1
|
Sun M, Ma J, Zhang G, Song M, Lv R, Liang J, Shi Y, Zhao L. Brain Targeting Bacterial Extracellular Vesicles Enhance Ischemic Stroke Therapy via Efficient ROS Elimination and Suppression of Immune Infiltration. ACS NANO 2025; 19:15491-15508. [PMID: 40249658 DOI: 10.1021/acsnano.4c16161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Abstract
Ischemic stroke (IS) as a detrimental neurological disease is accompanied by oxidative-stress-induced injury, concurrent inflammatory response, overactivated brain immune microenvironment, and disruption of the blood-brain barrier (BBB). This cascade of events ultimately leads to neuronal death and significantly impairs the recovery of neurological function. In this study, we presented extracellular vesicles derived from the gut probiotic Lactobacillus reuteri (LrEVs) integrated with brain targeting, reactive oxygen species (ROS) scavenging, and reduced infiltration of immune cells for effective multiple therapeutic interventions of IS. LrEVs inherited peptidoglycan (PGN) specifically targeted upregulated toll-like receptor 2 (TLR2) in the injured region of the ischemic brain, achieving the effective penetration of the BBB and accumulation in the ischemic brain. In the meantime, LrEVs prevented neuronal apoptosis after stroke by scavenging ROS overproduction and modulating microglial polarization through inhibition of the MAPK and NF-κB pathways. Furthermore, LrEVs inhibited the aggregation of C-C motif chemokine ligand 2 (CCL2), reduced the infiltration of peripheral immune cells such as macrophages and neutrophils into ischemic brain tissue, and suppressed the impairment of BBB, thereby improving the overactivated brain immune microenvironment. The findings provide a vesicle that combines ROS scavenging and modulation of the immune microenvironment, showcasing the potential of gut-probiotic-derived vesicles to treat neurological damage.
Collapse
Affiliation(s)
- Mengdi Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jinghan Ma
- Institution of Life Science, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ge Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Mingzhu Song
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ruizhen Lv
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jia Liang
- Institution of Life Science, Jinzhou Medical University, Jinzhou 121000, PR China
- Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, PR China
- Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
- Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, PR China
- Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121000, PR China
| |
Collapse
|
2
|
Zhu J, Peng F, Yang H, Luo J, Zhang L, Chen X, Liao H, Lei H, Liu S, Yang T, Luo G, Chen G, Zhao H. Probiotics and muscle health: the impact of Lactobacillus on sarcopenia through the gut-muscle axis. Front Microbiol 2025; 16:1559119. [PMID: 40160272 PMCID: PMC11952772 DOI: 10.3389/fmicb.2025.1559119] [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: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Sarcopenia refers to the decline in skeletal muscle mass and function. Due to its increased mortality rate and severe disability, the clinical importance of sarcopenia is becoming increasingly prominent. Although the exact cause of sarcopenia is not fully understood, the gut microbiota (GM) plays a crucial role in the pathogenesis of sarcopenia, and increasing evidence suggests that gut dysbiosis may be associated with disease development. In the past few decades, the use of probiotics has surged, few studies have explored their impact on sarcopenia prevention and treatment. Lactobacillus probiotics are commonly used for gut health and immune support, but their mechanism in sarcopenia via the gut-muscle axis remains uncertain. This review highlights the treatment challenges, GM's role in sarcopenia, and the potential of Lactobacillus as an adjunct therapy. In addition, we also discuss the possible mechanisms by which Lactobacillus affect muscle function, such as alleviating inflammatory states, clearing excessive reactive oxygen species (ROS), improving skeletal muscle metabolism, enhancing intestinal barrier function and modulating the gut microbiota and its metabolites. These mechanisms may collectively contribute to the preservation of muscle mass and function, offering a promising avenue for advancing microbial therapies for sarcopenia.
Collapse
Affiliation(s)
- Jingjun Zhu
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fei Peng
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huixin Yang
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, China
| | - Jing Luo
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Zhang
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaolong Chen
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huazhi Liao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hao Lei
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shuai Liu
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Tingqian Yang
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Guanghua Luo
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guodong Chen
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Wan X, Zhang C, Lei P, Wang H, Chen R, Yang Q, Cheng Y, Wu W, Sun D, Hong X. Precision therapeutics for inflammatory bowel disease: advancing ROS-responsive nanoparticles for targeted and multifunctional drug delivery. J Mater Chem B 2025; 13:3245-3269. [PMID: 39905851 DOI: 10.1039/d4tb02868f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a severe chronic intestinal disorder with a rising global incidence. Current therapies, including the delivery of anti-inflammatory drugs and probiotics, face significant challenges in terms of safety, stability, and efficacy. In IBD patients, the activity of antioxidant enzymes (e.g., superoxide dismutase, glutathione peroxidase, and glutathione reductase) is reduced at the site of intestinal inflammation, leading to the accumulation of reactive oxygen species (ROS). This accumulation damages the intestinal mucosa, disrupts tight junctions between cells, and compromises the integrity of the intestinal barrier, exacerbating IBD symptoms. Therefore, nanoparticles responsive to ROS and capable of mimicking antioxidant enzyme activity, such as boronates, polydopamine, sulfides, and metal nanozymes, have emerged as promising tools. These nanoparticles can respond to elevated ROS levels in inflamed intestinal regions and release drugs to effectively neutralize ROS, making them ideal candidates for IBD treatment. This review discusses the application of various ROS-responsive nanomaterial delivery systems in IBD therapy, highlights current challenges, and outlines future research directions. Furthermore, we explore the "layered programmable delivery" strategy, which combines ROS-responsive nanoparticles with pH-responsive and cell membrane-targeted nanoparticles. This strategy has the potential to overcome the limitations of single-mechanism targeted drug delivery, enabling multi-range and multi-functional treatment approaches that significantly enhance delivery efficiency, providing new insights for the future of localized IBD treatment.
Collapse
Affiliation(s)
- Xiuping Wan
- Department of Gastroenterology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Caijie Zhang
- The People's Hospital of Yuhuan (Yuhuan People's Hospital Health Community Group), Taizhou 317600, China
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Hanbing Wang
- Department of biotechnology, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yongwei Cheng
- National Engineering Research Center of Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University, Wenzhou 325000, China
- MedTech (Wenzhou) Health Innovation Achievement Transformation Institute, Wenzhou Institute of Industry & Science, Wenzhou 325000, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Xiaofei Hong
- Department of Gastroenterology, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu Central Hospital, Yiwu 322000, China.
| |
Collapse
|
4
|
Ibrahim D, Khater SI, Sherkawy HS, Elgamal A, Hasan AA, Muhammed AA, Farag MFM, Eissa SA, Ismail TA, Eissa HM, Eskandrani AA, Alansari WS, El-Emam MMA. Protective Role of Nano-encapsulated Bifidobacterium breve, Bacilllus coagulans, and Lactobacillus plantarum in Colitis Model: Insights Toward Propagation of Short-Chain Fatty Acids and Reduction of Exaggerated Inflammatory and Oxidative Response. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10472-y. [PMID: 39900879 DOI: 10.1007/s12602-025-10472-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
Irritable bowel disease (IBD), also known as ulcerative colitis and Crohn's disease, is a chronic inflammatory disorder affecting millions of people worldwide. Herein, nano-encapsulated multi-strain probiotics formulation, comprising Bifidobacterium breve DSM24732 and B. coagulans SANK 70258 and L. plantarum DSM24730 (BBLNPs) is used as an effective intervention technique for attenuating IBD through gut microenvironment regulation. The efficacy of the prophylactic role of BBLNPs in alleviating injury induced by dextran sulfate sodium (DSS) was evaluated by assessing oxidative and inflammatory responses, levels of short-chain fatty acids (SCFAs) and their regulation on GPR41/43 pathway, expression of genes related to tight-junctions and autophagy, immunohistochemistry of IL1β and GPR43, and histological examination of inflamed colonic tissue. The severity of clinical signs and paracellular permeability to FITC (fluorescein isothiocyanate)-labeled dextran was significantly decreased after BBLNP treatment. Reduction of oxidative stress-associated biomarkers (MDA, ROS, and H2O2) and acceleration of antioxidant enzyme activities (SOD, CAT, and GSH-Px) were noted in the BBLNP-treated group. Subsiding of inflammatory markers (TNF-α, IL-18, IL-6, TRL-4, CD-8, NLRP3, and caspase 1) and upregulation of tight-junction-related genes (occludin and JAM) was detected in BBLNPs. Administration of BBLNPs remarkably resulted in a higher level of SCFAs which parrel with colonic upregulation of GPR41 and GPR43 expression compared to DSS-treated rats. Notable modulation of autophagy-related genes (p62, mTOR, LC3, and Beclin-1) was identified post BBLNP treatment. The mRNA expressions of p62 and mTOR were significantly downregulated, while LC3 and Beclin-1 were upregulated after prophylactic treatment with BBLNPs. Immune-stained labeled cells showed lower expression of IL-1β and higher expression levels of GPR43 in BBLNPs compared to the DSS-induced group. The intestinal damage caused by DSSwas effectively mitigated by oral BBLNP treatment, as supported by the restoration of healthy colonic tissue architecture. The findings suggest that BBLNPs have a promising avenue in the remission of IBD by modulating inflammation, oxidative stress, microbial metabolites such as SCFAs, and autophagy.
