1
|
Liu A, Ma J, Liu Z, Qiu T, Zhao Q, Li G, Liang X, Li Q. "Shield" Armed Programmable Probiotics Harboring α-Aminoadipate Aminotransferase Gene Regulate Tryptophan Metabolism and Gut Microbiota to Alleviate the Inflammatory Bowel Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7920-7932. [PMID: 40116595 DOI: 10.1021/acs.jafc.4c13017] [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: 03/23/2025]
Abstract
Current treatment of inflammatory bowel disease (IBD) relies on anti-inflammatory and immunosuppressive agents. However, this concept is considered outdated due to its restricted efficacy and unavoidable side effects. Herein, a polynorepinephrine-coated programmable probiotic expressing α-aminoadipate aminotransferase (NE-EcN-pA) was constructed to improve the levels of kynurenic acid and xanthurenic acid in the intestine by modulating the endogenous tryptophan metabolism. The NE layer could protect EcN-pA against the harsh environment of the gastrointestinal tract, enhancing its survival and colonization. In UC mice, oral administration of NE-EcN-pA effectively alleviated intestinal inflammation and restored the intestinal epithelial barrier owing to the activation of the aryl hydrocarbon receptor pathway. Furthermore, NE-EcN-pA promoted the diversity of intestinal flora, improved the imbalance of flora, and enhanced the content of short-chain fatty acids in the colon. Overall, NE-EcN-pA can regulate endogenous tryptophan metabolism and gut microbiota, showing promise in the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Aijiang Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jun Ma
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zengguang Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Tianyuan Qiu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qixuan Zhao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Guangquan Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130012, China
| | - Xiao Liang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
2
|
Ma C, Liang Z, Wang Y, Luo H, Yang X, Yao B, Tu T. p-Hydroxycinnamic Acids: Advancements in Synthetic Biology, Emerging Regulatory Targets in Gut Microbiota Interactions, and Implications for Animal Health. J Nutr 2025; 155:1041-1056. [PMID: 39900184 DOI: 10.1016/j.tjnut.2025.01.034] [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: 12/09/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025] Open
Abstract
p-hydroxycinnamic acids (p-HCAs), a class of natural phenolic acid compounds extracted from plant resources and widely distributed, feature a C6-C3 phenylpropanoid structure. Their antioxidant, anti-inflammatory, and antibacterial activities have shown great potential for applications in food and animal feed. The interactions between p-HCAs and the gut microbiota, as well as their subsequent effects on animal health, have increasingly attracted the attention of researchers. In the context of a greener and safer future, the progress and innovation in biosynthetic technology have occupied a central position in ensuring the safety of food and feed. This review emphasizes the complex mechanisms underlying the interactions between p-HCAs and the gut microbiota, providing a solid explanation for the remarkable bioactivities of p-HCAs and their subsequent impact on animal health. Furthermore, it explores the advancements in the synthetic biology of p-HCAs. This review could aid in a basis for better understanding the underlying interactions between p-HCAs and gut microbiota and animal health.
Collapse
Affiliation(s)
- Chunlai Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ziqi Liang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huiying Luo
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Bin Yao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tao Tu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
3
|
Guo J, Shi Y, Yu X, Zhao Y, Wei B, Huo M, Lu L, Li L, Gao Q, Sun M. THRA1/PGC-1α/SIRT3 pathway regulates oxidative stress and is implicated in hypertension of maternal hypothyroid rat offspring. Hypertens Res 2025; 48:1080-1098. [PMID: 39639130 DOI: 10.1038/s41440-024-02023-9] [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: 04/03/2024] [Revised: 10/21/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Many epidemiologic and animal studies have shown that maternal hypothyroidism is associated with an increased risk of hypertension in offspring in later life. In this study, we established a maternal hypothyroidism rat model to explore the underlying mechanism that contributes to elevated blood pressure in adult male offspring of hypothyroid mothers. The levels of thyroid hormones (THs) in the offspring were measured using ELISA kits. Blood pressure (BP) and depressor response were recorded in conscious, freely moving rats. Vascular reactivity was conducted in isolated mesenteric arteries (MAs) using a myograph. We used real-time quantitative PCR (RT-qPCR) and Western blots to examine the mRNA and protein expression of relevant molecules in MAs. The A7r5 cells were transfected with small interfering RNA (siRNA) to further investigate the gene functions. The following findings were observed: Basal systolic BP and diastolic BP was significantly increased, accompanied by attenuated depressor response and decreased vascular sensitivity to sodium nitroprusside (SNP). Reactive Oxygen Species (ROS) levels in the MAs were enhanced, along with decreased expression of the THRA1/PGC-1α/SIRT3 pathway. In A7r5 cells, triiodothyronine (T3) pretreatment improved the PGC-1α/SIRT3 pathway and reduced ROS levels after H2O2-induced oxidative stress. In contrast, the knockdown of THRA1 or SIRT3 diminished the above effects of T3. Down-regulation of THRA1 contributed to a decline in the PGC-1α/SIRT3 pathway, which causes an increased production of ROS. This indicates that the T3-THRA1/PGC-1α/SIRT3 pathway plays a protective role in the regulation of BP and may be a potential therapeutic strategy against hypertension.
Collapse
Affiliation(s)
- Jun Guo
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yajun Shi
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Bin Wei
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Ming Huo
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Likui Lu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Kleniewska P, Pawliczak R. The Link Between Dysbiosis, Inflammation, Oxidative Stress, and Asthma-The Role of Probiotics, Prebiotics, and Antioxidants. Nutrients 2024; 17:16. [PMID: 39796449 PMCID: PMC11722634 DOI: 10.3390/nu17010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Asthma (a chronic inflammatory disease of the airways) is characterized by a variable course, response to treatment, and prognosis. Its incidence has increased significantly in recent decades. Unfortunately, modern lifestyle and environmental factors contribute to the further increase in the incidence of this disease. Progressive industrialization and urbanization, widespread use of antibiotic therapy, excessive sterility and inappropriate, highly processed diets are some of the many risk factors that are relevant today. Over the years, a lot of evidence has been gathered showing the influence of microorganisms of the gut or airways on human health. Studies published in recent years indicate that dysbiosis (microbial imbalance) and oxidative stress (pro-oxidant-antioxidant imbalance) are important elements of the pathogenesis of this inflammatory disease. Scientists have attempted to counteract the effects of this process by using probiotics, prebiotics, and antioxidants. The use of probiotic microorganisms positively modulates the immune system by maintaining homeostasis between individual fractions of immune system cells. Moreover, recently conducted experiments have shown that probiotics have antioxidant, anti-inflammatory, and protective properties in oxidative stress (OS). The aim of this study is to present the current state of knowledge on the role of dysbiosis and OS in the pathogenesis of asthma. Conclusions: This review highlights the importance of using probiotics, prebiotics, and antioxidants as potential strategies to support the treatment and prevention of this disease.
Collapse
Affiliation(s)
- Paulina Kleniewska
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | | |
Collapse
|
5
|
Jiang Z, Mei L, Li Y, Guo Y, Yang B, Huang Z, Li Y. Enzymatic Regulation of the Gut Microbiota: Mechanisms and Implications for Host Health. Biomolecules 2024; 14:1638. [PMID: 39766345 PMCID: PMC11727233 DOI: 10.3390/biom14121638] [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: 10/19/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
The gut microbiota, a complex ecosystem, is vital to host health as it aids digestion, modulates the immune system, influences metabolism, and interacts with the brain-gut axis. Various factors influence the composition of this microbiota. Enzymes, as essential catalysts, actively participate in biochemical reactions that have an impact on the gut microbial community, affecting both the microorganisms and the gut environment. Enzymes play an important role in the regulation of the intestinal microbiota, but the interactions between enzymes and microbial communities, as well as the precise mechanisms of enzymes, remain a challenge in scientific research. Enzymes serve both traditional nutritional functions, such as the breakdown of complex substrates into absorbable small molecules, and non-nutritional roles, which encompass antibacterial function, immunomodulation, intestinal health maintenance, and stress reduction, among others. This study categorizes enzymes according to their source and explores the mechanistic principles by which enzymes drive gut microbial activity, including the promotion of microbial proliferation, the direct elimination of harmful microbes, the modulation of bacterial interaction networks, and the reduction in immune stress. A systematic understanding of enzymes in regulating the gut microbiota and the study of their associated molecular mechanisms will facilitate the application of enzymes to precisely regulate the gut microbiota in the future and suggest new therapeutic strategies and dietary recommendations. In conclusion, this review provides a comprehensive overview of the role of enzymes in modulating the gut microbiota. It explores the underlying molecular and cellular mechanisms and discusses the potential applications of enzyme-mediated microbiota regulation for host gut health.
Collapse
Affiliation(s)
- Zipeng Jiang
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510641, China
| | - Liang Mei
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yuqi Li
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yuguang Guo
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Bo Yang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510641, China
| | - Zhiyi Huang
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yangyuan Li
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| |
Collapse
|
6
|
Wu H, Shi C, Li Q, Wang L, Wang R, Chen F, Li R, Guo X, Chen Y, She J. Oral Administration of Bioactive Nanoparticulates for Inflammatory Bowel Disease Therapy by Mitigating Oxidative Stress and Restoring Intestinal Microbiota Homeostasis. Mol Pharm 2024. [PMID: 39462848 DOI: 10.1021/acs.molpharmaceut.4c00499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The management of inflammatory bowel disease (IBD) continues to pose significant challenges due to the absence of curative therapies and a high rate of recurrence. Therefore, it is imperative to explore novel approaches to enhance the efficacy of IBD therapy. Herein, a bioactive nanoparticulate s is tailored designed to achieve a "Pull-Push" approach for efficient and safe IBD treatment by integrating reactive oxygen species (ROS) scavenging (Pull) with anti-inflammatory agent delivery (Push) in the inflammatory microenvironment. The multifunctional nanomedicine, designated MON-PAMAM@SASP, is developed through the encapsulation of sulfasalazine (SASP), a widely utilized clinical drug for the treatment of IBD, within cationic diselenide-bridged mesoporous organosilica nanoparticles (MONs) that possess significant antioxidant properties. Herein, poly(amidoamine) (PAMAM) endows the original MONs with positive charge characteristics. The MON-PAMAM@SASP not only displays the remarkable capability of neutralizing ROS to ameliorates intestinal damage, but also achieves controllable release of SASP to mitigate intestinal inflammation. Consequently, this nanomedicine effectively mitigates IBD by colitis in mouse models, and our current research has not identified any significant drug toxicity. Beyond regulating inflammatory microenvironment in intestine, treatment with MON-PAMAM@SASP results in increased richness and restores intestinal microbiota homeostasis, thereby mitigating IBD to a certain extent. Together, our work provides a highly versatile "Pull-Push" approach for IBD management and encourages the development of similar nanomedicine to treating multiple inflammatory diseases of gastrointestinal tract.
Collapse
Affiliation(s)
- Hong Wu
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- The Third Department of General Surgery, Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an 710016, China
| | - Chengxin Shi
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qixin Li
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Lizhao Wang
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ruochen Wang
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Fangman Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Ruizhe Li
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiaolong Guo
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Yinnan Chen
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Junjun She
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| |
Collapse
|
7
|
Abanikannda MF, Shiflett MB, Morais ARC, Hong J, Sealey WM, Bledsoe JW. Evaluating Inclusion of Commercial Pistachio By-Product as a Functional Ingredient in Rainbow Trout Fishmeal and Plant Meal-Based Diets. Antioxidants (Basel) 2024; 13:1280. [PMID: 39594422 PMCID: PMC11591393 DOI: 10.3390/antiox13111280] [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: 08/28/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
To meet the growing demand for sustainable aquaculture, plant proteins are being explored as alternative sources in fish diets. However, some plant proteins can have adverse health effects on fish, prompting research into functional feed ingredients to mitigate these issues. This study investigated pistachio shell powder (PSP), rich in antioxidants, as a functional feed ingredient for rainbow trout (Oncorhynchus mykiss). The effects of PSP inclusion (0%, 0.5%, 1%, 2%) on growth performance, intestinal health, and gut microbiota were assessed in fish fed either a fishmeal (FM) or plant meal (PM) diet over a 12-week feeding period. The results indicated that PSP inclusion at 1% significantly (p < 0.05) improved weight gain and growth performance in FM treatments, with no impact on growth in PM treatments. No significant differences were observed in other growth parameters, intestinal morphology, or oxidative stress markers, although a trend toward the downregulation of inflammatory genes was noted in PM treatments at 2% PSP inclusion. PSP inclusion did not significantly alter gut microbiota alpha diversity but affected beta diversity at the 0.5% level in the FM treatments (p < 0.05). Differential abundance analysis of gut microbiota revealed taxa-specific responses to PSP, particularly the genus Candidatus arthromitus, increasing in relative abundance with PSP inclusion in both the FM- and PM-based treatments. Overall, PSP inclusion up to 2% did not have significant adverse effects on the growth, intestinal health, or antioxidant status of rainbow trout.
