caused significant inhibition in colonic carrier-mediated TPP uptake and in cTPPT protein, mRNA, and hnRNA expression. The effect of LPS on colonic TPP uptake and cTTPT expression was also found to be associated with a significant reduction in activity of the SLC44A4 promoter as well as in decreased expression of the nuclear factor Elf-3 (E74-like ETS transcription factor 3), which is needed for promoter activity. Finally, we found that knocking down the Toll-like receptor 4 (TLR4) and blocking the nuclear factor kappa B (NF-κB), JNK, and p38 signaling pathways with the use of pharmacological inhibitors lead to significant abrogation in the degree of LPS-mediated inhibition in TPP uptake and cTPPT expression. These results demonstrated that exposure of colonic epithelia to LPS inhibits colonic TPP uptake via transcriptional mechanism(s) and that the effect is mediated via TLR4 receptor and NF-κB/p38/JNK signaling pathways.NEW & NOTEWORTHY This study examined the effect of the bacterial lipopolysaccharide (LPS) on the colonic uptake of thiamin pyrophosphate (TPP), the biologically active form of vitamin B1. Three complementary models were used: in vitro (human NCM460 cells), ex vivo (human colonoids), and in vivo (mice). The results showed LPS to significantly suppress TPP uptake and the expression of its transporter, and that these effects are mediated via the membrane TLR4 receptor, and involve the NF-κB/p38/JNK signaling pathways.
Collapse
Affiliation(s)
- Selvaraj Anthonymuthu
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Subrata Sabui
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| | - Katherine Lee
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Veterans Affairs Medical Center, St. Louis, Missouri, United States
| | - Hamid M Said
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medicine, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| |
Collapse
|
9
|
He Z, Liu J, Liu Y. Daphnetin attenuates intestinal inflammation, oxidative stress, and apoptosis in ulcerative colitis via inhibiting REG3A-dependent JAK2/STAT3 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:2132-2142. [PMID: 37209277 DOI: 10.1002/tox.23837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Daphnetin is a natural coumarin compound with anti-inflammatory, anti-oxidant, and anti-apoptotic effects, which has been previously demonstrated to ameliorate DSS-induced ulcerative colitis (UC). However, the molecular mechanism involved in the daphnetin-mediated pathological process of UC remains unclarified. The current study used DSS-induced mice and LPS-challenged Caco-2 cells as UC models. Bodyweight, disease activity index (DAI) score, and colon length were used to evaluate the severity of colitis. The histological changes in colon tissues were observed using H&E and PAS staining. Protein levels were detected by western blot. The malondialdehyde (MDA) and superoxide dismutase (SOD) activities were used to assess oxidative stress. Inflammatory responses were evaluated by detecting the levels of inflammatory cytokines (IFN-r, IL-1β, IL-6, and TNF-α) using flow cytometry. CCK-8 and TUNEL assay were employed to determine cell growth and cell death, respectively. The results showed that daphnetin could ameliorate the severity of colitis and attenuate the damage to intestinal structure in DSS-induced mice. Compared with the DSS group, the expression of ZO-1, occludin, and anti-apoptotic protein (BCL-2) was increased while the level of pro-apoptotic proteins (Bax and cleaved caspase 3) was decreased in DSS + daphnetin group. The activity of MDA and SOD, as well as the levels of inflammatory cytokines were substantially suppressed by daphnetin. In consistency, in vitro assays indicated that daphnetin protected Caco-2 cells from LPS-stimulated viability impairment, apoptosis, oxidative stress, and inflammation. Furthermore, daphnetin suppressed the activity of JAK2/STAT signaling in LPS-induced Caco-2 cells in a REG3A-dependent manner. REG3A overexpression abated the ameliorative effects of daphnetin while JAK2/STAT signaling inhibition functioned synergically with daphnetin in LPS-stimulated Caco-2 cells. Collectively, this study deepened the understanding of the therapeutic effects of daphnetin on UC and uncovered for the first time that daphnetin functioned through REG3A-activated JAK2/STAT3 signaling in UC, which may provide novel insights for the treatment of UC.