Collapse
Affiliation(s)
- Doaa Ibrahim
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Safaa I Khater
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Hoda S Sherkawy
- Department of Medical Biochemistry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Aya Elgamal
- Department of Animal Histology and Anatomy, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Cairo, Egypt
| | - Asmaa A Hasan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Asmaa A Muhammed
- Department of Medical Physiology, Faculty of Medicine, Aswan University, Aswan, 81511, Egypt
| | - Mohamed F M Farag
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Samar A Eissa
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Tamer Ahmed Ismail
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Hemmat M Eissa
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Areej A Eskandrani
- Chemistry Department, College of Science, Taibah University, Medina, Saudi Arabia
| | - Wafa S Alansari
- Biochemistry Department, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mahran Mohamed Abd El-Emam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
5
|
Ma C, Zheng X, Zhang Q, Renaud SJ, Yu H, Xu Y, Chen Y, Gong J, Cai Y, Hong Y, Li H, Liao Q, Guo Y, Kang L, Xie Z. A postbiotic exopolysaccharide synergizes with Lactobacillus acidophilus to reduce intestinal inflammation in a mouse model of colitis. Int J Biol Macromol 2025; 291:138931. [PMID: 39732236 DOI: 10.1016/j.ijbiomac.2024.138931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease marked by gut inflammation and microbial dysbiosis. Exopolysaccharides (EPS) from probiotic bacteria have been shown to regulate microbial composition and metabolism, but their role in promoting probiotic growth and alleviating inflammation in UC remains unclear. Here, we investigate BLEPS-1, a novel EPS derived from Bifidobacterium longum subsp. longum XZ01, for its ability to promote the growth of Lactobacillus strains. We then tested a synbiotic formulation of BLEPS-1 and L. acidophilus in a DSS-induced UC mouse model. The combination of BLEPS-1 and L. acidophilus alleviated DSS-induced intestinal inflammation, outperforming either component alone. Administration of BLEPS-1 decreased the proportion of M1 macrophages in the intestine, while M2 macrophages were more abundant following L. acidophilus treatment. Together, BLEPS-1 and L. acidophilus synergistically modulated macrophage polarization toward the M2-type. Administration of BLEPS-1 and L. acidophilus together modulated gut microbiota composition and altered the gut metabolic profile, with BLEPS-1 and L. acidophilus promoting metabolism of short-chain fatty acids and aromatic amino acids, respectively. Our study identified a novel synbiotic formulation with potent immunomodulatory and metabolic activity, laying the groundwork for a promising therapeutic strategy to treat intestinal inflammatory diseases such as colitis.
Collapse
Affiliation(s)
- Chong Ma
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Xiaobin Zheng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qian Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Stephen James Renaud
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada
| | - Hansheng Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Yaning Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Yuchun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Jing Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yonghua Cai
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanjun Hong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Hao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ying Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, China
| | - Liang Kang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510006, China.
| |
Collapse
|
6
|
Zhou L, Wong KY, Xie H. Modulation of Intestinal Inflammation and Protection of Dopaminergic Neurons in Parkinson's Disease Mice through a Probiotic Formulation Targeting NLRP3 Inflammasome. J Neuroimmune Pharmacol 2025; 20:9. [PMID: 39826038 PMCID: PMC11742874 DOI: 10.1007/s11481-024-10163-5] [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: 07/10/2024] [Accepted: 11/29/2024] [Indexed: 01/20/2025]
Abstract
Emerging evidence highlights the significance of peripheral inflammation in the pathogenesis of Parkinson's disease (PD) and suggests the gut as a viable therapeutic target. This study aimed to explore the neuroprotective effects of the probiotic formulation VSL#3® and its underlying mechanism in a PD mouse model induced by MPTP. Following MPTP administration, the striatal levels of dopamine and its metabolites, as along with the survival rate of dopaminergic neurons in the substantia nigra, were significantly reduced in PD mice. MPTP also significantly increased the mRNA expression of pro-inflammatory cytokines TNF-α and IL-1β, while reducing anti-inflammation mediators, like glia cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) in the striatum. These pathological changes were notably mitigated by VSL#3® treatment, suggesting its neuroprotective and anti-inflammatory effects in the brain. Additionally, VSL#3® significantly lowered the circulating levels of pro-inflammatory cytokines, and reduced TNF-α and IL-1β mRNA expression in the liver, indicating an inhibition of cytokine transfer. In the intestine, the probiotic treatment markedly decreased the mRNA expression of pro-inflammatory cytokines, (TNF-α, IL-1β, IL-6 and IL-17), and the other two key components of the NLRP3 inflammasome, caspase-1 and NLRP3, demonstrating an inhibition of VSL#3® on gut NLRP3 inflammasome. VSL#3® exerts neuroprotective effects in PD mice through the suppression of intestinal inflammation, particularly inhibiting the intestinal NLRP3 inflammasome. This study supports the therapeutic potential of targeting intestinal inflammation and utilizing probiotics in PD treatment.
Collapse
Affiliation(s)
- Liping Zhou
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Ka Ying Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Hongxiang Xie
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, PR China.
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, PR China.
| |
Collapse
|
7
|
Li JK, Veeraperumal S, Aweya JJ, Liu Y, Cheong KL. Fucoidan modulates gut microbiota and immunity in Peyer's patches against inflammatory bowel disease. Carbohydr Polym 2024; 342:122421. [PMID: 39048206 DOI: 10.1016/j.carbpol.2024.122421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Although fucoidan has potential use as an anti-inflammatory agent, the specific mechanisms by which it influences signaling and immunomodulatory pathways between gut microbiota and Peyer's patches remain unclear. Therefore, the aim of this study was to investigate the therapeutic potential of fucoidan in a dextran sulfate sodium (DSS)-induced mouse model of inflammatory bowel disease (IBD) by examining the effects on gut microbiota and the underlying anti-inflammatory mechanisms. Purified fucoidan, which upon characterization revealed structural fragments comprising →3)-β-D-Galp-(1→, →4)-α-L-Fucp-(1→, and →3)-α-L-Fucp-(1→ residues with a sulfation at position C2 was used. Treatment of the mice with fucoidan significantly alleviated the symptoms of IBD and restored the diversity of gut microbiota by enhancing the abundance of Bacteroidetes and reducing the proportion of Firmicutes. The administration of fucoidan also elevated levels of short-chain fatty acids while reducing the levels of pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ. Most importantly, fucoidan attenuated the expression of integrin α4β7/MAdCAM-1 and CCL25/CCR9, which are involved in homing intestinal lymphocytes within Peyer's patches. These findings indicate that fucoidan is a promising gut microbiota modulator and an anti-inflammatory agent for IBD.
Collapse
Affiliation(s)
- Jia-Kang Li
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China
| | - Suresh Veeraperumal
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China
| | - Jude Juventus Aweya
- College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian, China
| | - Yang Liu
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China
| | - Kit-Leong Cheong
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China.
| |
Collapse
|
8
|
Freitas ADS, Barroso FAL, Campos GM, Américo MF, Viegas RCDS, Gomes GC, Vital KD, Fernandes SOA, Carvalho RDDO, Jardin J, Miranda APGDS, Ferreira E, Martins FS, Laguna JG, Jan G, Azevedo V, de Jesus LCL. Exploring the anti-inflammatory effects of postbiotic proteins from Lactobacillus delbrueckii CIDCA 133 on inflammatory bowel disease model. Int J Biol Macromol 2024; 277:134216. [PMID: 39069058 DOI: 10.1016/j.ijbiomac.2024.134216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Lactobacillus delbrueckii CIDCA 133 is a promising health-promoting bacterium shown to alleviate intestinal inflammation. However, the specific bacterial components responsible for these effects remain largely unknown. Here, we demonstrated that consuming extractable proteins from the CIDCA 133 strain effectively relieved acute ulcerative colitis in mice. This postbiotic protein fraction reduced the disease activity index and prevented colon shortening in mice. Furthermore, histological analysis revealed colitis prevention with reduced inflammatory cell infiltration into the colon mucosa. Postbiotic consumption also induced an immunomodulatory profile in colitic mice, as evidenced by both mRNA transcript levels (Tlr2, Nfkb1, Nlpr3, Tnf, and Il6) and cytokines concentration (IL1β, TGFβ, and IL10). Additionally, it enhanced the levels of secretory IgA, upregulated the transcript levels of tight junction proteins (Hp and F11r), and improved paracellular intestinal permeability. More interestingly, the consumption of postbiotic proteins modulated the gut microbiota (Bacteroides, Arkkemansia, Dorea, and Oscillospira). Pearson correlation analysis indicated that IL10 and IL1β levels were positively associated with Bacteroides and Arkkemansia_Lactobacillus abundance. Our study reveals that CIDCA 133-derived proteins possess anti-inflammatory properties in colonic inflammation.