Collapse
Affiliation(s)
- Mosope F. Abanikannda
- Department of Animal Veterinary & Food Sciences, University of Idaho, Moscow, ID 83844, USA;
- Aquaculture Research Institute, Hagerman Fish Culture Experiment Station, University of Idaho, Hagerman, ID 83332, USA;
| | - Mark B. Shiflett
- Wonderful Institute for Sustainable Engineering, University of Kansas, Lawrence, KS 66045, USA; (M.B.S.); (A.R.C.M.)
| | - Ana Rita C. Morais
- Wonderful Institute for Sustainable Engineering, University of Kansas, Lawrence, KS 66045, USA; (M.B.S.); (A.R.C.M.)
| | - Jeoungwhui Hong
- Aquaculture Research Institute, Hagerman Fish Culture Experiment Station, University of Idaho, Hagerman, ID 83332, USA;
| | - Wendy M. Sealey
- US Department of Agriculture (USDA) Agriculture Research Service (ARS), Bozeman, MT 59715, USA;
| | - Jacob W. Bledsoe
- Department of Animal Veterinary & Food Sciences, University of Idaho, Moscow, ID 83844, USA;
- Aquaculture Research Institute, Hagerman Fish Culture Experiment Station, University of Idaho, Hagerman, ID 83332, USA;
| |
Collapse
|
8
|
Yao H, Chen J, Wang Y, Li Y, Jiang Q. Assessing causal relationships between gut microbiota and abortion: evidence from two sample Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1415730. [PMID: 39050566 PMCID: PMC11266152 DOI: 10.3389/fendo.2024.1415730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Background While some studies have suggested a link between gut microbiota (GM) and abortion, the causal relationship remains unclear. Methods To explore the causal relationship between GM and abortion, including spontaneous abortion (SA) and habitual abortion (HA), we performed a two-sample Mendelian randomization (MR) analysis. We used summary statistics data from MiBioGen and FinnGen for genome-wide association studies (GWAS), with GM data as the exposure variable and abortion data as the outcome variable. Results In the absence of heterogeneity and horizontal pleiotropy, the inverse-variance weighted (IVW) method identified five genetically predicted GM genera linked to the risk of abortions. Lactococcus was negatively correlated with the risk of SA, whereas the Eubacterium fissicatena group was positively correlated with the risk of SA. Genetic predictions of Coprococcus3 and Odoribacter were linked to a reduced risk of HA, while the Eubacterium ruminantium group was associated with an increased risk of HA. Conclusion Our study suggests a genetic causal relationship between specific GM and two types of abortions, improving our understanding of the pathological relationship between GM and abortion.
Collapse
Affiliation(s)
- Hang Yao
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Jiahao Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Wang
- Graduate School of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yuxin Li
- Graduate School of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qingling Jiang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
9
|
Gryaznova M, Burakova I, Smirnova Y, Morozova P, Chirkin E, Gureev A, Mikhaylov E, Korneeva O, Syromyatnikov M. Effect of Probiotic Bacteria on the Gut Microbiome of Mice with Lipopolysaccharide-Induced Inflammation. Microorganisms 2024; 12:1341. [PMID: 39065109 PMCID: PMC11278525 DOI: 10.3390/microorganisms12071341] [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: 05/21/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The role of lipopolysaccharide (LPS) in the development of diseases is clear, but the specific mechanisms remain poorly understood. This study aimed to investigate the microbiome aberrations in the guts of mice against the background of LPS, as well as the anti-inflammatory effect of probiotic supplementation with Lactobacillus plantarum from the gut, a mix of commercial probiotic lactic acid bacteria, and Weissella confusa isolated from milk using next-generation sequencing. LPS injections were found to induce inflammatory changes in the intestinal mucosa. These morphological changes were accompanied by a shift in the microbiota. We found no significant changes in the microbiome with probiotic supplementation compared to the LPS group. However, when Lactobacillus plantarum and a mix of commercial probiotic lactic acid bacteria were used, the intestinal mucosa was restored. Weissella confusa did not contribute to the morphological changes of the intestinal wall or the microbiome. Changes in the microbiome were observed with probiotic supplementation of Lactobacillus plantarum and a mix of commercial probiotic lactic acid bacteria compared to the control group. In addition, when Lactobacillus plantarum was used, we observed a decrease in the enrichment of the homocysteine and cysteine interconversion pathways with an increase in the L-histidine degradation pathway.
Collapse
Affiliation(s)
- Mariya Gryaznova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia; (M.G.); (I.B.); (Y.S.); (P.M.); (O.K.)
| | - Inna Burakova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia; (M.G.); (I.B.); (Y.S.); (P.M.); (O.K.)
| | - Yuliya Smirnova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia; (M.G.); (I.B.); (Y.S.); (P.M.); (O.K.)
| | - Polina Morozova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia; (M.G.); (I.B.); (Y.S.); (P.M.); (O.K.)
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (E.C.); (A.G.)
| | - Egor Chirkin
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (E.C.); (A.G.)
| | - Artem Gureev
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (E.C.); (A.G.)
| | - Evgeny Mikhaylov
- FSBSI All-Russian Veterinary Research Institute of Pathology, Pharmacology and Therapy, 394061 Voronezh, Russia;
| | - Olga Korneeva
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia; (M.G.); (I.B.); (Y.S.); (P.M.); (O.K.)
| | - Mikhail Syromyatnikov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia; (M.G.); (I.B.); (Y.S.); (P.M.); (O.K.)
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (E.C.); (A.G.)
| |
Collapse
|
10
|
Hu C, Yuan X, Zhao R, Hong B, Chen C, Zhu Q, Zheng Y, Hu J, Yuan Y, Wu Z, Zhang J, Tang C. Scale-Up Preparation of Manganese-Iron Prussian Blue Nanozymes as Potent Oral Nanomedicines for Acute Ulcerative Colitis. Adv Healthc Mater 2024; 13:e2400083. [PMID: 38447228 DOI: 10.1002/adhm.202400083] [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: 01/08/2024] [Revised: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Prussian blue (PB) nanozymes are demonstrated as effective therapeutics for ulcerative colitis (UC), yet an unmet practical challenge remains in the scalable production of these nanozymes and uncertainty over their efficacy. With a novel approach, a series of porous manganese-iron PB (MnPB) colloids, which are shown to be efficient scavengers for reactive oxygen species (ROS) including hydroxyl radical, superoxide anion, and hydrogen peroxide, are prepared. In vitro cellular experiments confirm the capability of the nanozyme to protect cells from ROS attack. In vivo, the administration of MnPB nanozyme through gavage at a dosage of 10 mg kg-1 per day for three doses in total potently ameliorates the pathological symptoms of acute UC in a murine model, resulting in mitigated inflammatory responses and improved viability rate. Significantly, the nanozyme produced at a large scale can be achieved at an unprecedented yield weighting ≈11 g per batch of reaction, demonstrating comparable anti-ROS activities and treatment efficacy to its small-scale counterpart. This work represents the first demonstration of the scale-up preparation of PB analog nanozymes for UC without compromising treatment efficacy, laying the foundation for further testing of these nanozymes on larger animals and promising clinical translation.
Collapse
Affiliation(s)
- Chengyun Hu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xue Yuan
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Ronghua Zhao
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Biao Hong
- College & Hospital of Stomatology, Anhui Provincial Key Laboratory of Oral Diseases Research, Anhui Medical University, Hefei, 230032, China
| | - Chuang Chen
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Qingjun Zhu
- Anhui Provincial Key Laboratory of High Magnetic Resonance Image, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yanmin Zheng
- Anhui Provincial Key Laboratory of High Magnetic Resonance Image, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jinming Hu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Yue Yuan
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Zhengyan Wu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Jia Zhang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Chaoliang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
11
|
Yang F, Su Y, Yan C, Chen T, Cheung PCK. Attenuation of inflammatory bowel disease by oral administration of mucoadhesive polydopamine-coated yeast β-glucan via ROS scavenging and gut microbiota regulation. J Nanobiotechnology 2024; 22:166. [PMID: 38610032 PMCID: PMC11010398 DOI: 10.1186/s12951-024-02434-3] [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: 09/25/2023] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Treatment for inflammatory bowel disease (IBD) is challenging since current anti-inflammatory and immunosuppressive therapies do not address the underlying causes of the illness, which include increased levels of reactive oxygen species (ROS) and dysbiosis of the gut commensal microbiota. Additionally, these treatments often have systemic off-target effects and adverse side effects. In this study, we have developed a prebiotic yeast β-glucan nanocomplex coated with bio-adhesive polydopamine (YBNs@PDA) to effectively prolong their retention time in the gastrointestinal (GI) tract. The oral administration of YBNs@PDA restored the epithelium barriers, reduced ROS levels, and minimized systemic drug exposure while improved therapeutic efficacy in an acute colitis mouse model. Furthermore, 16S ribosomal RNA genes sequencing demonstrated a higher richness and diversity in gut microflora composition following the treatments. In particular, YBNs@PDA markedly augmented the abundance of Lachnospiraceae NK4A136 and Bifidobacterium, both of which are probiotics with crucial roles in relieving colitis via retaining gut homeostasis. Cumulatively, these results demonstrate that the potential of YBNs@PDA as a novel drug-free, ROS-scavenging and gut microbiota regulation nanoplatform for the treatment of GI disorders.
Collapse
Affiliation(s)
- Fan Yang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yuting Su
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Tianfeng Chen
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China
| | - Peter Chi Keung Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
12
|
Zhang C, Xiang C, Zhou K, Liu X, Qiao G, Zhao Y, Dong K, Sun K, Liu Z. Intestinal lysozyme1 deficiency alters microbiota composition and impacts host metabolism through the emergence of NAD +-secreting ASTB Qing110 bacteria. mSystems 2024; 9:e0121423. [PMID: 38364095 PMCID: PMC10949482 DOI: 10.1128/msystems.01214-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/26/2024] [Indexed: 02/18/2024] Open
Abstract
The intestine plays a pivotal role in nutrient absorption and host defense against pathogens, orchestrated in part by antimicrobial peptides secreted by Paneth cells. Among these peptides, lysozyme has multifaceted functions beyond its bactericidal activity. Here, we uncover the intricate relationship between intestinal lysozyme, the gut microbiota, and host metabolism. Lysozyme deficiency in mice led to altered body weight, energy expenditure, and substrate utilization, particularly on a high-fat diet. Interestingly, these metabolic benefits were linked to changes in the gut microbiota composition. Cohousing experiments revealed that the metabolic effects of lysozyme deficiency were microbiota-dependent. 16S rDNA sequencing highlighted differences in microbial communities, with ASTB_g (OTU60) highly enriched in lysozyme knockout mice. Subsequently, a novel bacterium, ASTB Qing110, corresponding to ASTB_g (OTU60), was isolated. Metabolomic analysis revealed that ASTB Qing110 secreted high levels of NAD+, potentially influencing host metabolism. This study sheds light on the complex interplay between intestinal lysozyme, the gut microbiota, and host metabolism, uncovering the potential role of ASTB Qing110 as a key player in modulating metabolic outcomes. IMPORTANCE The impact of intestinal lumen lysozyme on intestinal health is complex, arising from its multifaceted interactions with the gut microbiota. Lysozyme can both mitigate and worsen certain health conditions, varying with different scenarios. This underscores the necessity of identifying the specific bacterial responses elicited by lysozyme and understanding their molecular foundations. Our research reveals that a deficiency in intestinal lysozyme1 may offer protection against diet-induced obesity by altering bacterial populations. We discovered a strain of bacterium, ASTB Qing110, which secretes NAD+ and is predominantly found in lyz1-deficient mice. Qing110 demonstrates positive effects in both C. elegans and mouse models of ataxia telangiectasia. This study sheds light on the intricate role of lysozyme in influencing intestinal health.