Collapse
Affiliation(s)
- Zhi He
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jingjing Liu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yang Liu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
10
|
Jeerawattanawart S, Hansakon A, Roytrakul S, Angkasekwinai P. Regulation and function of adiponectin in the intestinal epithelial cells in response to Trichinella spiralis infection. Sci Rep 2023; 13:14004. [PMID: 37635188 PMCID: PMC10460792 DOI: 10.1038/s41598-023-41377-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/25/2023] [Indexed: 08/29/2023] Open
Abstract
Besides metabolic homeostasis regulation, adipokines are recently emerged as important players in regulating immunity and inflammation. Helminth infection has known to modulate circulating adipokine secretion; however, the regulation and function of adipokines in response to helminth infection is still unclear. Here, we investigated the regulation and function of adiponectin during T. spiralis infection. While there was no change in circulating level of adiponectin, we found an increased adiponectin, but not leptin expression in the small intestine. Interestingly, the intestinal adiponectin expression was strongly associated with the expression of epithelial cell-derived cytokines IL-25, IL-33, and TSLP following infection. Indeed, mice deficiency of IL-25 receptor exhibited no intestinal adiponectin induction upon helminth infection. Interestingly, IL-25-induced adiponectin modulated intestinal epithelial cell responses by enhancing occludin and CCL17 expression. Using LPS-induced intestinal epithelial barrier dysfunctions in a Caco-2 cell monolayer model, adiponectin pretreatment enhanced a Transepithelial electrical resistance (TEER) and occludin expression. More importantly, adiponectin pretreatment of Caco2 cells prevented T. spiralis larval invasion in vitro and its administration during infection enhanced intestinal IL-13 secretion and worm expulsion in vivo. Altogether, our data suggest that intestinal adiponectin expression induced by helminth infection through the regulation of IL-25 promotes worm clearance and intestinal barrier function.
Collapse
Affiliation(s)
- Siranart Jeerawattanawart
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
| | - Adithap Hansakon
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand.
- Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathum Thani, 12120, Thailand.
| |
Collapse
|
11
|
Han Y, Liu L, Chen Y, Zheng H, Yao M, Cao L, Sferra TJ, Ke X, Peng J, Shen A. Qing Hua Chang Yin alleviates chronic colitis of mice by protecting intestinal barrier function and improving colonic microflora. Front Pharmacol 2023; 14:1176579. [PMID: 37576825 PMCID: PMC10413571 DOI: 10.3389/fphar.2023.1176579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Background: Qing Hua Chang Yin (QHCY) is a famous formula of traditional Chinese medicine (TCM) and has been proven to have protective effect on ulcerative colitis. However, its protective effect and potential therapeutic mechanisms in chronic colitis remain unclear. The purpose of this study is to explore the effects and underlying mechanisms of QHCY on dextran sulfate sodium (DSS)-induced chronic colitis mice model. Methods: The chronic colitis model was established by administration of 2% DSS for three consecutive cycles of 7 days with two intervals of 14 days for recovery by drinking water. The experiment lasted 49 days. The DSS + QHCY group received QHCY administration by oral gavage at doses of 1.6 g/kg/d, DSS + Mesalazine group was administrated Mesalazine by oral gavage at doses of 0.2 g/kg/d. The control and DSS group were given equal volume of distilled water. The body weight, stool consistency and blood in stool were monitored every 2 days. The disease activity index (DAI) was calculated. The colon length was measured after the mice were sacrificed. The histomorphology of colonic tissues was checked by the HE and PAS staining. Immunohistochemistry was performed to detect the expressions of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), tight junction proteins (ZO-1, occludin) and Mucin2 (MUC2). 16S rRNA sequencing analysis was conducted to study the diversity and abundance of gut microbiota changes. Results: QHCY treatment not only significantly attenuated DSS-induced the weight loss, DAI score increase, colon shortening and histological damage in mice, but also decreased the expression of pro-inflammatory cytokines in colonic tissues and increased the expression of ZO-1, occludin, and MUC2. Furthermore, QHCY enhanced the diversity of gut microbes and regulated the structure and composition of intestinal microflora in mice with chronic colitis. Conclusion: QHCY has a therapeutic effect on a murine model of chronic colitis. It can effectively reduce the clinical and pathological manifestations of colitis and prevent alterations in the gut microbiota.