Collapse
Affiliation(s)
- Andria Dos Santos Freitas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Gabriela Munis Campos
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Monique Ferrary Américo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Gabriel Camargos Gomes
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Kátia Duarte Vital
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | - Enio Ferreira
- Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Guimarães Laguna
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Vasco Azevedo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| | - Luís Cláudio Lima de Jesus
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
9
|
Chen T, Jin N, Zhang Q, Li Z, Wang Q, Fang X. Auraptene Mitigates Colitis Induced by Dextran Sulfate Sodium in Mice by Regulating Specific Intestinal Flora and Repairing the Intestinal Barrier. Inflammation 2024; 47:1127-1141. [PMID: 38236384 DOI: 10.1007/s10753-023-01965-5] [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: 10/14/2023] [Revised: 12/11/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024]
Abstract
Auraptene (AUT) is widely known to possess both antioxidant and anti-inflammatory properties. This study attempted to evaluate the protective effects of AUT in dextran sodium sulfate (DSS)-induced colitis in mice and to determine the underlying molecular mechanisms. Our results suggest that AUT substantially minimizes the severity and worsening of DSS-induced colitis in mice, indicated by the lengthening of the colon, lower disease activity index, reduced oxidation levels, and attenuated inflammatory factors. Molecular studies revealed that AUT reduces the nuclear translocation of nuclear factor-κB (NF-κB), thereby inhibiting the expression of inflammatory factors. Additionally, AUT promotes the diversity of the intestinal flora in mice with colitis by increasing the number of beneficial bacteria such as Lactobacillaceae and lowering the number of harmful bacteria. In conclusion, AUT mitigates DSS-induced colitis by maintaining the integrity of the intestinal barrier and modulating the levels of the intestinal microbial species.
Collapse
Affiliation(s)
- Tong Chen
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Naizhong Jin
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Qi Zhang
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Zhongming Li
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Qiutao Wang
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| |
Collapse
|
10
|
Gu X, Liao S, Li M, Wang J, Tan B. Chloroquine Downregulation of Intestinal Autophagy Changed Intestinal Microbial Community Compositions and Metabolite Profiles in Piglets. Vet Sci 2024; 11:333. [PMID: 39195787 PMCID: PMC11360670 DOI: 10.3390/vetsci11080333] [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: 06/04/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 08/29/2024] Open
Abstract
Our previous study demonstrated that moderate inhibition of intestinal autophagy was beneficial to alleviate early weaning stress in piglets, but the detailed mechanism behind this was unclear. Microbiota-mediated enterocyte autophagy helps maintain intestinal homeostasis. This study investigated the effects of inhibition or activation of autophagy in intestinal microbial community compositions and metabolite profiles in piglets. Eighteen 24-day-old weaned piglets were divided into three groups (each treatment of six piglets) and treated daily with rapamycin (RAPA), chloroquine (CQ) or a control volume of normal saline (CON group). Before the formal trial, the piglets were allowed to acclimatize for 3 days, and then the trial period was 14 days. Collected samples from the ileum and colon underwent 16S rRNA gene sequencing and metabolite analysis. Significant differences in microbial composition were observed in both the ileum and colon of the RAPA and CQ groups compared to the CON group (p < 0.05). In addition, the relative levels of abundance of Peptostreptococcus, Fusobacterium, Dialister, Selenomonas and Oceanobacillus in the ileum and Porphyromonas, Bacteroides, unidentified_Lachnospiraceae, Akkermansia, Sharpea, Peptococcus, Pseudoalteromonas, Peptoclostridium and unidentified_Acidobacteria in the colon were improved in piglets fed the RAPA diet, whereas the relative levels of abundance of Turicibacter, Rickettsiella and Sarcina in the ileum and Roseburia and Kroppenstedtia in the colon were enhanced in the CQ group (p < 0.05). Meanwhile, metabolomic analysis showed that there were significant differences in metabolites among all groups (p < 0.05), and KEGG enrichment analysis revealed that differential metabolites were mainly enriched in the ABC transporters and biosynthesis of amino acids pathways. Furthermore, these metabolites were closely related to differential microorganisms (p < 0.05). Overall, autophagy inhibition regulates the composition of intestinal microorganisms and their metabolites, and these differential metabolites are significantly correlated with differential intestinal microorganisms, which may in turn affect the production performance of weaned piglets.
Collapse
Affiliation(s)
- Xueling Gu
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.G.); (S.L.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China;
| | - Simeng Liao
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.G.); (S.L.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China;
| | - Meng Li
- Yuelushan Laboratory, Changsha 410128, China;
| | - Jing Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.G.); (S.L.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China;
| | - Bie Tan
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.G.); (S.L.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China;
| |
Collapse
|
11
|
Ma J, Xie H, Yuan C, Shen J, Chen J, Chen Q, Liu J, Tong Q, Sun J. The gut microbial signatures of patients with lacunar cerebral infarction. Nutr Neurosci 2024; 27:620-636. [PMID: 37538045 DOI: 10.1080/1028415x.2023.2242121] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND Emerging evidence revealed that gut microbial dysbiosis is involved in the pathogenesis of multiple neurological diseases, but there is little available data on the relationship between gut microbiota and lacunar cerebral infarction (LCI). METHODS Fecal samples from acute LCI patients (n = 65) and matched healthy controls (n = 65) were collected. The compositions and potential functions of the gut microbiota were estimated. RESULTS The results showed that there were significant gut microbial differences between LCI and control groups. Patients with LCI had higher abundances of genus Lactobacillus, Streptococcus, Veillonella, Acidaminococcus, Bacillus, Peptoclostridium, Intestinibacter, Alloscardovia and Cloacibacillus but lower proportions of genus Agathobacter and Lachnospiraceae_UCG-004. Investigating further these microbes such as Lactobacillus and Veillonella were correlated with clinical signs. Moreover, we found that 9 gene functions of gut microbiota were different between LCI patients and controls, which were associated with amino acid metabolism and inflammatory signal transduction. Notably, four optimal microbial markers were determined, and the combination of Streptococcus, Lactobacillus, Agathobacter, Lachnospiraceae_UCG-004 and the three risk factors achieved an area under the curve (AUC) value of 0.854 to distinguish LCI from controls. CONCLUSION These findings revealed the characterizing of gut microbiota in LCI patients and provided potential microbial biomarkers for clinical diagnosis of LCI.
Collapse
Affiliation(s)
- Jiaying Ma
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Huijia Xie
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chengxiang Yuan
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jie Shen
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jiaxin Chen
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qionglei Chen
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qiuling Tong
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jing Sun
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
12
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
13
|
Wei FH, Xie WY, Zhao PS, Gao W, Gao F. Echinacea purpurea Polysaccharide Ameliorates Dextran Sulfate Sodium-Induced Colitis by Restoring the Intestinal Microbiota and Inhibiting the TLR4-NF-κB Axis. Nutrients 2024; 16:1305. [PMID: 38732552 PMCID: PMC11085647 DOI: 10.3390/nu16091305] [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: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic intestinal ailment which cannot be completely cured. The occurrence of UC has been on the rise in recent years, which is highly detrimental to patients. The effectiveness of conventional drug treatment is limited. The long-term usage of these agents can lead to substantial adverse effects. Therefore, the development of a safe and efficient dietary supplement is important for the prevention of UC. Echinacea purpurea polysaccharide (EPP) is one of the main bioactive substances in Echinacea purpurea. EPP has many favorable effects, such as antioxidative, anti-inflammatory, and antitumor effects. However, whether EPP can prevent or alleviate UC is still unclear. This study aims to analyze the effect and mechanism of EPP on UC in mice using a 3% dextran sulfate sodium (DSS)-induced UC model. The results showed that dietary supplementation with 200 mg/kg EPP significantly alleviated the shortening of colon length, weight loss, and histopathological damage in DSS-induced colitis mice. Mechanistically, EPP significantly inhibits the activation of the TLR4/NF-κB pathway and preserves the intestinal mechanical barrier integrity by enhancing the expression of claudin-1, ZO-1, and occludin and reducing the loss of goblet cells. Additionally, 16S rRNA sequencing revealed that EPP intervention reduced the abundance of Bacteroides, Escherichia-Shigella, and Klebsiella; the abundance of Lactobacillus increased. The results of nontargeted metabonomics showed that EPP reshaped metabolism. In this study, we clarified the effect of EPP on UC, revealed the potential function of EPP, and supported the use of polysaccharide dietary supplements for UC prevention.
Collapse
Affiliation(s)
| | | | | | | | - Fei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.-H.W.); (W.-Y.X.); (P.-S.Z.); (W.G.)
| |
Collapse
|
14
|
Zhou K, Xie J, Su Y, Fang J. Lactobacillus reuteri for chronic periodontitis: focus on underlying mechanisms and future perspectives. Biotechnol Genet Eng Rev 2024; 40:381-408. [PMID: 36856460 DOI: 10.1080/02648725.2023.2183617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023]
Abstract
Chronic periodontitis is a common oral disorder caused by pathogenic bacteria. Despite the wide use of antibiotics as the conventional adjunctive treatment, the challenges of increased antibiotic resistance and limited therapeutic effect receive considerable attention and the developments of alternative treatments gain increasing consideration. Growing evidence showed that Lactobacillus reuteri (LR) may represent a promising alternative adjunct for chronic periodontitis. It can attenuate inflammation and reduce tissue disruption. LR-assisted treatment has been shown to be effective and relatively safe in multiple clinical trials, and accumulating evidence suggests its significant biological roles. In the current review, we focus on capturing the underlying mechanisms of LR involved in chronic periodontitis, thereby representing a scientific foundation for LR-assisted therapy. Furthermore, we point out the challenges and future directions for further clinical trials to improve the clinical applicability for LR.