Collapse
Affiliation(s)
- Chengye Zhang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Chen Xiang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Kaichen Zhou
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xingchen Liu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Guofeng Qiao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Yabo Zhao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Kemeng Dong
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ke Sun
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Zhihua Liu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
13
|
Jastrząb R, Tomecki R, Jurkiewicz A, Graczyk D, Szczepankowska AK, Mytych J, Wolman D, Siedlecki P. The strain-dependent cytostatic activity of Lactococcus lactis on CRC cell lines is mediated through the release of arginine deiminase. Microb Cell Fact 2024; 23:82. [PMID: 38481270 PMCID: PMC10938756 DOI: 10.1186/s12934-024-02345-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most commonly diagnosed cancers, posing a serious public health challenge that necessitates the development of new therapeutics, therapies, and prevention methods. Among the various therapeutic approaches, interventions involving lactic acid bacteria (LAB) as probiotics and postbiotics have emerged as promising candidates for treating and preventing CRC. While human-isolated LAB strains are considered highly favorable, those sourced from environmental reservoirs such as dairy and fermented foods are also being recognized as potential sources for future therapeutics. RESULTS In this study, we present a novel and therapeutically promising strain, Lactococcus lactis ssp. lactis Lc4, isolated from dairy sources. Lc4 demonstrated the ability to release the cytostatic agent - arginine deiminase (ADI) - into the post-cultivation supernatant when cultured under conditions mimicking the human gut environment. Released arginine deiminase was able to significantly reduce the growth of HT-29 and HCT116 cells due to the depletion of arginine, which led to decreased levels of c-Myc, reduced phosphorylation of p70-S6 kinase, and cell cycle arrest. The ADI release and cytostatic properties were strain-dependent, as was evident from comparison to other L. lactis ssp. lactis strains. CONCLUSION For the first time, we unveil the anti-proliferative properties of the L. lactis cell-free supernatant (CFS), which are independent of bacteriocins or other small molecules. We demonstrate that ADI, derived from a dairy-Generally Recognized As Safe (GRAS) strain of L. lactis, exhibits anti-proliferative activity on cell lines with different levels of argininosuccinate synthetase 1 (ASS1) expression. A unique feature of the Lc4 strain is also its capability to release ADI into the extracellular space. Taken together, we showcase L. lactis ADI and the Lc4 strain as promising, potential therapeutic agents with broad applicability.
Collapse
Affiliation(s)
- Rafał Jastrząb
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Adolfa Pawińskiego 5A, Warsaw, 02-106, Poland
- Olimp Laboratories, Pustynia 84F, Debica, 39-200, Poland
| | - Rafał Tomecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Adolfa Pawińskiego 5A, Warsaw, 02-106, Poland
- Faculty of Biology, University of Warsaw, Miecznikowa 1, Warsaw, 02-089, Poland
| | - Aneta Jurkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Adolfa Pawińskiego 5A, Warsaw, 02-106, Poland
| | - Damian Graczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Adolfa Pawińskiego 5A, Warsaw, 02-106, Poland
| | - Agnieszka K Szczepankowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Adolfa Pawińskiego 5A, Warsaw, 02-106, Poland
| | | | - Damian Wolman
- Olimp Laboratories, Pustynia 84F, Debica, 39-200, Poland
| | - Pawel Siedlecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Adolfa Pawińskiego 5A, Warsaw, 02-106, Poland.
| |
Collapse
|
14
|
Lau LYJ, Quek SY. Probiotics: Health benefits, food application, and colonization in the human gastrointestinal tract. FOOD BIOENGINEERING 2024; 3:41-64. [DOI: 10.1002/fbe2.12078] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/29/2024] [Indexed: 01/04/2025]
Abstract
AbstractProbiotics have become increasingly popular over the past two decades due to the continuously expanding scientific evidence indicating their beneficial effects on human health. Therefore, they have been applied in the food industry to produce functional food, which plays a significant role in human health and reduces disease risk. However, maintaining the viability of probiotics and targeting the successful delivery to the gastrointestinal tract remain two challenging tasks in food applications. Specifically, this paper reviews the potentially beneficial properties of probiotics, highlighting the use and challenges of probiotics in food application and the associated health benefits. Of foremost importance, this paper also explores the potential underlying molecular mechanisms of the enhanced effect of probiotics on gastrointestinal epithelial cells, including a discussion on various surface adhesion‐related proteins on the probiotic cell surface that facilitate colonization.
Collapse
Affiliation(s)
- Li Ying Jessie Lau
- Food Science, School of Chemical Sciences The University of Auckland Auckland New Zealand
| | - Siew Young Quek
- Food Science, School of Chemical Sciences The University of Auckland Auckland New Zealand
| |
Collapse
|
15
|
Rauf A, Khalil AA, Awadallah S, Khan SA, Abu‐Izneid T, Kamran M, Hemeg HA, Mubarak MS, Khalid A, Wilairatana P. Reactive oxygen species in biological systems: Pathways, associated diseases, and potential inhibitors-A review. Food Sci Nutr 2024; 12:675-693. [PMID: 38370049 PMCID: PMC10867483 DOI: 10.1002/fsn3.3784] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 02/20/2024] Open
Abstract
Reactive oxygen species (ROS) are produced under normal physiological conditions and may have beneficial and harmful effects on biological systems. ROS are involved in many physiological processes such as differentiation, proliferation, necrosis, autophagy, and apoptosis by acting as signaling molecules or regulators of transcription factors. In this case, maintaining proper cellular ROS levels is known as redox homeostasis. Oxidative stress occurs because of the imbalance between the production of ROS and antioxidant defenses. Sources of ROS include the mitochondria, auto-oxidation of glucose, and enzymatic pathways such as nicotinamide adenine dinucleotide phosphate reduced (NAD[P]H) oxidase. The possible ROS pathways are NF-κB, MAPKs, PI3K-Akt, and the Keap1-Nrf2-ARE signaling pathway. This review covers the literature pertaining to the possible ROS pathways and strategies to inhibit them. Additionally, this review summarizes the literature related to finding ROS inhibitors.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of ChemistryUniversity of SwabiAnbarPakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Samir Awadallah
- Department of Medical Lab Sciences, Faculty of Allied Medical SciencesZarqa UniversityZarqaJordan
| | - Shahid Ali Khan
- Department of Chemistry, School of Natural SciencesNational University of Science and Technology (NUST)IslamabadPakistan
| | - Tareq Abu‐Izneid
- Pharmaceutical Sciences, College of PharmacyAl Ain UniversityAl Ain, Abu DhabiUAE
| | - Muhammad Kamran
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical SciencesTaibah UniversityAl‐Medinah Al‐MonawaraSaudi Arabia
| | | | - Ahood Khalid
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| |
Collapse
|
16
|
Abdelgawad A, Nicola T, Martin I, Halloran BA, Tanaka K, Adegboye CY, Jain P, Ren C, Lal CV, Ambalavanan N, O'Connell AE, Jilling T, Willis KA. Antimicrobial peptides modulate lung injury by altering the intestinal microbiota. MICROBIOME 2023; 11:226. [PMID: 37845716 PMCID: PMC10578018 DOI: 10.1186/s40168-023-01673-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/21/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Mammalian mucosal barriers secrete antimicrobial peptides (AMPs) as critical, host-derived regulators of the microbiota. However, mechanisms that support microbiota homeostasis in response to inflammatory stimuli, such as supraphysiologic oxygen, remain unclear. RESULTS We show that supraphysiologic oxygen exposure to neonatal mice, or direct exposure of intestinal organoids to supraphysiologic oxygen, suppresses the intestinal expression of AMPs and alters intestinal microbiota composition. Oral supplementation of the prototypical AMP lysozyme to hyperoxia-exposed neonatal mice reduced hyperoxia-induced alterations in their microbiota and was associated with decreased lung injury. CONCLUSIONS Our results identify a gut-lung axis driven by intestinal AMP expression and mediated by the intestinal microbiota that is linked to lung injury in newborns. Together, these data support that intestinal AMPs modulate lung injury and repair. Video Abstract.
Collapse
Affiliation(s)
- Ahmed Abdelgawad
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Teodora Nicola
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Isaac Martin
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brian A Halloran
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kosuke Tanaka
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Comfort Y Adegboye
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pankaj Jain
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Changchun Ren
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charitharth V Lal
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amy E O'Connell
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tamás Jilling
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kent A Willis
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
17
|
Li B, Guo Y, Jia X, Cai Y, Zhang Y, Yang Q. Luteolin alleviates ulcerative colitis in rats via regulating immune response, oxidative stress, and metabolic profiling. Open Med (Wars) 2023; 18:20230785. [PMID: 37693835 PMCID: PMC10487402 DOI: 10.1515/med-2023-0785] [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: 01/17/2023] [Revised: 05/30/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease and associated with metabolic imbalance. Luteolin (LUT) reportedly exhibits anti-inflammatory activity. However, its regulatory effects on metabolites remain indistinct. Here, the effects of LUT on immune response and oxidative stress in UC were determined. Serum metabolomics profiles of UC rats treated with LUT were obtained utilizing liquid chromatography-mass spectrometry. The results revealed that LUT treatment alleviated colon tissue injury, colon shortening, weight loss, and inflammatory response in UC rats. Additionally, the levels of superoxide dismutase and total antioxidant capacity were elevated, but malondialdehyde content was reduced in serum of UC rats, while these changes were abrogated by LUT. Metabolomics analysis unveiled that l-malic acid, creatinine, l-glutamine, and l-lactic acid levels were remarkably decreased, while dimethyl sulfone, 5-methylcytosine, cysteine-S-sulfate, and jasmonic acid levels were notably increased after LUT treatment. Furthermore, differential metabolites primarily participated in d-glutamine and d-glutamate metabolism, glutathione metabolism, and citrate cycle pathways. In summary, these results demonstrated that LUT improved immune response, alleviated oxidative stress, and altered metabolites in UC rats. This study lays the root for further exploring the mechanism of LUT in the treatment of UC.
Collapse
Affiliation(s)
- Bolin Li
- Department of Gastroenterology, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
- Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research (Hebei), Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Turbidity Toxin Syndrome, Shijiazhuang, Hebei, China
| | - Yuxi Guo
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xuemei Jia
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yanru Cai
- Department of Gastroenterology, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yunfeng Zhang
- Hebei Key Laboratory of Turbidity Toxin Syndrome, Shijiazhuang, Hebei, China
- Department of Gastroenterology, Hebei Provincial Hospital of Chinese Medicine, 389 Zhongshan East Road, Chang’an District, Shijiazhuang, Hebei, China
| | - Qian Yang
- Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research (Hebei), Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Turbidity Toxin Syndrome, Shijiazhuang, Hebei, China
- Department of Gastroenterology, Hebei Provincial Hospital of Chinese Medicine, 389 Zhongshan East Road, Chang’an District, Shijiazhuang, Hebei, China
| |
Collapse
|
18
|
Li Q, Zheng T, Ding H, Chen J, Li B, Zhang Q, Yang S, Zhang S, Guan W. Exploring the Benefits of Probiotics in Gut Inflammation and Diarrhea-From an Antioxidant Perspective. Antioxidants (Basel) 2023; 12:1342. [PMID: 37507882 PMCID: PMC10376667 DOI: 10.3390/antiox12071342] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammatory bowel disease (IBD), characterized by an abnormal immune response, includes two distinct types: Crohn's disease (CD) and ulcerative colitis (UC). Extensive research has revealed that the pathogeny of IBD encompasses genetic factors, environmental factors, immune dysfunction, dysbiosis, and lifestyle choices. Furthermore, patients with IBD exhibit both local and systemic oxidative damage caused by the excessive presence of reactive oxygen species. This oxidative damage exacerbates immune response imbalances, intestinal mucosal damage, and dysbiosis in IBD patients. Meanwhile, the weaning period represents a crucial phase for pigs, during which they experience pronounced intestinal immune and inflammatory responses, leading to severe diarrhea and increased mortality rates. Pigs are highly similar to humans in terms of physiology and anatomy, making them a potential choice for simulating human IBD. Although the exact mechanism behind IBD and post-weaning diarrhea remains unclear, the oxidative damage, in its progression and pathogenesis, is well acknowledged. Besides conventional anti-inflammatory drugs, certain probiotics, particularly Lactobacillus and Bifidobacteria strains, have been found to possess antioxidant properties. These include the scavenging of reactive oxygen species, chelating metal ions to inhibit the Fenton reaction, and the regulation of host antioxidant enzymes. Consequently, numerous studies in the last two decades have committed to exploring the role of probiotics in alleviating IBD. Here, we sequentially discuss the oxidative damage in IBD and post-weaning diarrhea pathogenesis, the negative consequences of oxidative stress on IBD, the effectiveness of probiotics in IBD treatment, the application of probiotics in weaned piglets, and the potential antioxidant mechanisms of probiotics.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hanting Ding
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
19
|
Cao F, Jin L, Gao Y, Ding Y, Wen H, Qian Z, Zhang C, Hong L, Yang H, Zhang J, Tong Z, Wang W, Chen X, Mao Z. Artificial-enzymes-armed Bifidobacterium longum probiotics for alleviating intestinal inflammation and microbiota dysbiosis. NATURE NANOTECHNOLOGY 2023; 18:617-627. [PMID: 36973397 DOI: 10.1038/s41565-023-01346-x] [Citation(s) in RCA: 172] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Inflammatory bowel disease can be caused by the dysfunction of the intestinal mucosal barrier and dysregulation of gut microbiota. Traditional treatments use drugs to manage inflammation with possible probiotic therapy as an adjuvant. However, current standard practices often suffer from metabolic instability, limited targeting and result in unsatisfactory therapeutic outcomes. Here we report on artificial-enzyme-modified Bifidobacterium longum probiotics for reshaping a healthy immune system in inflammatory bowel disease. Probiotics can promote the targeting and retention of the biocompatible artificial enzymes to persistently scavenge elevated reactive oxygen species and alleviate inflammatory factors. The reduced inflammation caused by artificial enzymes improves bacterial viability to rapidly reshape the intestinal barrier functions and restore the gut microbiota. The therapeutic effects are demonstrated in murine and canine models and show superior outcomes to traditional clinical drugs.