Collapse
Affiliation(s)
- Yuying Han
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Liya Liu
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Youqin Chen
- Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Huifang Zheng
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mengying Yao
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Liujing Cao
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Thomas J. Sferra
- Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Xiao Ke
- Department of Gastroenterology, The Second People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Clinical Medical Research Centre of Chinese Medicine for Spleen and Stomach, Fuzhou, China
| | - Jun Peng
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Aling Shen
- Clinical Research Institute, The Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
12
|
Crosstalk between gut microbiota and lung inflammation in murine toxicity models of respiratory exposure or co-exposure to carbon nanotube particles and cigarette smoke extract. Toxicol Appl Pharmacol 2022; 447:116066. [PMID: 35595072 DOI: 10.1016/j.taap.2022.116066] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 11/23/2022]
Abstract
Carbon nanotubes (CNTs) are emerging environmental and occupational toxicants known to induce lung immunotoxicity. While the underlying mechanisms are evolving, it is yet unknown whether inhaled CNTs would cause abnormalities in gut microbiota (dysbiosis), and if such microbiota alteration plays a role in the modulation of CNT-induced lung immunotoxicity. It is also unknown whether co-exposure to tobacco smoke will modulate CNT effects. We compared the effects of lung exposure to multi-wall CNT, cigarette smoke extract (CSE), and their combination (CNT + CSE) in a 4-week chronic toxicity mouse model. The exposures induced differential perturbations in gut microbiome as evidenced by altered microbial α- and β- diversity, indicating a lung-to-gut communication. The gut dysbiosis due to CNTs, unlike CSE, was characterized by an increase in Firmicutes/Bacteroidetes ratio typically associated with proinflammatory condition. Notably, while all three exposures reduced Proteobacteria, the CNT exposure and co-exposure induced appearance of Tenericutes and Cyanobacteria, respectively, implicating them as potential biomarkers of exposure. CNTs differentially induced certain lung proinflammatory mediators (TNF-α, IL-1β, CCL2, CXCL5) whereas CNTs and CSE commonly induced other mediators (CXCL1 and TGF-β). The co-exposure showed either a component-dominant effect or a summative effect for both dysbiosis and lung inflammation. Depletion of gut microbiota attenuated both the differentially-induced and commonly-induced (TGF-β) lung inflammatory mediators as well as granulomas implying gut-to-lung communication and a modulatory role of gut dysbiosis. Taken together, the results demonstrated gut dysbiosis as a systemic effect of inhaled CNTs and provided the first evidence of a bidirectional gut-lung crosstalk modulating CNT lung immunotoxicity.
Collapse
|
13
|
Chen Z, Lv Y, Xu H, Deng L. Herbal Medicine, Gut Microbiota, and COVID-19. Front Pharmacol 2021; 12:646560. [PMID: 34305582 PMCID: PMC8293616 DOI: 10.3389/fphar.2021.646560] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus Disease 19 (COVID-19) is a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has grown to a worldwide pandemic with substantial mortality. The symptoms of COVID-19 range from mild flu-like symptoms, including cough and fever, to life threatening complications. There are still quite a number of patients with COVID-19 showed enteric symptoms including nausea, vomiting, and diarrhea. The gastrointestinal tract may be one of the target organs of SARS-CoV-2. Angiotensin converting enzyme 2 (ACE2) is the main receptor of SARS-CoV-2 virus, which is significantly expressed in intestinal cells. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Intestinal flora imbalance and endotoxemia may accelerate the progression of COVID-19. Many herbs have demonstrated properties relevant to the treatment of COVID-19, by supporting organs and systems of the body affected by the virus. Herbs can restore the structure of the intestinal flora, which may further modulate the immune function after SARS-CoV-2 infection. Regulation of intestinal flora by herbal medicine may be helpful for the treatment and recovery of the disease. Understanding the role of herbs that regulate intestinal flora in fighting respiratory virus infections and maintaining intestinal flora balance can provide new ideas for preventing and treating COVID-19.
Collapse
Affiliation(s)
- Ziqi Chen
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Medical College, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Lv
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Huachong Xu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|