Collapse
Affiliation(s)
- Keyi Zhou
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| | - Jiaman Xie
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| | - Yuan Su
- Department of Periodontology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| | - Jingxian Fang
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
15
|
Li T, Zhu K, Wang L, Dong Y, Huang J. Stabilization by Chaperone GroEL in Biogenic Selenium Nanoparticles Produced from Bifidobacterium animalis H15 for the Treatment of DSS-Induced Colitis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13439-13452. [PMID: 38456847 DOI: 10.1021/acsami.3c16340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Inflammatory bowel diseases have a high rate of mortality and pose a serious threat to global public health. Selenium is an essential trace element, which has been shown to play important roles in redox control and antioxidant defense. Microorganisms play important roles in the reduction of toxic inorganic selenium (selenite and selenate) to less-toxic biogenic selenium nanoparticles (Bio-SeNPs), which have higher biocompatibility. In the present study, novel Bio-SeNPs with high stability were synthesized using probiotic Bifidobacterium animalis subsp. lactis H15, which was isolated from breastfed infant feces. The Bio-SeNPs with a size of 122 nm showed stability at various ionic strengths, temperatures, and in simulated gastrointestinal fluid, while chemosynthetic SeNPs underwent aggregation. The main surface protein in the Bio-SeNPs was identified as chaperone GroEL by liquid chromatography-tandem mass spectrometry. The overexpression and purification of GroEL demonstrated that GroEL controlled the assembly of Bio-SeNPs both in vitro and in vivo. In vivo, oral administration of Bio-SeNPs could alleviate dextran sulfate sodium-induced colitis by decreasing cell apoptosis, increasing antioxidant capacity and the number of proliferating cells, and improving the function of the intestinal mucosal barrier. In vitro experiments verified that Bio-SeNPs inhibited lipopolysaccharide-induced toll-like receptor 4/NF-κB signaling pathway activation. These results suggest that the Bio-SeNPs with high stability could have potential as a nutritional supplement for the treatment of colitis in nanomedicine applications.
Collapse
Affiliation(s)
- Tong Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Kongdi Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Lianshun Wang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning 116023, China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiaqiang Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| |
Collapse
|
16
|
Bai B, Liu Q, Kong R, Jia Z, Chen H, Zhi W, Wang B, Ma C, Ma D. Role of Nrf2/HO-1 pathway on inhibiting activation of ChTLR15/ChNLRP3 inflammatory pathway stimulated by E. tenella sporozoites. Poult Sci 2024; 103:103445. [PMID: 38262340 PMCID: PMC10835464 DOI: 10.1016/j.psj.2024.103445] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/25/2024] Open
Abstract
The aim of this study is to explore whether Nrf2 antioxidant pathway negatively regulates the ChTLR15/NLRP3 inflammatory pathway stimulated by Eimeria tenella infection. Firstly, levels of molecules in the Nrf2/HO-1 pathway in DF-1 cells pre-treated with an optimized dose of Corilagine or probiotics Levilactobacillus brevis 23017 were quantified using real-time PCR (qRT-PCR) and Western blot. Then, DF-1 cells pre-treated with Corilagine or L. brevis 23017 were stimulated with E. tenella sporozoites, and mRNA levels of molecules in Nrf2/HO-1 and ChTLR15/NLRP3 pathways, protein levels of p-Nrf2, Nrf2, HO-1, ChTLR15 and ChNLRP3, levels of malondialdehyde (MDA) and reactive oxygen species (ROS) were quantified. Further, expression level of Nrf2 and ChTLR15 in DF-1 cells was knocked down by RNA interfering (RNAi) method, and target cells were pre-treated with Corilagine or L. brevis 23017, followed by stimulation with E. tenella sporozoites, and the expression levels of key molecules in Nrf2/HO-1 and ChTLR15/NLRP3 pathways were quantified. The results showed that mRNA and protein levels of key molecules in the Nrf2/HO-1 pathway in DF-1 cells was significantly upregulated after pretreating with 15 μM Corilagine and supernatant of L. brevis 23017. After stimulating with E. tenella sporozoites, levels of molecules in the ChTLR15/NLRP3 pathway, levels of MDA and ROS in DF-1 cells pre-treated with 15 μM Corilagine or bacterial supernatant were all significantly down-regulated. The results from the knock-down experiment also displayed that Corrigine and L. brevis 23017 inhibited the activation of the ChTLR15/ChNLRP3 inflammatory pathway stimulated by E. tenella sporozoites through activating Nrf2/HO-1 antioxidant pathway. This study provides new ideas for the development of novel anticoccidial products.
Collapse
Affiliation(s)
- Bingrong Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qiuju Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Rui Kong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhipeng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenjing Zhi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Biao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunli Ma
- College of Food Science, Northeast Agricultural University, Harbin, China
| | - Dexing Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China; Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
17
|
Sun Z, Huang S, Yan X, Zhang X, Hao Y, Jiang L, Dai Z. Living, Heat-Killed Limosilactobacillus mucosae and Its Cell-Free Supernatant Differentially Regulate Colonic Serotonin Receptors and Immune Response in Experimental Colitis. Nutrients 2024; 16:468. [PMID: 38398793 PMCID: PMC10893098 DOI: 10.3390/nu16040468] [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: 01/18/2024] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Lactobacillus species have been shown to alleviate gut inflammation and oxidative stress. However, the effect of different lactobacilli components on gut inflammation has not been well studied. This study aims to identify the differences in the effect and mechanisms of different forms and components of Limosilactobacillus mucosae (LM) treatment in the alleviation of gut inflammation using a colitis mouse model that is induced by dextran sodium sulfate (DSS). Seventy-two C57BL/6 mice were divided into six groups: control, DSS, live LM+DSS (LM+DSS), heat-killed LM+DSS (HKLM+DSS), LM cell-free supernatant + DSS (LMCS+DSS), and MRS medium + DSS (MRS+DSS). The mice were treated with different forms and components of LM for two weeks before DSS treatment. After that, the mice were sacrificed for an assessment of their levels of inflammatory cytokines, serotonin (5-HT) receptors (HTRs), and tryptophan metabolites. The results showed that, compared to other treatments, LMCS was more effective (p < 0.05) in the alleviation of DSS-induced body weight loss and led to an increase in the disease activity index score. All three forms and components of LM increased (p < 0.05) the levels of indole-3-acetic acid but reduced (p < 0.05) the levels of 5-HT in the colon. HKLM or LMCS reduced (p < 0.05) the percentages of CD3+CD8+ cytotoxic T cells but increased (p < 0.05) the percentages of CD3+CD4+ T helper cells in the spleen. LM or HKLM increased (p < 0.05) abundances of CD4+Foxp3+ regulatory T cells in the spleen. The LM and LMCS treatments reduced (p < 0.05) the expression of the pro-inflammatory cytokines Il6 and Il17a. The mice in the HKLM+DSS group had higher (p < 0.05) mRNA levels of the anti-inflammatory cytokine Il10, the cell differentiation and proliferation markers Lgr5 and Ki67, the 5-HT degradation enzyme Maoa, and HTRs (Htr1a, Htr2a, and Htr2b) in the colon. All three forms and components of LM reduced the phosphorylation of STAT3. The above findings can help to optimize the functionality of probiotics and develop new dietary strategies that aid in the maintenance of a healthy gut.
Collapse
Affiliation(s)
- Zhiyuan Sun
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (Z.S.); (X.Y.); (X.Z.); (Y.H.); (L.J.)
| | - Siqi Huang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Xing Yan
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (Z.S.); (X.Y.); (X.Z.); (Y.H.); (L.J.)
| | - Xiuwen Zhang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (Z.S.); (X.Y.); (X.Z.); (Y.H.); (L.J.)
| | - Youling Hao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (Z.S.); (X.Y.); (X.Z.); (Y.H.); (L.J.)
| | - Lili Jiang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (Z.S.); (X.Y.); (X.Z.); (Y.H.); (L.J.)
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (Z.S.); (X.Y.); (X.Z.); (Y.H.); (L.J.)
| |
Collapse
|
18
|
Guo X, Xu J, Huang C, Zhang Y, Zhao H, Zhu M, Wang J, Nie Y, Xu H, Zhou Y, Zhou Y. Rapamycin extenuates experimental colitis by modulating the gut microbiota. J Gastroenterol Hepatol 2023; 38:2130-2141. [PMID: 37916431 DOI: 10.1111/jgh.16381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 09/03/2023] [Accepted: 09/30/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND AND AIM Autophagy and gut microbiota correlates closely with the inflammatory bowel disease. Herein, we aimed to study the roles of rapamycin on the gut microbiota in inflammatory bowel disease. METHODS Acute colitis was induced with dextran sodium sulfate (DSS) and 2,4,6-trinitrobenzenesulfonic acid solution in mice. Mice were administered with rapamycin or hydroxychloroquine. Weight loss, disease activity index scores, histopathological score, serum inflammatory cytokines, intestinal permeability, and colonic autophagy-related proteins were detected. Cecal content was also preserved in liquid nitrogen and subsequently analyzed following the 16S DNA sequencing. The antibiotic cocktail-induced microbiome depletion was performed to further investigate the relationship between autophagy activation and gut microbiota. RESULTS Compared with the control group, the colonic autophagy-related proteins of P62, mTOR, and p-mTOR increased significantly, while the levels of LC3B and ATG16L1 decreased (all P < 0.05) in the model group. After rapamycin intervention, the colonic pathology of mice improved, while the disease activity index score decreased substantially; the colon length increased, and the expression of IL-6 and TNF-α decreased. Following hydroxychloroquine treatment, some indicators suggested aggravation of colitis. Principal coordinates analysis showed that the DSS group was located on a separate branch from the rapamycin group but was closer to the hydroxychloroquine group. Compared with the DSS group, the rapamycin group was associated with higher abundances of f_Lactobacillaceae (P = 0.0151), f_Deferribacteraceae (P = 0.0290), g_Lactobacillus (P = 0.0151), g_Mucispirillum (P = 0.0137), s_Lactobacillus_reuteri (P = 0.0028), and s_Clostridium_sp_Culture_Jar-13 (P = 0.0082) and a lower abundance of s_Bacteroides_sartorii (P = 0.0180). Linear discriminant analysis effect size showed that rapamycin increased the abundances of Lactobacillus-reuteri, Prevotellaceae, Paraprevotella, Christensenella and Streptococcus and decreased those of Peptostreptococcaceae and Romboutsia Bacteroides-sartorii. Besides, the improvement effect of autophagy activation on colitis disappears following gut microbiome depletion. CONCLUSION The therapeutic effects of rapamycin on extenuating experimental colitis may be related to the gut microbiota.