Collapse
Affiliation(s)
- Fangfang Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongyang Wen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chenyin Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jiaojiao Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zongrui Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Huang Q, Yang Y, Zhu Y, Chen Q, Zhao T, Xiao Z, Wang M, Song X, Jiang Y, Yang Y, Zhang J, Xiao Y, Nan Y, Wu W, Ai K. Oral Metal-Free Melanin Nanozymes for Natural and Durable Targeted Treatment of Inflammatory Bowel Disease (IBD). SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207350. [PMID: 36760016 DOI: 10.1002/smll.202207350] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/10/2023] [Indexed: 05/11/2023]
Abstract
Oral antioxidant nanozymes bring great promise for inflammatory bowel disease (IBD) treatment. To efficiently eliminate reactive oxygen species (ROS), various metal-based nanozymes have been developed for the treatment of IBD but their practical applications are seriously impaired by unstable ROS-eliminating properties and potential metal ion leakage in the digestive tract. Here, the authors for the first time propose metal-free melanin nanozymes (MeNPs) with excellent gastrointestinal stability and biocompatibility as a favorable therapy strategy for IBD. Moreover, MeNPs have extremely excellent natural and long-lasting characteristics of targeting IBD lesions. In view of the dominant role of ROS in IBD, the authors further reveal that oral administration of MeNPs can greatly alleviate the six major pathological features of IBD: oxidative stress, endoplasmic reticulum stress, apoptosis, inflammation, gut barrier disruption, and gut dysbiosis. Overall, this strategy highlights the great clinical application prospects of metal-free MeNPs via harnessing ROS scavenging at IBD lesions, offering a paradigm for antioxidant nanozyme in IBD or other inflammatory diseases.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yuqi Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yan Zhu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Mingyuan Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiangping Song
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yitian Jiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yunrong Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jinping Zhang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Wei Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
21
|
Tian H, Li J, Chen X, Ren Z, Pan X, Huang W, Bhatia M, Pan LL, Sun J. Oral Delivery of Mouse β-Defensin 14 (mBD14)-Producing Lactococcus lactis NZ9000 Attenuates Experimental Colitis in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:5185-5194. [PMID: 36943701 DOI: 10.1021/acs.jafc.2c07098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Antimicrobial peptides (AMPs) play essential roles in maintaining intestinal health and have been suggested as possible therapeutic strategies against inflammatory bowel disease (IBD). However, the instability of AMPs in the process of transmission in vivo limits their application in the treatment of IBD. In this study, we constructed the mBD14-producing Lactococcus lactis NZ9000 (L. lactis/mBD14) to achieve enteric delivery of mBD14 and evaluated its protective effect on dextran sodium sulfate (DSS)-induced colitis. Mice treated with L. lactis/mBD14 exhibited milder symptoms of colitis (P < 0.01). Additionally, L. lactis/mBD14 treatment reversed DSS-induced epithelial dysfunction and reduced the production of pro-inflammatory cytokines in colon (P < 0.01). Mechanistically, L. lactis/mBD14 significantly inhibited NOD-like receptor pyrin domain containing three inflammasome-mediated pro-inflammatory response (P < 0.05) and regulated microbiota homeostasis by promoting the abundance of probiotic bacteria Akkermansia muciniphila and Faecalibacterium prausnitzii and decreasing the pathogenic Escherichia coli (P < 0.01). Taken together, this study demonstrates the protective effect of L. lactis/mBD14 in DSS-induced colitis, and suggests that oral administration of L. lactis/mBD14 may represent a potential therapeutic strategy for IBD.
Collapse
Affiliation(s)
- Haizhi Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Xiaopei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Zhengnan Ren
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Weining Huang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 9016, New Zealand
| | - Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi 214126, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| |
Collapse
|
22
|
Abdelgawad A, Nicola T, Martin I, Halloran BA, Tanaka K, Adegboye CY, Jain P, Ren C, Lal CV, Ambalavanan N, O'Connell AE, Jilling T, Willis KA. Antimicrobial peptides modulate lung injury by altering the intestinal microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.529700. [PMID: 36993189 PMCID: PMC10054967 DOI: 10.1101/2023.03.14.529700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Mammalian mucosal barriers secrete antimicrobial peptides (AMPs) as critical host-derived regulators of the microbiota. However, mechanisms that support homeostasis of the microbiota in response to inflammatory stimuli such as supraphysiologic oxygen remain unclear. Here, we show that neonatal mice breathing supraphysiologic oxygen or direct exposure of intestinal organoids to supraphysiologic oxygen suppress the intestinal expression of AMPs and alters the composition of the intestinal microbiota. Oral supplementation of the prototypical AMP lysozyme to hyperoxia exposed neonatal mice reduced hyperoxia-induced alterations in their microbiota and was associated with decreased lung injury. Our results identify a gut-lung axis driven by intestinal AMP expression and mediated by the intestinal microbiota that is linked to lung injury. Together, these data support that intestinal AMPs modulate lung injury and repair. In Brief Using a combination of murine models and organoids, Abdelgawad and Nicola et al. find that suppression of antimicrobial peptide release by the neonatal intestine in response to supra-physiological oxygen influences the progression of lung injury likely via modulation of the ileal microbiota. Highlights Supraphysiologic oxygen exposure alters intestinal antimicrobial peptides (AMPs).Intestinal AMP expression has an inverse relationship with the severity of lung injury.AMP-driven alterations in the intestinal microbiota form a gut-lung axis that modulates lung injury.AMPs may mediate a gut-lung axis that modulates lung injury.
Collapse
|
23
|
Walsh AM, Leech J, Huttenhower C, Delhomme-Nguyen H, Crispie F, Chervaux C, Cotter P. Integrated molecular approaches for fermented food microbiome research. FEMS Microbiol Rev 2023; 47:fuad001. [PMID: 36725208 PMCID: PMC10002906 DOI: 10.1093/femsre/fuad001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Molecular technologies, including high-throughput sequencing, have expanded our perception of the microbial world. Unprecedented insights into the composition and function of microbial communities have generated large interest, with numerous landmark studies published in recent years relating the important roles of microbiomes and the environment-especially diet and nutrition-in human, animal, and global health. As such, food microbiomes represent an important cross-over between the environment and host. This is especially true of fermented food microbiomes, which actively introduce microbial metabolites and, to a lesser extent, live microbes into the human gut. Here, we discuss the history of fermented foods, and examine how molecular approaches have advanced research of these fermented foods over the past decade. We highlight how various molecular approaches have helped us to understand the ways in which microbes shape the qualities of these products, and we summarize the impacts of consuming fermented foods on the gut. Finally, we explore how advances in bioinformatics could be leveraged to enhance our understanding of fermented foods. This review highlights how integrated molecular approaches are changing our understanding of the microbial communities associated with food fermentation, the creation of unique food products, and their influences on the human microbiome and health.
Collapse
Affiliation(s)
- Aaron M Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork and APC Microbiome Ireland, P61 C996, Ireland
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - John Leech
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork and APC Microbiome Ireland, P61 C996, Ireland
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Fiona Crispie
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork and APC Microbiome Ireland, P61 C996, Ireland
| | - Christian Chervaux
- Danone Nutricia Research, Centre Daniel Carasso, Palaiseau 91120, France
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork and APC Microbiome Ireland, P61 C996, Ireland
| |
Collapse
|
24
|
Jiang Q, Charoensiddhi S, Xue X, Sun B, Liu Y, El-Seedi HR, Wang K. A review on the gastrointestinal protective effects of tropical fruit polyphenols. Crit Rev Food Sci Nutr 2022; 63:7197-7223. [PMID: 36397724 DOI: 10.1080/10408398.2022.2145456] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tropical fruits are popular because of their unique, delicious flavors and good nutritional value. Polyphenols are considered to be the main bioactive ingredients in tropical fruits, and these exert a series of beneficial effects on the human gastrointestinal tract that can enhance intestinal health and prevent intestinal diseases. Moreover, they are distinct from the polyphenols in fruits grown in other geographical zones. Thus, the comprehensive effects of polyphenols in tropical fruits on gut health warrant in-depth review. This article reviews, first, the biological characteristics of several representative tropical fruits, including mango, avocado, noni, cashew apple, passion fruit and lychee; second, the types and content of the main polyphenols in these tropical fruits; third, the effects of each of these fruit polyphenols on gastrointestinal health; and, fourth, the protective mechanism of polyphenols. Polyphenols and their metabolites play a crucial role in the regulation of the gut microbiota, increasing intestinal barrier function, reducing oxidative stress, inhibiting the secretion of inflammatory factors and regulating immune function. Thus, review highlights the value of tropical fruits, highlighting their significance for future research on their applications as functional foods that are oriented to gastrointestinal protection.
Collapse
Affiliation(s)
- Qianer Jiang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Suvimol Charoensiddhi
- Department of Food Science and Technology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Xiaofeng Xue
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Biqi Sun
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yang Liu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Hesham R El-Seedi
- Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, Uppsala, Sweden
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
| | - Kai Wang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
25
|
Koga J, Ojiro K, Yanagida A, Suto T, Hiki H, Inoue Y, Sakai C, Nakamoto K, Fujisawa Y, Orihara A, Murakami H, Hirasawa S, Nakajima K, Sakazawa T, Yamane H. Ingestion of Indigestible Cacao Proteins Promotes Defecation and Alters the Intestinal Microbiota in Mice. Curr Dev Nutr 2022; 6:nzac129. [PMID: 36204331 PMCID: PMC9529219 DOI: 10.1093/cdn/nzac129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/26/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background In animals, the health effects of ingested cacao proteins are unknown because the proteins are difficult to extract and purify from cacao beans. Objectives This study aimed to develop an extraction and purification method for cacao proteins and reveal the effect of ingestion of cacao proteins on defecation and intestinal microbiota in mice. Methods Three groups of mice were fed a control diet (AIN-93 G), a cacao lignin diet (AIN-93 G containing 1.22% cacao lignin), or a cacao protein and lignin diet (AIN-93 G containing 1.97% cacao proteins and 1.22% cacao lignin) by pair-feeding for 8 d. Feces were collected as 2 bulked samples from days 1 to 4 and days 5 to 8 on each diet. The collected feces were weighed and the intestinal microbiota was analyzed by next-generation sequencing-based 16S rRNA. Results A new extraction and purification method for cacao proteins has been developed, then found that the proteins are resistant to digestive enzymes. However, the cacao protein powder made by this method contained 34.9% of lignin in addition to 56.4% of proteins. Therefore, to reveal the effect by cacao proteins alone, the fecal weight and intestinal microbiota of mice fed the cacao protein and lignin diet were compared with those of mice fed the cacao lignin diet. The fecal weight of mice fed the cacao protein and lignin diet was significantly greater than of mice fed the cacao lignin diet. The relative abundance of Lactococcus and Mucispirillum species in mice fed the cacao protein and lignin diet was significantly higher than in mice fed the cacao lignin diet, but the relative abundance of Anaerotruncus, Oscillospira, and Roseburia species in mice fed the cacao protein and lignin diet was significantly lower than in mice fed the cacao lignin diet. Conclusions Ingestion of indigestible cacao proteins promoted defecation and altered the intestinal microbiota such as Lactococcus, Mucispirillum, Anaerotruncus, Oscillospira, and Roseburia species in mice.