Collapse
Affiliation(s)
- Xue Guo
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yan Zhang
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Hailan Zhao
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Minzheng Zhu
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jiaqi Wang
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yuqiang Nie
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
19
|
Jiang Z, Jiang P, Ji S, Su D, Xu G, Zhang M. Research progress on Limosilactibacilus reuteri in diseases. Microbiol Res 2023; 276:127482. [PMID: 37660453 DOI: 10.1016/j.micres.2023.127482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/20/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
Limosilactibacilus reuteri (L. reuteri) is a gram-positive probiotic that does not produce peroxidase. Certain strains of L. reuteri have been approved for use in human health products in China. The probiotic mechanism of L. reuteri in organisms can be divided into two directions: first, L. reuteri directly regulates the gut microbiota and indirectly affecting the host; second, L. reuteri secretes substances that directly affect the host. Numerous studies have shown that a deficiency in this commensal bacterium is associated with various diseases in different systems (such as inflammation in the digestive system, systemic lupus erythematosus in the autoimmune system, metabolic syndrome in the endocrine system, and mastitis in the reproductive system). However, although recent studies have found that L. reuteri can also promote disease progression, but overall, it is more beneficial than harmful in general. Further, more in-depth experiments are needed to determine whether L. reuteri should be removed from probiotics in the future. In this review, we provide an overview of the research history of L. reuteri and conclude with the main mechanisms through which this intestinal symbiont can improve health or aggravate diseases.
Collapse
Affiliation(s)
- Zhuoru Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Ping Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Siqi Ji
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Dan Su
- FUJIFILM Diosynth Biotechnologies, Watertown 02472, MA, United States
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China.
| | - Mingming Zhang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China.
| |
Collapse
|
20
|
Lei P, Yu H, Ma J, Du J, Fang Y, Yang Q, Zhang K, Luo L, Jin L, Wu W, Sun D. Cell membrane nanomaterials composed of phospholipids and glycoproteins for drug delivery in inflammatory bowel disease: A review. Int J Biol Macromol 2023; 249:126000. [PMID: 37532186 DOI: 10.1016/j.ijbiomac.2023.126000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Inflammatory bowel disease (IBD) is a serious chronic intestinal disorder with an increasing global incidence. However, current treatment strategies, such as anti-inflammatory drugs and probiotics, have limitations in terms of safety, stability, and effectiveness. The emergence of targeted nanoparticles has revolutionized IBD treatment by enhancing the biological properties of drugs and promoting efficiency and safety. Unlike synthetic nanoparticles, cell membrane nanomaterials (CMNs) consist primarily of biological macromolecules, including phospholipids, proteins, and sugars. CMNs include red blood cell membranes, macrophage membranes, and leukocyte membranes, which possess abundant glycoprotein receptors and ligands on their surfaces, allowing for the formation of cell-to-cell connections with other biological macromolecules. Consequently, they exhibit superior cell affinity, evade immune responses, and target inflammation effectively, making them ideal material for targeted delivery of IBD therapies. This review explores various CMNs delivery systems for IBD treatment. However, due to the complexity and harsh nature of the intestinal microenvironment, the lack of flexibility or loss of selectivity poses challenges in designing single CMNs delivery strategies. Therefore, we propose a hierarchically programmed delivery modality that combines CMNs with pH, charge, ROS and ligand-modified responsive nanoparticles. This approach significantly improves delivery efficiency and points the way for future research in this area.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Li Luo
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523059, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
21
|
Li C, Peng K, Xiao S, Long Y, Yu Q. The role of Lactobacillus in inflammatory bowel disease: from actualities to prospects. Cell Death Discov 2023; 9:361. [PMID: 37773196 PMCID: PMC10541886 DOI: 10.1038/s41420-023-01666-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Inflammatory Bowel Disease (IBD), a chronic nonspecific intestinal inflammatory disease, is comprised of Ulcerative Colitis (UC) and Crohn's Disease (CD). IBD is closely related to a systemic inflammatory reaction and affects the progression of many intestinal and extraintestinal diseases. As one of the representative bacteria for probiotic-assisted therapy in IBD, multiple strains of Lactobacillus have been proven to alleviate intestinal damage and strengthen the intestinal immunological barrier, epithelial cell barrier, and mucus barrier. Lactobacillus also spares no effort in the alleviation of IBD-related diseases such as Colitis-associated Colorectal cancer (CAC), Alzheimer's Disease (AD), Depression, Anxiety, Autoimmune Hepatitis (AIH), and so on via gut-brain axis and gut-liver axis. This article aims to discuss the role of Lactobacillus in IBD and IBD-related diseases, including its underlying mechanisms and related curative strategies from the present to the future.
Collapse
Affiliation(s)
- Congxin Li
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kaixin Peng
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Siqi Xiao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Yuanyuan Long
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Qin Yu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China.
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
22
|
Peng Y, Ma Y, Luo Z, Jiang Y, Xu Z, Yu R. Lactobacillus reuteri in digestive system diseases: focus on clinical trials and mechanisms. Front Cell Infect Microbiol 2023; 13:1254198. [PMID: 37662007 PMCID: PMC10471993 DOI: 10.3389/fcimb.2023.1254198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Objectives Digestive system diseases have evolved into a growing global burden without sufficient therapeutic measures. Lactobacillus reuteri (L. reuteri) is considered as a new potential economical therapy for its probiotic effects in the gastrointestinal system. We have provided an overview of the researches supporting various L. reuteri strains' application in treating common digestive system diseases, including infantile colic, diarrhea, constipation, functional abdominal pain, Helicobacter pylori infection, inflammatory bowel disease, diverticulitis, colorectal cancer and liver diseases. Methods The summarized literature in this review was derived from databases including PubMed, Web of Science, and Google Scholar. Results The therapeutic effects of L. reuteri in digestive system diseases may depend on various direct and indirect mechanisms, including metabolite production as well as modulation of the intestinal microbiome, preservation of the gut barrier function, and regulation of the host immune system. These actions are largely strain-specific and depend on the activation or inhibition of various certain signal pathways. It is well evidenced that L. reuteri can be effective both as a prophylactic measure and as a preferred therapy for infantile colic, and it can also be recommended as an adjuvant strategy to diarrhea, constipation, Helicobacter pylori infection in therapeutic settings. While preclinical studies have shown the probiotic potential of L. reuteri in the management of functional abdominal pain, inflammatory bowel disease, diverticulitis, colorectal cancer and liver diseases, its application in these disease settings still needs further study. Conclusion This review focuses on the probiotic effects of L. reuteri on gut homeostasis via certain signaling pathways, and emphasizes the importance of these probiotics as a prospective treatment against several digestive system diseases.
Collapse
Affiliation(s)
- Yijing Peng
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Wuxi Children’s Hospital, Children’s Hospital of Jiangnan University, Wuxi, China
| | - Yizhe Ma
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Department of Pediatric, Jiangyin People’s Hospital of Nantong University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Yifan Jiang
- School of Medicine, Nantong University, Nantong, China
| | - Zhimin Xu
- College of Resources and Environment, Innovative Institute for Plant Health, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Renqiang Yu
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
23
|
Yao S, Zhao Y, Chen H, Sun R, Chen L, Huang J, Yu Z, Chen S. Exploring the Plasticity of Diet on Gut Microbiota and Its Correlation with Gut Health. Nutrients 2023; 15:3460. [PMID: 37571397 PMCID: PMC10420685 DOI: 10.3390/nu15153460] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Dietary habits have been proven to help alter the composition of gut microbiota, and exploring the impact of nutritional patterns on gut microbiota changes can help protect gut health. However, few studies have focused on the dietary impact on the gut microbiota over an experimental timeframe. In this study, 16S rRNA gene sequencing was employed to investigate the gut microbiota of mice under different dietary patterns, including AIN-93G diet (Control), high protein diet (HPD), high fiber diet (HFD), and switch diet (Switch). The alpha diversity of the HPD group significantly decreased, but HFD can restore this decline. During HPD, some genera were significantly upregulated (e.g., Feacalibaculum) and downregulated (e.g., Parabacteroides). However, after receiving HFD, other genera were upregulated (e.g., Akkermansia) and downregulated (e.g., Lactobacillus). In addition, the interaction between pathogenic bacteria was more pronounced during HPD, while the main effect was probiotics during HFD. In conclusion, the plasticity exhibited by the gut microbiota was subject to dietary influences, wherein disparate dietary regimens hold pivotal significance in upholding the well-being of the host. Therefore, our findings provide new ideas and references for the relationship between diets and gut microbiota.
Collapse
Affiliation(s)
- Siqi Yao
- Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha 410008, China;
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Yiming Zhao
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Hao Chen
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410078, China; (H.C.); (J.H.)
| | - Ruizheng Sun
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Liyu Chen
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410078, China; (H.C.); (J.H.)
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410078, China; (Y.Z.); (R.S.); (L.C.)
| | - Shuijiao Chen
- Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410008, China
| |
Collapse
|
24
|
Shen Y, Jiang B, Zhang C, Wu Q, Li L, Jiang P. Combined Inhibition of the TGF-β1/Smad Pathway by Prevotella copri and Lactobacillus murinus to Reduce Inflammation and Fibrosis in Primary Sclerosing Cholangitis. Int J Mol Sci 2023; 24:11010. [PMID: 37446187 DOI: 10.3390/ijms241311010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Primary sclerosing cholangitis (PSC) is a chronic cholestatic disease characterized by inflammation and fibrosis of the bile ducts. Cholestasis may lead to hepatic inflammation and fibrosis, and amelioration of cholestasis may allow recovery from inflammatory and fibrotic pathological damage. Prevotella copri (P. copri) interventions have been reported to significantly improve cholestasis and liver fibrosis in 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced PSC mouse models. Even though P. copri treatment alone cannot bring about recovery from DDC-induced inflammation, it increases the abundance of Lactobacillus murinus (L. murinus) compared with DDC treatment, which has been reported to have anti-inflammatory effects. The abundance of L. murinus still not recovering to a normal level may underlie hepatic inflammation in P. copri + DDC mice. Separate or combined interventions of P. copri and L. murinus were used to investigate the molecular mechanism underlying the improvement in PSC inflammation and fibrosis. P. copri and L. murinus significantly reduced the hepatic inflammatory cell aggregation and inflammatory factor expression as well as the hepatic collagen content and fibrin factor expression in the PSC mice. Further analysis of phosphorylation and dephosphorylation levels revealed that treating the PSC mice with the P. copri and L. murinus combined intervention inhibited the activity of the DDC-activated TGF-β1/Smad pathway, thereby reducing liver inflammation and fibrosis. The combination of P. copri and L. murinus inhibits the TGF-β1/Smad pathway and reduces inflammation and fibrosis in PSC.
Collapse
Affiliation(s)
- Yu Shen
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Baorong Jiang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Chenchen Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Qian Wu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Ping Jiang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| |
Collapse
|
25
|
Mignini I, Ainora ME, Di Francesco S, Galasso L, Gasbarrini A, Zocco MA. Tumorigenesis in Inflammatory Bowel Disease: Microbiota-Environment Interconnections. Cancers (Basel) 2023; 15:3200. [PMID: 37370812 PMCID: PMC10295963 DOI: 10.3390/cancers15123200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Colo-rectal cancer (CRC) is undoubtedly one of the most severe complications of inflammatory bowel diseases (IBD). While sporadic CRC develops from a typical adenoma-carcinoma sequence, IBD-related CRC follows different and less understood pathways and its pathophysiological mechanisms were not completely elucidated. In contrast to chronic inflammation, which is nowadays a well-recognised drive towards neoplastic transformation in IBD, only recently was gut microbiota demonstrated to interfere with both inflammation processes and immune-mediated anticancer surveillance. Moreover, the role of microbiota appears particularly complex and intriguing when also considering its multifaceted interactions with multiple environmental stimuli, notably chronic pathologies such as diabetes and obesity, lifestyle (diet, smoking) and vitamin intake. In this review, we presented a comprehensive overview on current evidence of the influence of gut microbiota on IBD-related CRC, in particular its mutual interconnections with the environment.
Collapse
Affiliation(s)
| | - Maria Elena Ainora
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy; (I.M.); (S.D.F.); (L.G.); (A.G.); (M.A.Z.)
| | | | | | | | | |
Collapse
|
26
|
Nosaka T, Murata Y, Takahashi K, Naito T, Ofuji K, Matsuda H, Ohtani M, Hiramatsu K, Imamura Y, Goi T, Nakamoto Y. Hepatocellular carcinoma progression promoted by 5-lipoxygenase activity in CD163(+) tumor-associated macrophages. Biomed Pharmacother 2023; 162:114592. [PMID: 36966664 DOI: 10.1016/j.biopha.2023.114592] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
Arachidonic acid 5-lipoxygenase (5-LOX), an enzyme that synthesizes leukotrienes (LTs), is involved in cancer development including proliferation, invasion, metastasis and drug resistance. However, the functional role of 5-LOX in hepatocellular carcinoma (HCC) remains to be elucidated. In this study, we analyzed the contribution of 5-LOX in HCC progression and investigated the potential of targeted therapy. Analysis of 86 resected HCC specimens and the clinical data of 362 cases of liver cancer from The Cancer Genome Atlas Liver Hepatocellular Carcinoma dataset, showed that 5-LOX expression was associated with postoperative survival. The cancer proliferative and stem cell potential were correlated with the levels of 5-LOX in CD163(+) tumor-associated macrophages (TAMs). In an HCC mouse model, CD163(+) TAMs expressed 5-LOX and produced LTB4 and LTC/D/E4; the 5-LOX inhibitor, zileuton, suppressed HCC progression. LTB4 and LTC/D/E4 promoted cancer proliferation and stem cell capacity via phosphorylation of extracellular signal-regulated kinase 1/2 and stem cell-associated genes. Taken together, we identified a novel mechanism of HCC progression in which CD163(+) TAMs express 5-LOX and produce LTB4 and LTC/D/E4, thereby enhancing the proliferative and stem cell potential of HCC cells. Furthermore, inhibition of 5-LOX activity regulates HCC progression, suggesting it has potential as a new therapeutic target.
Collapse
Affiliation(s)
- Takuto Nosaka
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yosuke Murata
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kazuto Takahashi
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tatsushi Naito
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kazuya Ofuji
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidetaka Matsuda
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masahiro Ohtani
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Katsushi Hiramatsu
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yoshiaki Imamura
- Division of Diagnostic Pathology/Surgical Pathology, University of Fukui Hospital, Fukui, Japan
| | - Takanori Goi
- First Department of Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| |
Collapse
|
27
|
Uesugi T, Mori S, Miyanaga K, Yamamoto N. GroEL Secreted from Bacillus subtilis Natto Exerted a Crucial Role for Anti-Inflammatory IL-10 Induction in THP-1 Cells. Microorganisms 2023; 11:1281. [PMID: 37317255 DOI: 10.3390/microorganisms11051281] [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: 04/19/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
Although diverse immunomodulatory reactions of probiotic bacteria have been reported, this effect via Bacillus subtilis natto remains unclear, despite its long consumption history in Japan and usage in Natto production. Hence, we performed a comparative analysis of the immunomodulatory activities of 23 types of B. subtilis natto isolated from Natto products to elucidate the key active components. Among the isolated 23 strains, the supernatant from B. subtilis strain 1 fermented medium showed the highest induction of anti-inflammatory IL-10 and pro-inflammatory IL-12 in THP-1 dendritic cells (THP-1 DC) after co-incubation. We isolated the active component from strain 1 cultured medium and employed DEAE-Sepharose chromatography with 0.5 M NaCl elution for fractionation. IL-10-inducing activity was specific to an approximately 60 kDa protein, GroEL, which was identified as a chaperone protein and was significantly reduced with anti-GroEL antibody. Differential expression analysis of strains 1 and 15, which had the lowest cytokine-producing activity, showed a higher expression of various genes involved in chaperones and sporulation in strain 1. Furthermore, GroEL production was induced in spore-forming medium. The present study is the first to show that the chaperone protein GroEL, secreted by B. subtilis natto during sporulation, plays a crucial role in IL-10 and IL-12 production in THP-1 DC.
Collapse
Affiliation(s)
- Taisuke Uesugi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Kanagawa, Japan
- Ezaki Glico Co., Ltd., 4-6-5 Utajima, Nishiyodogawa-ku, Osaka 555-8502, Osaka, Japan
| | - Suguru Mori
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Kanagawa, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Kanagawa, Japan
- Department of Infection and Immunity, School of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-Shi 329-0498, Tochigi, Japan
| | - Naoyuki Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Kanagawa, Japan
| |
Collapse
|
28
|
Wei Y, Qi M, Liu C, Li L. Astragalus polysaccharide attenuates bleomycin-induced pulmonary fibrosis by inhibiting TLR4/ NF-κB signaling pathway and regulating gut microbiota. Eur J Pharmacol 2023; 944:175594. [PMID: 36804541 DOI: 10.1016/j.ejphar.2023.175594] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
PURPOSE Astragalus polysaccharide (APS) is a naturally-occurring compound derived from Astragalus membranaceus with anti-inflammatory and antioxidant properties. However, its beneficial effects and mechanisms on pulmonary fibrosis are unknown. Gut microbiota impact lung diseases via the gut-lung axis. Herein, we investigated APS progression to intervene in pulmonary fibrosis via the toll-like receptor 4(TLR4)/nuclear factor-kappa B(NF-κB) signaling pathway and gut microbiota homeostasis regulation. METHODS We used bleomycin (BLM) to construct an idiopathic pulmonary fibrosis (IPF) mouse model and assessed the pathology with Masson, hematoxylin-eosin (HE), and Sirius red staining. Enzyme-linked immunosorbent assay (ELISA) kits were employed to evaluate the inflammatory cytokine levels. Western blot evaluated TLR4/NF-κB signaling pathway expression. TUNEL staining to detect apoptosis. Mice feces samples were gathered for 16S rRNA gene sequencing. RESULTS Our findings revealed that APS ameliorated the extent of damage and collagen deposition in lung tissues, reduced inflammatory cytokines TNF-α, IL-6, and IL-1β levels, and decreased apoptosis. APS might attenuate the inflammatory response through TLR4/NF-κB signaling pathway inhibition. Meanwhile, the IPF mice model exhibited dysregulation of gut microbiota, and these changes were restored after APS intervention. APS may increase the proportion of probiotics, decrease that of harmful bacteria, and balance the gut microbiota via regulating metabolic pathways. CONCLUSION APS ameliorated lung tissue injury in the IPF mice model, inhibited TLR4/NF-κB signaling pathway, suppressed inflammatory cytokines activation, and reduced apoptosis. Moreover, APS regulated the metabolism of gut microbiota besides beneficial bacteria content elevation.