Collapse
Affiliation(s)
- Jinichiro Koga
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Kota Ojiro
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Ayumi Yanagida
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Takahisa Suto
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Hideaki Hiki
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Yuki Inoue
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Chihiro Sakai
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Kohei Nakamoto
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Yuta Fujisawa
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Ayaka Orihara
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Haruka Murakami
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Shintaro Hirasawa
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Kengo Nakajima
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Tomoko Sakazawa
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| | - Hisakazu Yamane
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, Japan
| |
Collapse
|
26
|
Xu J, Chu T, Yu T, Li N, Wang C, Li C, Zhang Y, Meng H, Nie G. Design of Diselenide-Bridged Hyaluronic Acid Nano-antioxidant for Efficient ROS Scavenging to Relieve Colitis. ACS NANO 2022; 16:13037-13048. [PMID: 35861614 DOI: 10.1021/acsnano.2c05558] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Overproduction of reactive oxygen species (ROS), a key characteristic of inflammatory bowel disease (IBD), is responsible for dysregulation of signal transduction, inflammatory response, and DNA damage, which ultimately leads to disease progression and deterioration. Thus, ROS scavenging has become a promising strategy to navigate IBD. Inspired by the targeting capability of hyaluronic acid (HA) to CD44-overexpressed inflammatory cells together with the redox regulation capacity of diselenide compounds, we developed an oral nanoformulation, i.e., diselenide-bridged hyaluronic acid nanogel (SeNG), with a view to treat colitis through a ROS scavenging mechanism. Our data demonstrated that SeNG specifically accumulated in colitis tissue that was mediated by highly efficient CD44-HA interaction. This has allowed us to demonstrate a significant anti-inflammatory effect in an acute colitis mouse model induced by dextran sulfate sodium and trinitrobenzenesulfonic acid. Mechanistically, we continued to show SeNG reduced the ROS level via both direct elimination and up-regulation of the Nrf2/HO-1 signal pathway. Collectively, our work provides proof-of-principle evidence for a SeNG-mediated nano-antioxidant strategy, by which colitis could be effectively managed.
Collapse
Affiliation(s)
- Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
| | - Tianjiao Chu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School & Hospital of Stomatology, Zhongguancun South Avenue 22, Beijing 100081, China
| | - Naishi Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinlong Zhang
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Zhongguancun Beiyitiao 11, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
Dong Y, Xu T, Xiao G, Hu Z, Chen J. Opportunities and challenges for synthetic biology in the therapy of inflammatory bowel disease. Front Bioeng Biotechnol 2022; 10:909591. [PMID: 36032720 PMCID: PMC9399643 DOI: 10.3389/fbioe.2022.909591] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a complex, chronic intestinal inflammatory disorder that primarily includes Crohn’s disease (CD) and ulcerative colitis (UC). Although traditional antibiotics and immunosuppressants are known as the most effective and commonly used treatments, some limitations may be expected, such as limited efficacy in a small number of patients and gut flora disruption. A great many research studies have been done with respect to the etiology of IBD, while the composition of the gut microbiota is suggested as one of the most influential factors. Along with the development of synthetic biology and the continuing clarification of IBD etiology, broader prospects for novel approaches to IBD therapy could be obtained. This study presents an overview of the currently existing treatment options and possible therapeutic targets at the preclinical stage with respect to microbial synthesis technology in biological therapy. This study is highly correlated to the following topics: microbiota-derived metabolites, microRNAs, cell therapy, calreticulin, live biotherapeutic products (LBP), fecal microbiota transplantation (FMT), bacteriophages, engineered bacteria, and their functional secreted synthetic products for IBD medical implementation. Considering microorganisms as the main therapeutic component, as a result, the related clinical trial stability, effectiveness, and safety analysis may be the major challenges for upcoming research. This article strives to provide pharmaceutical researchers and developers with the most up-to-date information for adjuvant medicinal therapies based on synthetic biology.
Collapse
Affiliation(s)
- Yumeng Dong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Suzhou U-Synbio Co., Ltd., Suzhou, China
| | - Tiangang Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Guozheng Xiao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ziyan Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jingyu Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- *Correspondence: Jingyu Chen,
| |
Collapse
|
28
|
Laroute V, Beaufrand C, Gomes P, Nouaille S, Tondereau V, Daveran-Mingot ML, Theodorou V, Eutamene H, Mercier-Bonin M, Cocaign-Bousquet M. Lactococcus lactis NCDO2118 exerts visceral antinociceptive properties in rat via GABA production in the gastro-intestinal tract. eLife 2022; 11:77100. [PMID: 35727704 PMCID: PMC9213000 DOI: 10.7554/elife.77100] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/01/2022] [Indexed: 12/01/2022] Open
Abstract
Gut disorders associated to irritable bowel syndrome (IBS) are combined with anxiety and depression. Evidence suggests that microbially produced neuroactive molecules, like γ-aminobutyric acid (GABA), can modulate the gut-brain axis. Two natural strains of Lactococcus lactis and one mutant were characterized in vitro for their GABA production and tested in vivo in rat by oral gavage for their antinociceptive properties. L. lactis NCDO2118 significantly reduced visceral hypersensitivity induced by stress due to its glutamate decarboxylase (GAD) activity. L. lactis NCDO2727 with similar genes for GABA metabolism but no detectable GAD activity had no in vivo effect, as well as the NCDO2118 ΔgadB mutant. The antinociceptive effect observed for the NCDO2118 strain was mediated by the production of GABA in the gastro-intestinal tract and blocked by GABAB receptor antagonist. Only minor changes in the faecal microbiota composition were observed after the L. lactis NCDO2118 treatment. These findings reveal the crucial role of the microbial GAD activity of L. lactis NCDO2118 to deliver GABA into the gastro-intestinal tract for exerting antinociceptive properties in vivo and open avenues for this GRAS (Generally Recognized As safe) bacterium in the management of visceral pain and anxious profile of IBS patients.
Collapse
Affiliation(s)
- Valérie Laroute
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Catherine Beaufrand
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Pedro Gomes
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.,Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sébastien Nouaille
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Valérie Tondereau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Vassilia Theodorou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Hélène Eutamene
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Cocaign-Bousquet
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| |
Collapse
|
29
|
Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat Commun 2022; 13:3432. [PMID: 35701435 PMCID: PMC9198027 DOI: 10.1038/s41467-022-31171-0] [Citation(s) in RCA: 277] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/08/2022] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) play vital roles in intestinal inflammation. Therefore, eliminating ROS in the inflammatory site by antioxidant enzymes such as catalase and superoxide dismutase may effectively curb inflammatory bowel disease (IBD). Here, Escherichia coli Nissle 1917 (ECN), a kind of oral probiotic, was genetically engineered to overexpress catalase and superoxide dismutase (ECN-pE) for the treatment of intestinal inflammation. To improve the bioavailability of ECN-pE in the gastrointestinal tract, chitosan and sodium alginate, effective biofilms, were used to coat ECN-pE via a layer-by-layer electrostatic self-assembly strategy. In a mouse IBD model induced by different chemical drugs, chitosan/sodium alginate coating ECN-pE (ECN-pE(C/A)2) effectively relieved inflammation and repaired epithelial barriers in the colon. Unexpectedly, such engineered EcN-pE(C/A)2 could also regulate the intestinal microbial communities and improve the abundance of Lachnospiraceae_NK4A136 and Odoribacter in the intestinal flora, which are important microbes to maintain intestinal homeostasis. Thus, this study lays a foundation for the development of living therapeutic proteins using probiotics to treat intestinal-related diseases. Inflammatory bowel disease (IBD) is a complex disease that is associated with multiple genetic and environmental variables. Here the authors develop genetically engineered probiotics with selfproducing functional proteins and biofilm self-coating for safe and efficient IBD treatment in mice.
Collapse
|
30
|
Liu Y, Charamis N, Boeren S, Blok J, Lewis AG, Smid EJ, Abee T. Physiological Roles of Short-Chain and Long-Chain Menaquinones (Vitamin K2) in Lactococcus cremoris. Front Microbiol 2022; 13:823623. [PMID: 35369466 PMCID: PMC8965153 DOI: 10.3389/fmicb.2022.823623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/17/2022] [Indexed: 01/07/2023] Open
Abstract
Lactococcus cremoris and L. lactis are well known for their occurrence and applications in dairy fermentations, but their niche extends to a range of natural and food production environments. L. cremoris and L. lactis produce MKs (vitamin K2), mainly as the long-chain forms represented by MK-9 and MK-8, and a detectable number of short-chain forms represented by MK-3. The physiological significance of the different MK forms in the lifestyle of these bacterial species has not been investigated extensively. In this study, we used L. cremoris MG1363 to construct mutants producing different MK profiles by deletion of genes encoding (i) a menaquinone-specific isochorismate synthase, (ii) a geranyltranstransferase, and (iii) a prenyl diphosphate synthase. These gene deletions resulted in (i) a non-MK producer (ΔmenF), (ii) a presumed MK-1 producer (ΔispA), and (iii) an MK-3 producer (Δllmg_0196), respectively. By examining the phenotypes of the MG1363 wildtype strain and respective mutants, including biomass accumulation, stationary phase survival, oxygen consumption, primary metabolites, azo dye/copper reduction, and proteomes, under aerobic, anaerobic, and respiration-permissive conditions, we could infer that short-chain MKs like MK-1 and MK-3 are preferred to mediate extracellular electron transfer and reaction with extracellular oxygen, while the long-chain MKs like MK-9 and MK-8 are more efficient in aerobic respiratory electron transport chain. The different electron transfer routes mediated by short-chain and long-chain MKs likely support growth and survival of L. cremoris in a range of (transiently) anaerobic and aerobic niches including food fermentations, highlighting the physiological significance of diverse MKs in L. cremoris.
Collapse
Affiliation(s)
- Yue Liu
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Nikolaos Charamis
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, Netherlands
| | - Joost Blok
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | | | - Eddy J Smid
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Tjakko Abee
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
31
|
Wang R, Deng Y, Zhang Y, Li X, Sun L, Deng Q, Liu Y, Gooneratne R, Li J. Modulation of Intestinal Barrier, Inflammatory Response, and Gut Microbiota by Pediococcus pentosaceus zy-B Alleviates Vibrio parahaemolyticus Infection in C57BL/6J Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1865-1877. [PMID: 35107008 DOI: 10.1021/acs.jafc.1c07450] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Modulation of the intestinal barrier, inflammation, and gut microbiota by Pediococcus pentosaceus zy-B (zy-B) in Vibrio parahaemolyticus (Vp)-infected C57BL/6J mice was studied. Mice intragastrically pretreated with 108 colony-forming units (CFU) zy-B significantly alleviated Vp infection as evidenced by maintaining body weight and reduced disease activity index score and intestine ratio. In addition, zy-B reduced the Vp load in the ileum and cecum, significantly reduced the load in the colon, prevented colonic atrophy, and strengthened mucosal integrity. Mechanistically, zy-B ameliorated intestinal barrier dysfunction by upregulating tight junction protein expression, which in turn reduced the lipopolysaccharide, d-lactic acid (d-LA), and diamine oxidase concentrations and downregulated the cannabinoid receptor 1 (CB1) and CB2 mRNA expressions. Moreover, zy-B systemically reduced inflammation by decreasing interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α levels, and increased interleukin-10 (IL-10), immunoglobulin M (IgM), and immunoglobulin G (IgG) levels in the colon and serum. Furthermore, zy-B markedly altered the gut microbiota composition by enriching Bifidobacterium, Akkermansia, and Lactobacillus in the colon. Overall, zy-B appears to act as a probiotic to alleviate Vp infection by protecting the intestinal barrier, reducing inflammation, and promoting the growth of "beneficial" gut microbiota.
Collapse
Affiliation(s)
- Rundong Wang
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
- College of Food Science and Engineering, Lingnan Normal University, Zhanjiang 524048, China
| | - Yijia Deng
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Xuepeng Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Lijun Sun
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qi Deng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ying Liu
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury 7647, New Zealand
| | - Jianrong Li
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| |
Collapse
|
32
|
Su X, Yao B. Exploiting enzymes as a powerful tool to modulate the gut microbiota. Trends Microbiol 2022; 30:314-317. [PMID: 35120774 DOI: 10.1016/j.tim.2022.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
Abstract
Orally administered enzymes can have profound effects on the composition of the gut microbiota and may serve as an appealing alternative modulating agent. We summarize the three ways through which enzymes can influence the gut microbiota and discuss the challenges in choosing the right enzyme to modulate the gut microbiota.
Collapse
Affiliation(s)
- Xiaoyun Su
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 West Yuanmingyuan Road, Haidian District, Beijing 100193, China.
| | - Bin Yao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 West Yuanmingyuan Road, Haidian District, Beijing 100193, China.
| |
Collapse
|
33
|
Ren Y, Jiang W, Luo C, Zhang X, Huang M. Atractylenolide III Ameliorates TNBS-Induced Intestinal Inflammation in Mice by Reducing Oxidative Stress and Regulating Intestinal Flora. Chem Biodivers 2021; 18:e2001001. [PMID: 34156157 DOI: 10.1002/cbdv.202001001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/03/2021] [Indexed: 12/16/2022]
Abstract
The present study aimed to explore the therapeutic effects of the main active ingredients of Atractylodes macrocephala on the 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced mouse colitis model. TNBS-induced colitis was established in mice, which were treated with 8-β-Hydroxyasterolide (Atractylenolide III) for 14 days. The body weight of the mice in the middle and high dose groups of Atractylenolide III was increased compared with that of the model group. The disease activity index score was significantly reduced. The activity levels of myeloperoxidase were significantly decreased following increase in the dosage of Atractylenolide III, as determined by histological analysis. Moreover, Atractylenolide III downregulated the expression levels of the inflammatory factors interleukin-1β and tumor necrosis factor-α, and greatly suppressed the levels of the pro-oxidant markers, reactive oxygen species and malondialdehyde, while enhancing the expression levels of the antioxidant enzymes catalase, superoxide dismutase and glutathione peroxidase. The protein expression levels of formyl peptide receptor 1 (FPR1) and nuclear respiratory factor 2 (Nrf2) were upregulated in the colonic tissues of TNBS-treated mice. This effect was effectively reversed by Atractylenolide III treatment. In vivo studies indicated that TNBS alone induced a decrease in the abundance of lactobacilli and in the biodiversity of the colon. In conclusion, the present study suggested that Atractylenolide III attenuated TNBS-induced acute colitis by regulating oxidative stress via the FPR1 and Nrf2 pathways and by affecting the development of intestinal flora.
Collapse
Affiliation(s)
- Yan Ren
- College of Pharmaceutical Science, Guizhou University, Guiyang, 550025, P. R. China
| | - Wenwen Jiang
- College of Pharmaceutical Science, Guizhou University, Guiyang, 550025, P. R. China
| | - Chunli Luo
- College of Agriculture, Guizhou University, Guiyang, 550025, P. R. China
| | - Xiaohan Zhang
- College of Pharmaceutical Science, Guizhou University, Guiyang, 550025, P. R. China
| | - Mingjin Huang
- College of Agriculture, Guizhou University, Guiyang, 550025, P. R. China
| |
Collapse
|
34
|
Wilkinson JE, Franzosa EA, Everett C, Li C, Hu FB, Wirth DF, Song M, Chan AT, Rimm E, Garrett WS, Huttenhower C. A framework for microbiome science in public health. Nat Med 2021; 27:766-774. [PMID: 33820996 DOI: 10.1038/s41591-021-01258-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022]
Abstract
Human microbiome science has advanced rapidly and reached a scale at which basic biology, clinical translation and population health are increasingly integrated. It is thus now possible for public health researchers, practitioners and policymakers to take specific action leveraging current and future microbiome-based opportunities and best practices. Here we provide an outline of considerations for research, education, interpretation and scientific communication concerning the human microbiome and public health. This includes guidelines for population-scale microbiome study design; necessary physical platforms and analysis methods; integration into public health areas such as epidemiology, nutrition, chronic disease, and global and environmental health; entrepreneurship and technology transfer; and educational curricula. Particularly in the near future, there are both opportunities for the incorporation of microbiome-based technologies into public health practice, and a growing need for policymaking and regulation around related areas such as prebiotic and probiotic supplements, novel live-cell therapies and fecal microbiota transplants.
Collapse
Affiliation(s)
- Jeremy E Wilkinson
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric A Franzosa
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Christine Everett
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Chengchen Li
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Frank B Hu
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dyann F Wirth
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric Rimm
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Wendy S Garrett
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Curtis Huttenhower
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
35
|
Yu S, Qian H. Deoxyschizandrin treats mice with ulcerative colitis possibly via the TLR4/NF-κB signaling pathway. Am J Transl Res 2021; 13:3856-3863. [PMID: 34017577 PMCID: PMC8129281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/18/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE This study aimed to investigate the effects and mechanisms of deoxyschizandrin (DSD) on treatment of ulcerative colitis (UC). METHODS The models of mice with UC were established through dextran sulfate sodium (DSS) administration, and the successful models were treated with DSD. The therapeutic effects of DSD on UC mice were evaluated and its behind mechanisms were analyzed. RESULTS After DSS induction, the mice showed increased body weight and colon length, worse disease activity index (DAI) and body inflammation, oxidative stress injury and increased apoptosis of colonic epithelial cells, which were remarkably relieved after DSD intervention. Besides, the levels of TLR4, MyD88 and NF-κB in the colon tissues were elevated in UC mouse models, while DSD treatment reduced the levels of these markers. CONCLUSION DSD can alleviate the symptoms of mice with DSS-induced UC via inhibiting body inflammation, improving oxidative stress and reducing the apoptosis of colonic epithelial cells, which may be attributed to DSD inhibition of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Sheng Yu
- Surgery of Traditional Chinese Medicine, Nanjing University of Traditional Chinese Medicine, The First Clinical Medical CollegeNanjing 210000, Jiangsu Province, China
| | - Haihua Qian
- Department of Anorectal Diseases, North Hospital of Jiangsu Province Traditional Chinese MedicineNanjing 210000, Jiangsu Province, China
| |
Collapse
|
36
|
Martínez B, Rodríguez A, Kulakauskas S, Chapot-Chartier MP. Cell wall homeostasis in lactic acid bacteria: threats and defences. FEMS Microbiol Rev 2021; 44:538-564. [PMID: 32495833 PMCID: PMC7476776 DOI: 10.1093/femsre/fuaa021] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
Abstract
Lactic acid bacteria (LAB) encompasses industrially relevant bacteria involved in food fermentations as well as health-promoting members of our autochthonous microbiota. In the last years, we have witnessed major progresses in the knowledge of the biology of their cell wall, the outermost macrostructure of a Gram-positive cell, which is crucial for survival. Sophisticated biochemical analyses combined with mutation strategies have been applied to unravel biosynthetic routes that sustain the inter- and intra-species cell wall diversity within LAB. Interplay with global cell metabolism has been deciphered that improved our fundamental understanding of the plasticity of the cell wall during growth. The cell wall is also decisive for the antimicrobial activity of many bacteriocins, for bacteriophage infection and for the interactions with the external environment. Therefore, genetic circuits involved in monitoring cell wall damage have been described in LAB, together with a plethora of defence mechanisms that help them to cope with external threats and adapt to harsh conditions. Since the cell wall plays a pivotal role in several technological and health-promoting traits of LAB, we anticipate that this knowledge will pave the way for the future development and extended applications of LAB.
Collapse
Affiliation(s)
- Beatriz Martínez
- DairySafe research group. Department of Technology and Biotechnology of Dairy Products. Instituto de Productos Lácteos de Asturias, IPLA-CSIC. Paseo Río Linares s/n. 33300 Villaviciosa, Spain
| | - Ana Rodríguez
- DairySafe research group. Department of Technology and Biotechnology of Dairy Products. Instituto de Productos Lácteos de Asturias, IPLA-CSIC. Paseo Río Linares s/n. 33300 Villaviciosa, Spain
| | - Saulius Kulakauskas
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
37
|
Delgado-Venegas CS, Martínez-Hernández SL, Cervantes-García D, Montes de Oca-Luna R, de Jesús Loera-Arias M, Mata-Martínez MG, Ventura-Juárez J, Muñoz-Ortega MH. Modulating effects of the probiotic Lactococcus lactis on the hepatic fibrotic process induced by CCl 4 in Wistar rats. Exp Ther Med 2021; 21:339. [PMID: 33732312 PMCID: PMC7903445 DOI: 10.3892/etm.2021.9770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatic cirrhosis is a chronic disease that affects one fifth of the World's population and is the third leading cause of death in Mexico. Attempts have been made to develop treatments for this hepatic cirrhosis, which include manipulating the intestinal microbiota and thus decreasing the early inflammatory response. The microbiota is reportedly altered in patients with cirrhosis. Due to its immunomodulatory properties and its ability to survive in the gastrointestinal tract, Lactococcus lactis (L. lactis) has been used as a therapeutic measure in inflammatory disorders of the colon. The objective of the present study was to evaluate the efficacy of the L. lactis probiotic NZ9000 in preventing tetrachloromethane (CCl4)-induced experimental hepatic fibrosis. The following 4 groups were included in the experimental stage (n=5): i) Control group; ii) L. lactis group; iii) CCl4 group; and iv) L. lactis-CCl4 group. For the first 2 weeks, L. lactis was orally administered to the L. lactis and L. lactis-CCl4 groups; CCl4 was then peritoneally administered to the lactis-CCl4 group for a further 4 weeks (in addition to the probiotic), while the L. lactis group received the probiotic only. For the CCl4 group, CCl4 was administered for 4 weeks. The experimental groups were all compared with the control group and the L. lactis + CCl4 group. Tissue samples were analyzed histologically and biochemically, and the gene expression levels of interleukin (IL)-1, IL-10 and forkhead box protein P3 (FoxP3) were determined. L. lactis decreased hepatic cirrhosis by preventing steatosis and fibrosis, and by reducing the levels of AST and ALT. Subchronic CCl4 injury induced upregulation of the IL-1β gene in the liver, which was decreased by L. lactis. It was also found that the group treated with L. lactis showed increased expression of Foxp3 in the liver and IL-10 in the gut. These results suggested that oral administration of L. lactis may be a potential probiotic to prevent or protect against CCl4-induced liver injury.