Collapse
Affiliation(s)
- Yi Wei
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ming Qi
- Department of Primary Healthcare, Qingdao Hospital of Traditional Chinese Medicine, Qingdao, 266014, China
| | - Chao Liu
- Department of Medical Imaging, Qingdao Hospital of Traditional Chinese Medicine, Qingdao, 266014, China.
| | - Lujia Li
- Department of Health Care, People's Liberation Army Navy 971 Hospital, Qingdao, 266071, China.
| |
Collapse
|
29
|
Nishiyama K, Yong CC, Moritoki N, Kitazawa H, Odamaki T, Xiao JZ, Mukai T. Sharing of Moonlighting Proteins Mediates the Symbiotic Relationship among Intestinal Commensals. Appl Environ Microbiol 2023; 89:e0219022. [PMID: 36847513 PMCID: PMC10053696 DOI: 10.1128/aem.02190-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
The human gastrointestinal tract is inhabited by trillions of symbiotic bacteria that form a complex ecological community and influence human physiology. Symbiotic nutrient sharing and nutrient competition are the most studied relationships in gut commensals, whereas the interactions underlying homeostasis and community maintenance are not fully understood. Here, we provide insights into a new symbiotic relationship wherein the sharing of secreted cytoplasmic proteins, called "moonlighting proteins," between two heterologous bacterial strains (Bifidobacterium longum and Bacteroides thetaiotaomicron) was observed to affect the adhesion of bacteria to mucins. B. longum and B. thetaiotaomicron were cocultured using a membrane-filter system, and in this system the cocultured B. thetaiotaomicron cells showed greater adhesion to mucins compared to that shown by monoculture cells. Proteomic analysis showed the presence of 13 B. longum-derived cytoplasmic proteins on the surface of B. thetaiotaomicron. Moreover, incubation of B. thetaiotaomicron with the recombinant proteins GroEL and elongation factor Tu (EF-Tu)-two well-known mucin-adhesive moonlighting proteins of B. longum-led to an increase in the adhesion of B. thetaiotaomicron to mucins, a result attributed to the localization of these proteins on the B. thetaiotaomicron cell surface. Furthermore, the recombinant EF-Tu and GroEL proteins were observed to bind to the cell surface of several other bacterial species; however, the binding was species dependent. The present findings indicate a symbiotic relationship mediated by the sharing of moonlighting proteins among specific strains of B. longum and B. thetaiotaomicron. IMPORTANCE The adhesion of intestinal bacteria to the mucus layer is an important colonization strategy in the gut environment. Generally, the bacterial adhesion process is a characteristic feature of the individual cell surface-associated adhesion factors secreted by a particular bacterium. In this study, coculture experiments between Bifidobacterium and Bacteroides show that the secreted moonlighting proteins adhere to the cell surface of coexisting bacteria and alter the adhesiveness of the bacteria to mucins. This finding indicates that the moonlighting proteins act as adhesion factors for not only homologous strains but also for coexisting heterologous strains. The presence of a coexisting bacterium in the environment can significantly alter the mucin-adhesive properties of another bacterium. The findings from this study contribute to a better understanding of the colonization properties of gut bacteria through the discovery of a new symbiotic relationship between them.
Collapse
Affiliation(s)
- Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Cheng-Chung Yong
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| |
Collapse
|
30
|
Yu Z, Chen J, Liu Y, Meng Q, Liu H, Yao Q, Song W, Ren X, Chen X. The role of potential probiotic strains Lactobacillus reuteri in various intestinal diseases: New roles for an old player. Front Microbiol 2023; 14:1095555. [PMID: 36819028 PMCID: PMC9932687 DOI: 10.3389/fmicb.2023.1095555] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
Lactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., is a gut symbiont that can colonize many mammals. Since it was first isolated in 1962, a multitude of research has been conducted to investigate its function and unique role in different diseases as an essential probiotic. Among these, the basic functions, beneficial effects, and underlying mechanisms of L. reuteri have been noticed and understood profoundly in intestinal diseases. The origins of L. reuteri strains are diverse, with humans, rats, and piglets being the most common. With numerous L. reuteri strains playing significant roles in different intestinal diseases, DSM 17938 is the most widely used in humans, especially in children. The mechanisms by which L. reuteri improves intestinal disorders include protecting the gut barrier, suppressing inflammation and the immune response, regulating the gut microbiota and its metabolism, and inhibiting oxidative stress. While a growing body of studies focused on L. reuteri, there are still many unknowns concerning its curative effects, clinical safety, and precise mechanisms. In this review, we initially interpreted the basic functions of L. reuteri and its related metabolites. Then, we comprehensively summarized its functions in different intestinal diseases, including inflammatory bowel disease, colorectal cancer, infection-associated bowel diseases, and pediatric intestinal disorders. We also highlighted some important molecules in relation to the underlying mechanisms. In conclusion, L. reuteri has the potential to exert a beneficial impact on intestinal diseases, which should be further explored to obtain better clinical application and therapeutic effects.
Collapse
Affiliation(s)
- Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Hang Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenxuan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangfeng Ren
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Xin Chen ✉
| |
Collapse
|
31
|
Qin W, Xie Y, Ren Z, Xu C, Sun MA, Yin Z, Bao W. Integrative ATAC-seq and RNA-seq analyses of IPEC-J2 cells reveals porcine transcription and chromatin accessibility changes associated with Escherichia coli F18ac inhibited by Lactobacillus reuteri. Front Microbiol 2023; 14:1101111. [PMID: 36876070 PMCID: PMC9978113 DOI: 10.3389/fmicb.2023.1101111] [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: 11/20/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Escherichia coli is the main cause of postweaning diarrhea in pigs, leading to economic loss. As a probiotic, Lactobacillus reuteri has been used to inhibit E. coli in clinical applications; however, its integrative interactions with hosts remain unclear, especially in pigs. Here, we found that L. reuteri effectively inhibited E. coli F18ac adhering to porcine IPEC-J2 cells, and explored the genome-wide transcription and chromatin accessibility landscapes of IPEC-J2 cells by RNA-seq and ATAC-seq. The results showed that some key signal transduction pathways, such as PI3K-AKT and MAPK signaling pathways, were enriched in the differentially expressed genes (DEGs) between E. coli F18ac treatment with and without L. reuteri groups. However, we found less overlap between RNA-seq and ATAC-seq datasets; we speculated that this might be caused by histones modification through ChIP-qPCR detection. Furthermore, we identified the regulation of the actin cytoskeleton pathway and a number of candidate genes (ARHGEF12, EGFR, and DIAPH3) that might be associated with the inhibition of E. coli F18ac adherence to IPEC-J2 cells by L. reuteri. In conclusion, we provide a valuable dataset that can be used to seek potential porcine molecular markers of E. coli F18ac pathogenesis and L. reuteri antibacterial activity, and to guide the antibacterial application of L. reuteri.
Collapse
Affiliation(s)
- Weiyun Qin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yunxiao Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhanshi Ren
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chao Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming-An Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
32
|
Ramos Meyers G, Samouda H, Bohn T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients 2022; 14:5361. [PMID: 36558520 PMCID: PMC9788597 DOI: 10.3390/nu14245361] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
It is widely accepted that the gut microbiota plays a significant role in modulating inflammatory and immune responses of their host. In recent years, the host-microbiota interface has gained relevance in understanding the development of many non-communicable chronic conditions, including cardiovascular disease, cancer, autoimmunity and neurodegeneration. Importantly, dietary fibre (DF) and associated compounds digested by the microbiota and their resulting metabolites, especially short-chain fatty acids (SCFA), were significantly associated with health beneficial effects, such as via proposed anti-inflammatory mechanisms. However, SCFA metabolic pathways are not fully understood. Major steps include production of SCFA by microbiota, uptake in the colonic epithelium, first-pass effects at the liver, followed by biodistribution and metabolism at the host's cellular level. As dietary patterns do not affect all individuals equally, the host genetic makeup may play a role in the metabolic fate of these metabolites, in addition to other factors that might influence the microbiota, such as age, birth through caesarean, medication intake, alcohol and tobacco consumption, pathogen exposure and physical activity. In this article, we review the metabolic pathways of DF, from intake to the intracellular metabolism of fibre-derived products, and identify possible sources of inter-individual variability related to genetic variation. Such variability may be indicative of the phenotypic flexibility in response to diet, and may be predictive of long-term adaptations to dietary factors, including maladaptation and tissue damage, which may develop into disease in individuals with specific predispositions, thus allowing for a better prediction of potential health effects following personalized intervention with DF.