Collapse
Affiliation(s)
| | - Sandra Luz Martínez-Hernández
- Center for Basic Sciences, Department of Morphology, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico
| | - Daniel Cervantes-García
- National Council of Science and Technology, Center for Basic Sciences, Department of Microbiology, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico
| | - Roberto Montes de Oca-Luna
- Faculty of Medicine, Department of Histology, Autonomous University of Nuevo Léon, Monterrey, Nuevo León, 64460, Mexico
| | - María de Jesús Loera-Arias
- Faculty of Medicine, Department of Histology, Autonomous University of Nuevo Léon, Monterrey, Nuevo León, 64460, Mexico
| | - María Guadalupe Mata-Martínez
- Center for Basic Sciences, Department of Morphology, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico
| | - Javier Ventura-Juárez
- Center for Basic Sciences, Department of Morphology, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico
| | - Martín Humberto Muñoz-Ortega
- Center of Basic Sciences, Department of Chemistry, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico
| |
Collapse
|
38
|
Yang F, Zou Q, Gao B. GutBalance: a server for the human gut microbiome-based disease prediction and biomarker discovery with compositionality addressed. Brief Bioinform 2021; 22:6123951. [PMID: 33515036 DOI: 10.1093/bib/bbaa436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/17/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
The compositionality of the microbiome data is well-known but often neglected. The compositional transformation pertains to the supervised learning of microbiome data and is a critical step that decides the performance and reliability of the disease classifiers. We value the excellent performance of the distal discriminative balance analysis (DBA) method, which selects distal balances of pairs and trios of bacteria, in addressing the classification of high-dimensional microbiome data. By applying this method to the species-level abundances of all the disease phenotypes in the GMrepo database, we build a balance-based model repository for the classification of human gut microbiome-related diseases. The model repository supports the prediction of disease risks for new sample(s). More importantly, we highlight the concept of balance-disease associations rather than the conventional microbe-disease associations and develop the human Gut Balance-Disease Association Database (GBDAD). Each predictable balance for each disease model indicates a potential biomarker-disease relationship and can be interpreted as a bacteria ratio positively or negatively correlated with the disease. Furthermore, by linking the balance-disease associations to the evidenced microbe-disease associations in MicroPhenoDB, we surprisingly found that most species-disease associations inferred from the shotgun metagenomic datasets can be validated by external evidence beyond MicroPhenoDB. The balance-based species-disease association inference will accelerate the generation of new microbe-disease association hypotheses in gastrointestinal microecology research and clinical trials. The model repository and the GBDAD database are deployed on the GutBalance server, which supports interactive visualization and systematic interrogation of the disease models, disease-related balances and disease-related species of interest.
Collapse
Affiliation(s)
- Fenglong Yang
- University of Electronic Science and Technology of China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
- Hainan Key Laboratory for Computational Science and Application, Hainan Normal University, Haikou 571158, China
| | - Bo Gao
- Department of Radiology, The Second Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| |
Collapse
|
39
|
Larsen IS, Jensen BAH, Bonazzi E, Choi BSY, Kristensen NN, Schmidt EGW, Süenderhauf A, Morin L, Olsen PB, Hansen LBS, Schröder T, Sina C, Chassaing B, Marette A. Fungal lysozyme leverages the gut microbiota to curb DSS-induced colitis. Gut Microbes 2021; 13:1988836. [PMID: 34693864 PMCID: PMC8547870 DOI: 10.1080/19490976.2021.1988836] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/15/2021] [Accepted: 09/27/2021] [Indexed: 02/08/2023] Open
Abstract
Colitis is characterized by colonic inflammation and impaired gut health. Both features aggravate obesity and insulin resistance. Host defense peptides (HDPs) are key regulators of gut homeostasis and generally malfunctioning in above-mentioned conditions. We aimed here to improve bowel function in diet-induced obesity and chemically induced colitis through daily oral administration of lysozyme, a well-characterized HDP, derived from Acremonium alcalophilum.C57BL6/J mice were fed either low-fat reference diet or HFD ± daily gavage of lysozyme for 12 weeks, followed by metabolic assessment and evaluation of colonic microbiota encroachment. To further evaluate the efficacy of intestinal inflammation, we next supplemented chow-fed BALB/c mice with lysozyme during Dextran Sulfate Sodium (DSS)-induced colitis in either conventional or microbiota-depleted mice. We assessed longitudinal microbiome alterations by 16S amplicon sequencing in both models.Lysozyme dose-dependently alleviated intestinal inflammation in DSS-challenged mice and further protected against HFD-induced microbiota encroachment and fasting hyperinsulinemia. Observed improvements of intestinal health relied on a complex gut flora, with the observation that microbiota depletion abrogated lysozyme's capacity to mitigate DSS-induced colitis.Akkermansia muciniphila associated with impaired gut health in both models, a trajectory that was mitigated by lysozyme administration. In agreement with this notion, PICRUSt2 analysis revealed specific pathways consistently affected by lysozyme administration, independent of vivarium, disease model and mouse strain.Taking together, lysozyme leveraged the gut microbiota to curb DSS-induced inflammation, alleviated HFD-induced gastrointestinal disturbances and lowered fasting insulin levels in obese mice. Collectively, these data present A. alcalophilum-derived lysozyme as a promising candidate to enhance gut health.
Collapse
Affiliation(s)
- Ida Søgaard Larsen
- Quebec Heart and Lung Institute (Iucpq), Faculty of Medicine, and Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
| | - Benjamin A. H. Jensen
- Quebec Heart and Lung Institute (Iucpq), Faculty of Medicine, and Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erica Bonazzi
- Inserm U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, Université De Paris, Paris, France
| | - Béatrice S. Y. Choi
- Quebec Heart and Lung Institute (Iucpq), Faculty of Medicine, and Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
| | | | | | - Annika Süenderhauf
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Laurence Morin
- Quebec Heart and Lung Institute (Iucpq), Faculty of Medicine, and Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
| | | | | | - Torsten Schröder
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Christian Sina
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Benoît Chassaing
- Inserm U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, Université De Paris, Paris, France
| | - André Marette
- Quebec Heart and Lung Institute (Iucpq), Faculty of Medicine, and Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
| |
Collapse
|
40
|
Obesity Worsens Gulf War Illness Symptom Persistence Pathology by Linking Altered Gut Microbiome Species to Long-Term Gastrointestinal, Hepatic, and Neuronal Inflammation in a Mouse Model. Nutrients 2020; 12:nu12092764. [PMID: 32927823 PMCID: PMC7551189 DOI: 10.3390/nu12092764] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/30/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
Persistence of Gulf War illness (GWI) pathology among deployed veterans is a clinical challenge even after almost three decades. Recent studies show a higher prevalence of obesity and metabolic disturbances among Gulf War veterans primarily due to the existence of post-traumatic stress disorder (PTSD), chronic fatigue, sedentary lifestyle, and consumption of a high-carbohydrate/high-fat diet. We test the hypothesis that obesity from a Western-style diet alters host gut microbial species and worsens gastrointestinal and neuroinflammatory symptom persistence. We used a 5 month Western diet feeding in mice that received prior Gulf War (GW) chemical exposure to mimic the home phase obese phenotype of the deployed GW veterans. The host microbial profile in the Western diet-fed GWI mice showed a significant decrease in butyrogenic and immune health-restoring bacteria. The altered microbiome was associated with increased levels of IL6 in the serum, Claudin-2, IL6, and IL1β in the distal intestine with concurrent inflammatory lesions in the liver and hyperinsulinemia. Microbial dysbiosis was also associated with frontal cortex levels of increased IL6 and IL1β, activated microglia, decreased levels of brain derived neurotrophic factor (BDNF), and higher accumulation of phosphorylated Tau, an indicator of neuroinflammation-led increased risk of cognitive deficiencies. Mechanistically, serum from Western diet-fed mice with GWI significantly increased microglial activation in transformed microglial cells, increased tyrosyl radicals, and secreted IL6. Collectively, the results suggest that an existing obese phenotype in GWI worsens persistent gastrointestinal and neuronal inflammation, which may contribute to poor outcomes in restoring cognitive function and resolving fatigue, leading to the deterioration of quality of life.
Collapse
|
41
|
Liu Y, Luo L, Luo Y, Zhang J, Wang X, Sun K, Zeng L. Prebiotic Properties of Green and Dark Tea Contribute to Protective Effects in Chemical-Induced Colitis in Mice: A Fecal Microbiota Transplantation Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6368-6380. [PMID: 32419454 DOI: 10.1021/acs.jafc.0c02336] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Green and dark tea extract (GTE/DTE) ameliorate chemical-induced colitis in mice; however, the role of gut microbiota in the anticolitis effects of green and dark tea in mice remains unclear. This study aims to explore the role of modulations in gut microbes mediated by green and dark tea in colitis mice by fecal microbiota transplantation (FMT). Our results indicated that GTE and DTE (5 mg/kg bodyweight/day for 4 weeks) exhibited prebiotic effects on the donor mice. Moreover, the FMT treatments (transferring the microbiota daily from the 1 g/kg bodyweight fecal sample to each recipient) indicated that, compared with the fecal microbiota from the normal diet-treated donor mice, the fecal microbiota from the GTE- and DTE-treated donor mice significantly ameliorate colitis-related symptoms (e.g., loss of bodyweight, colonic inflammation, loss of barrier integrity, and gut microbiota dysbiosis) and downregulated the TLR4/MyD88/NF-κB pathway. Collectively, GTE and DTE ameliorate chemical-induced colitis by modulating gut microbiota.
Collapse
Affiliation(s)
- Yan Liu
- College of Food Science, Southwest University, Beibei, Chongqing 400715, People's Republic of China
| | - Liyong Luo
- College of Food Science, Southwest University, Beibei, Chongqing 400715, People's Republic of China
- Tea Research Institute, Southwest University, Beibei, Chongqing 400715, People's Republic of China
| | - Yakun Luo
- Tea Research Institute of Puer, Puer, Yunnan 665000, People's Republic of China
| | - Jun Zhang
- Tea Research Institute of Puer, Puer, Yunnan 665000, People's Republic of China
| | - Xinghua Wang
- Tea Research Institute of Puer, Puer, Yunnan 665000, People's Republic of China
| | - Kang Sun
- College of Food Science, Southwest University, Beibei, Chongqing 400715, People's Republic of China
- Tea Research Institute, Southwest University, Beibei, Chongqing 400715, People's Republic of China
| | - Liang Zeng
- College of Food Science, Southwest University, Beibei, Chongqing 400715, People's Republic of China
- Tea Research Institute, Southwest University, Beibei, Chongqing 400715, People's Republic of China
| |
Collapse
|
42
|
The High-Fat Diet Based on Extra-Virgin Olive Oil Causes Dysbiosis Linked to Colorectal Cancer Prevention. Nutrients 2020; 12:nu12061705. [PMID: 32517306 PMCID: PMC7352482 DOI: 10.3390/nu12061705] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
The present study aims to examine the effects of three different high-fat diet (HFD) on mice gut microbiota in order to analyse whether they create the microenvironmental conditions that either promote or prevent colorectal cancer (CRC). We evaluated colonic mucosa-associated microbiota in CD1 mice fed with HFD, based on 60% kcal from fat-containing coconut, sunflower or extra-virgin olive oil as the only source of fat. The main findings were as follows: (a) All HFD produced a decrease in the richness and diversity of the intestinal microbiota that was independent of mouse weight, (b) HFD switched Lactobacillus to Lactococcus. In general, the results showed that both sunflower- and coconut-HFD generated a pro-inflammatory intestinal microenvironment. In brief, coconut-HFD decreased Akkermansia and increased Staphylococcus, Prevotella and Bacteroides spp. abundance. Sunflower-HFD reduced Akkermansia and Bifidobacterium, while enhancing Sphingomonas and Neisseria spp. abundance. In contrast, EVOO-HFD produced an anti-inflammatory microenvironment characterised by a decreased Enterococcus, Staphylococcus, Neisseria and Pseudomonas spp. abundance. At the same time, it increased the Firmicutes/Bacteroidetes ratio and maintained the Akkermansia population. To conclude, EVOO-HFD produced changes in the gut microbiota that are associated with the prevention of CRC, while coconut and sunflower-HFD caused changes associated with an increased risk of CRC.
Collapse
|
43
|
Kim JH, Kordahi MC, Chac D, DePaolo RW. Toll-like Receptor-6 Signaling Prevents Inflammation and Impacts Composition of the Microbiota During Inflammation-Induced Colorectal Cancer. Cancer Prev Res (Phila) 2019; 13:25-40. [PMID: 31771941 DOI: 10.1158/1940-6207.capr-19-0286] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/06/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022]
Abstract
Tightly regulated immune responses must occur in the intestine to avoid unwanted inflammation, which may cause chronic sequela leading to diseases such as colorectal cancer. Toll-like receptors play an important role in preventing aberrant immune responses in the intestine by sensing endogenous commensal microbiota and delivering important regulatory signals to the tissue. However, the role that specific innate receptors may play in the development of chronic inflammation and their impact on the composition of the colonic microbiota is not well understood. Using a model of inflammation-induced colorectal cancer, we found that Lactobacillus species are lost more quickly in wild-type (WT) mice than TLR6-deficient mice resulting in overall differences in bacterial composition. Despite the longer retention of Lactobacillus, the TLR6-deficient mice presented with more tumors and a worse overall outcome. Restoration of the lost Lactobacillus species suppressed inflammation, reduced tumor number, and prevented change in the abundance of Proteobacteria only when given to WT mice, indicating the effect of these Lactobacillus are TLR6 dependent. We found that the TLR6-dependent effects of Lactobacillus could be dissociated from one another via the involvement of IL10, which was necessary to dampen the inflammatory microenvironment, but had no effect on bacterial composition. Altogether, these data suggest that innate immune signals can shape the composition of the microbiota under chronic inflammatory conditions, bias the cytokine milieu of the tissue microenvironment, and influence the response to microbiota-associated therapies.