Collapse
Affiliation(s)
- Guilherme Ramos Meyers
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
- Doctoral School in Science and Engineering, University of Luxembourg, 2, Avenue de l'Université, 4365 Esch-sur-Alzette, Luxembourg
| | - Hanen Samouda
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
| |
Collapse
|
33
|
Inhibition of GABAAR or Application of Lactobacillus casei Zhang Alleviates Ulcerative Colitis in Mice: GABAAR as a Potential Target for Intestinal Epithelial Renewal and Repair. Int J Mol Sci 2022; 23:ijms231911210. [PMID: 36232509 PMCID: PMC9570049 DOI: 10.3390/ijms231911210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Emerging evidence indicates that the gamma−aminobutyric acid type A receptor (GABAAR) and Lactobacillus casei Zhang regulate colitis in a variety of ways, such as by participating in host immune and inflammatory responses, altering the gut microbiota, and influencing intestinal barrier function. However, not much is known about the mechanisms by which GABAAR and L. casei affect colon epithelial cell renewal and the interaction between GABAAR and L. casei during this process. To elucidate this, we established a dextran sulfate sodium (DSS)−induced model and measured the mouse body weights, colon length, the disease activity index (DAI), and histological scores. Our results indicated that inhibition of GABAAR alleviated the DSS−induced colitis symptoms, resulting in less weight loss and more intact colon tissue. Moreover, treatment with bicuculline (Bic, a GABAAR inhibitor) increased the levels of PCNA, β−catenin, and TCF4 in mice with colitis. Interestingly, open field test performances showed that inhibition of GABAAR also attenuated colitis−related anxiety−like behavior. By 16S RNA gene sequencing analysis, we showed that inhibition of GABAAR partially reversed the gut dysbacteriosis of DSS−induced mice and increased the abundance of beneficial bacteria. Additionally, L. casei Zhang supplementation inhibited the expression of GABAAR in mice with colitis, promoted the proliferation and renewal of colon epithelial cells, and alleviated anxiety−like behavior and intestinal microflora disorder in mice. Thus, GABAAR plays a key role in the beneficial effects of L. casei on DSS−induced colitis in mice.
Collapse
|
34
|
Zhang C, Ma K, Nie K, Deng M, Luo W, Wu X, Huang Y, Wang X. Assessment of the safety and probiotic properties of Roseburia intestinalis: A potential “Next Generation Probiotic”. Front Microbiol 2022; 13:973046. [PMID: 36160246 PMCID: PMC9493362 DOI: 10.3389/fmicb.2022.973046] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/05/2022] [Indexed: 12/04/2022] Open
Abstract
Roseburia intestinalis is an anaerobic bacterium that produces butyric acid and belongs to the phylum Firmicutes. There is increasing evidence that this bacterium has positive effects on several diseases, including inflammatory bowel disease, atherosclerosis, alcoholic fatty liver, colorectal cancer, and metabolic syndrome, making it a potential “Next Generation Probiotic.” We investigated the genomic characteristics, probiotic properties, cytotoxicity, oral toxicity, colonization characteristics of the bacterium, and its effect on the gut microbiota. The genome contains few genes encoding virulence factors, three clustered regularly interspaced short palindromic repeat (CRISPR) sequences, two Cas genes, no toxic biogenic amine synthesis genes, and several essential amino acid and vitamin synthesis genes. Seven prophages and 41 genomic islands were predicted. In addition to a bacteriocin (Zoocin A), the bacterium encodes four metabolic gene clusters that synthesize short-chain fatty acids and 222 carbohydrate-active enzyme modules. This bacterium is sensitive to antibiotics specified by the European Food Safety Authority, does not exhibit hemolytic or gelatinase activity, and exhibits some acid resistance. R. intestinalis adheres to intestinal epithelial cells and inhibits the invasion of certain pathogens. In vitro experiments showed that the bacterium was not cytotoxic. R. intestinalis did not affect the diversity or abundance of the gut flora. Using the fluorescent labelling method, we discovered that R. intestinalis colonizes the cecum and mucus of the colon. An oral toxicity study did not reveal any obvious adverse effects. The lethal dose (LD)50 of R. intestinalis exceeded 1.9 × 109 colony forming units (CFU)/kg, whereas the no observed adverse effect level (NOAEL) derived from this study was 1.32 × 109 CFU/kg/day for 28 days. The current research shows that, R. intestinalis is a suitable next-generation probiotic considering its probiotic properties and safety.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kejia Ma
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kai Nie
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minzi Deng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Weiwei Luo
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xing Wu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yujun Huang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
- *Correspondence: Xiaoyan Wang,
| |
Collapse
|
35
|
Zhu Y, Xu Y, Wang X, Rao L, Yan X, Gao R, Shen T, Zhou Y, Kong C, Zhou L. Probiotic Cocktail Alleviates Intestinal Inflammation Through Improving Gut Microbiota and Metabolites in Colitis Mice. Front Cell Infect Microbiol 2022; 12:886061. [PMID: 35782138 PMCID: PMC9240319 DOI: 10.3389/fcimb.2022.886061] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/16/2022] [Indexed: 12/17/2022] Open
Abstract
The modulation of the gut microbiome has been widely suggested as a promising therapeutic strategy for inflammatory bowel disease (IBD). Here, we established a novel probiotic cocktail to investigate its therapeutic role in acute colitis mice. During dextran sulfate sodium (DSS)-induced colitis, the mice were treated with the probiotic cocktail, fecal microbiota transplantation (FMT) from a healthy mice donor, or 5-aminosalicylic acid (5-ASA), respectively. The inflammatory responses were assessed by symptoms, serum inflammatory factors, and histological scoring. The intestinal barrier function was assessed by detecting tight junction proteins. Gut microbiota and its metabolites were further identified using 16S rDNA sequencing and a liquid chromatograph mass spectrometer (LC-MS/MS). Compared with FMT and 5-ASA treatment, the probiotic cocktail performed better in alleviating symptoms of colitis and decreasing disease activity score and mucosal inflammation. The probiotic cocktail also significantly decreased serum IL-17 level and increased JAM-1 expression in colon. The gut microbiota analysis confirmed that the beneficial effects of the probiotic cocktail were attributed to increasing anti-inflammatory bacteria Akkermansia, Bifidobacterium, and Blautia, while decreasing pro-inflammatory bacteria Parasutterella. The targeted metabolome analysis further indicated a rise in the production of Bifidobacterium-related short-chain fatty acids (SCFAs) such as propanoic acid and isobutyric acid after probiotics treatment. Taken together, the probiotic cocktail effectively alleviated intestinal inflammation through improving gut microbiota and metabolites in colitis mice, suggesting its great potential to be a novel therapeutic approach for IBD patients.
Collapse
Affiliation(s)
- Yefei Zhu
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Medicine, Nantong University, Nantong, China
| | - Yu Xu
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- School of Medicine, Nantong University, Nantong, China
| | - Xinyue Wang
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Leiping Rao
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xuebing Yan
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Renyuan Gao
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Tongyi Shen
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Yuan Zhou
- School of Biological Engineering, Hefei Technology College, Hefei, China
| | - Cheng Kong
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- *Correspondence: Longxiang Zhou, ; Cheng Kong,
| | - Longxiang Zhou
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Longxiang Zhou, ; Cheng Kong,
| |
Collapse
|
36
|
Lyu S, Pan F, Ge H, Yang Q, Duan X, Feng M, Liu X, Zhang T, Liu J. Fermented egg-milk beverage alleviates dextran sulfate sodium-induced colitis in mice through the modulation of intestinal flora and short-chain fatty acids. Food Funct 2021; 13:702-715. [PMID: 34935826 DOI: 10.1039/d1fo03040j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Fermented egg-milk beverage (FEMB) can alleviate the symptoms of intestinal diseases by regulating intestinal flora and supplying nutrition. This study investigated the protective effect of FEMB on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. The results showed that FEMB relieved the UC mice's pathological abnormalities and colonic inflammation, and restructured the intestinal flora composition simultaneously. After FEMB treatment for 14 days, the body weight of the mice rose and the disease activity index (DAI) value decreased. Furthermore, the length and form of colons in the UC mice were notably restored. Inflammatory cells decreased or disappeared, and goblet cells and crypt were enriched and modified. 16S rRNA gene sequencing results demonstrated that FEMB treatment could increase the abundance of beneficial bacteria in the cecum content of mice, including unclassified_f_Lachnospiraceae and Lactobacillus. Moreover, probiotics that can increase the content of short-chain fatty acids (SCFAs) may contribute to inflammation alleviation. An increase in amino acids was observed in our experiment, which may benefit nutritional supplements. In conclusion, FEMB treatment can alleviate the damage of DSS-induced colitis in Balb/c mice. This study provides a theoretical basis for both the relief of inflammation and the application of FEMB.
Collapse
Affiliation(s)
- Siwen Lyu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Fengguang Pan
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Huifang Ge
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Qi Yang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Xuehui Duan
- College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Mengmeng Feng
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Xuanting Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Ting Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Jingbo Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China. .,College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| |
Collapse
|