Collapse
Affiliation(s)
- Jee-Hyun Kim
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Melissa C Kordahi
- Division of Pathology, Department of Medicine, University of Washington, Seattle, Washington
| | - Denise Chac
- Division of Pathology, Department of Medicine, University of Washington, Seattle, Washington
| | - R William DePaolo
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
44
|
Zhao F, Song S, Ma Y, Xu X, Zhou G, Li C. A Short-Term Feeding of Dietary Casein Increases Abundance of Lactococcus lactis and Upregulates Gene Expression Involving Obesity Prevention in Cecum of Young Rats Compared With Dietary Chicken Protein. Front Microbiol 2019; 10:2411. [PMID: 31708891 PMCID: PMC6824296 DOI: 10.3389/fmicb.2019.02411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Casein and chicken are assessed to contain high quality proteins, which are essential for human health. Studies have shown that ingestion of the two dietary proteins resulted in distinct effects on physiology, liver transcriptome and gut microbiota. However, its underlying mechanism is not fully understood, in particular for a crosstalk between gut microbiota and host under a specific diet intervention. We fed young rats with a casein or a chicken protein-based diet (CHPD) for 7 days, and characterized cecal microbiota composition and cecal gene expression. We found that a short-term intervention with a casein-based diet (CAD) induced a higher relative abundance of beneficial bacterium Lactococcus lactis as well as Bifidobacterium pseudolongum, which upregulated galactose metabolism of the microbiome compared with a CHPD. The CAD also upregulated gene expression involved in obesity associated pathways (e.g., Adipoq and Irs1) in cecal tissue of rats. These genes and the bacterial taxon were reported to play an important role in protecting development of obesity. Furthermore, the differentially represented bacterial taxon L. lactis was positively associated with these differentially expressed genes in the gut tissue. Our results provide a new insight into the crosstalk between gut microbiota and host in response to dietary proteins, indicating a potential mechanism of obesity prevention function by casein.
Collapse
Affiliation(s)
- Fan Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Key Laboratory of Meat Products Processing, Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, China
| | - Shangxin Song
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, China
| | - Yafang Ma
- College of Food Science and Technology, Nanjing Agricultural University, Key Laboratory of Meat Products Processing, Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, China
| | - Xinglian Xu
- College of Food Science and Technology, Nanjing Agricultural University, Key Laboratory of Meat Products Processing, Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, China
| | - Guanghong Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Key Laboratory of Meat Products Processing, Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, China
| | - Chunbao Li
- College of Food Science and Technology, Nanjing Agricultural University, Key Laboratory of Meat Products Processing, Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, China
| |
Collapse
|
45
|
Sheng Y, Li H, Liu M, Xie B, Wei W, Wu J, Meng F, Wang HY, Chen S. A Manganese-Superoxide Dismutase From Thermus thermophilus HB27 Suppresses Inflammatory Responses and Alleviates Experimentally Induced Colitis. Inflamm Bowel Dis 2019; 25:1644-1655. [PMID: 31067299 DOI: 10.1093/ibd/izz097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Superoxide dismutase (SOD) is an attractive therapeutic agent to ameliorate oxidative stress that is critical for the initiation and progression of inflammatory bowel disease (IBD). However, the short life of SOD limits its clinical application. In this study, we aim to examine the therapeutic effects of a hyperthermostable SOD from the Thermus thermophilus HB27 (TtSOD) for treatment of experimentally induced IBD. METHODS A recombinant TtSOD was expressed and purified from Escherichia coli, and its therapeutic effects were examined in 2 experimental IBD animal models. RESULTS In IBD induced by 2,4,6-trinitrobenzenesulfonic acid in zebrafish, TtSOD treatment decreased intestinal enlargement and attenuated neutrophil infiltration, resulting in alleviation of enterocolitis. In mice, SOD activity was substantially increased in the intestine after oral gavage of TtSOD, which ameliorated gut inflammation, preserved gut barrier function, and attenuated the severity of dextran sulfate sodium-induced colitis. Furthermore, TtSOD inhibited lipopolysaccharide-induced production of reactive oxygen species and inflammatory responses in mouse bone marrow-derived macrophages. CONCLUSIONS Our results demonstrate that TtSOD possesses therapeutic activities toward experimentally induced IBD, offering new clinical treatment options for patients with IBD.
Collapse
Affiliation(s)
- Yang Sheng
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Hailong Li
- Institute of Molecular Enzymology, Medical College of Soochow University, Jiangsu, China
| | - Minjun Liu
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Bingxian Xie
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Wen Wei
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Jiawei Wu
- Institute of Molecular Enzymology, Medical College of Soochow University, Jiangsu, China
| | - Fanguo Meng
- Redox Medical Center for Public Health, Medical College of Soochow University, Jiangsu, China
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Shamoon M, Martin NM, O'Brien CL. Recent advances in gut Microbiota mediated therapeutic targets in inflammatory bowel diseases: Emerging modalities for future pharmacological implications. Pharmacol Res 2019; 148:104344. [PMID: 31400403 DOI: 10.1016/j.phrs.2019.104344] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 02/09/2023]
Abstract
The inflammatory bowel diseases (IBDs) are chronic inflammatory conditions, which are increasing in prevalence worldwide. The IBDs are thought to result from an aberrant immune response to gut microbes in genetically susceptible individuals. Dysbiosis of the gut microbiome, both functional and compositional, promotes patient susceptibility to colonization by pathobionts. Manipulating gut microbial communities and gut microbiota-immune system interactions to restore gut homeostasis or reduce inflammation are appealing therapeutic models. We discuss the therapeutic potential of precision microbiota editing, natural and engineered probiotics, the use of gut microbiota-derived metabolites in colitogenic phenotypes, and intestinal stem cells, in maintaining gut microbiota balance, restoring the mucosal barrier, and having positive immunomodulatory effects in experimental IBD. This review highlights that we are only just beginning to understand the complexity of the microbiota and how it can be manipulated for health benefits, including treatment and prevention of the clinical IBDs in future.
Collapse
Affiliation(s)
- Muhammad Shamoon
- Biology Section, Shandong Normal University Affiliated Senior School, Jinan 250001, PR China
| | - Natalia M Martin
- Medical School, The Australian National University, Canberra ACT 2600, Australia
| | - Claire L O'Brien
- Medical School, The Australian National University, Canberra ACT 2600, Australia; IBD Research Group, Canberra Hospital, Canberra, Australia.
| |
Collapse
|
47
|
Bisanz JE, Upadhyay V, Turnbaugh JA, Ly K, Turnbaugh PJ. Meta-Analysis Reveals Reproducible Gut Microbiome Alterations in Response to a High-Fat Diet. Cell Host Microbe 2019; 26:265-272.e4. [PMID: 31324413 DOI: 10.1016/j.chom.2019.06.013] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/22/2019] [Accepted: 06/23/2019] [Indexed: 02/08/2023]
Abstract
Multiple research groups have shown that diet impacts the gut microbiome; however, variability in experimental design and quantitative assessment have made it challenging to assess the degree to which similar diets have reproducible effects across studies. Through an unbiased subject-level meta-analysis framework, we re-analyzed 27 dietary studies including 1,101 samples from rodents and humans. We demonstrate that a high-fat diet (HFD) reproducibly changes gut microbial community structure. Finer taxonomic analysis revealed that the most reproducible signals of a HFD are Lactococcus species, which we experimentally demonstrate to be common dietary contaminants. Additionally, a machine-learning approach defined a signature that predicts the dietary intake of mice and demonstrated that phylogenetic and gene-centric transformations of this model can be translated to humans. Together, these results demonstrate the utility of microbiome meta-analyses in identifying robust and reproducible features for mechanistic studies in preclinical models.
Collapse
Affiliation(s)
- Jordan E Bisanz
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vaibhav Upadhyay
- Department of Internal Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jessie A Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kimberly Ly
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
48
|
Gaudu P, Yamamoto Y, Jensen PR, Hammer K, Lechardeur D, Gruss A. Genetics of Lactococci. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0035-2018. [PMID: 31298208 PMCID: PMC10957224 DOI: 10.1128/microbiolspec.gpp3-0035-2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Indexed: 11/20/2022] Open
Abstract
Lactococcus lactis is the best characterized species among the lactococci, and among the most consumed food-fermenting bacteria worldwide. Thanks to their importance in industrialized food production, lactococci are among the lead bacteria understood for fundamental metabolic pathways that dictate growth and survival properties. Interestingly, lactococci belong to the Streptococcaceae family, which includes food, commensal and virulent species. As basic metabolic pathways (e.g., respiration, metal homeostasis, nucleotide metabolism) are now understood to underlie virulence, processes elucidated in lactococci could be important for understanding pathogen fitness and synergy between bacteria. This chapter highlights major findings in lactococci and related bacteria, and covers five themes: distinguishing features of lactococci, metabolic capacities including the less known respiration metabolism in Streptococcaceae, factors and pathways modulating stress response and fitness, interbacterial dialogue via metabolites, and novel applications in health and biotechnology.
Collapse
Affiliation(s)
| | - Yuji Yamamoto
- Laboratory of Cellular Microbiology, School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Aomori Japan
| | - Peter Ruhdal Jensen
- National Food Institute, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | - Karin Hammer
- DTU Bioengineering, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | | | | |
Collapse
|
49
|
The Bidirectional Interactions between Resveratrol and Gut Microbiota: An Insight into Oxidative Stress and Inflammatory Bowel Disease Therapy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5403761. [PMID: 31179328 PMCID: PMC6507241 DOI: 10.1155/2019/5403761] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/16/2022]
Abstract
Dysbiosis and oxidative stress in the gut have contributed to the progression of intestinal inflammatory bowel disease (IBD). The current study has reported that enteric bacteria mediate redox homeostasis through the regulation of reactive oxygen species (ROS) production. Resveratrol, one of the most abundant polyphenols, with poor oral bioavailability, is considered as a scavenger of ROS and other free radicals. Recent studies have shown that resveratrol effectively enhances the growth of Lactococcus lactis and inhibits the growth of Enterococcus faecalis. (1) In terms of the two-way relationship between gut microbiota and resveratrol, resveratrol modulates gut microbiota; (2) in terms of resveratrol biotransformation by gut microbiota, we speculate that gut microbiota could be a target of resveratrol to maintain gut homeostasis. Here, we reviewed the current researches about the cellular signaling pathways in intestinal epithelial cells triggered by gut microbiota in response to oxidative stress. These results suggest that the modulation of the gut microbiota through resveratrol supplementation appears as a promising potential approach for the therapy of inflammatory bowel disease.
Collapse
|
50
|
Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy. Microorganisms 2019; 7:microorganisms7040097. [PMID: 30987157 PMCID: PMC6518429 DOI: 10.3390/microorganisms7040097] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 12/12/2022] Open
Abstract
Pelvic radiotherapy has been frequently reported to cause acute and late onset gastrointestinal (GI) toxicities associated with significant morbidity and mortality. Although the underlying mechanisms of pelvic radiation-induced GI toxicity are poorly understood, they are known to involve a complex interplay between all cell types comprising the intestinal wall. Furthermore, increasing evidence states that the human gut microbiome plays a role in the development of radiation-induced health damaging effects. Gut microbial dysbiosis leads to diarrhea and fatigue in half of the patients. As a result, reinforcement of the microbiome has become a hot topic in various medical disciplines. To counteract GI radiotoxicities, apart from traditional pharmacological compounds, adjuvant therapies are being developed including food supplements like vitamins, prebiotics, and probiotics. Despite the easy, cheap, safe, and feasible approach to protect patients against acute radiation-induced toxicity, clinical trials have yielded contradictory results. In this review, a detailed overview is given of the various clinical, intestinal manifestations after pelvic irradiation as well as the role of the gut microbiome herein. Furthermore, whilst discussing possible strategies to prevent these symptoms, food supplements are presented as auspicious, prophylactic, and therapeutic options to mitigate acute pelvic radiation-induced GI injury by exploring their molecular mechanisms of action.
Collapse
|