1
|
Liu F, Zhang H, Fan L, Yu Q, Wang S. Hotspots and development trends of gut microbiota in atopic dermatitis: A bibliometric analysis from 1988 to 2024. Medicine (Baltimore) 2024; 103:e40931. [PMID: 39686442 DOI: 10.1097/md.0000000000040931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a prevalent inflammatory skin condition that commonly occurs in children. More and more scientific evidence suggests that gut microbiota plays an important role in the pathogenesis of AD, whereas there is no article providing a comprehensive summary and analysis. We aimed to analyze documents on AD and gut microbiota and identify hotspots and development trends in this field. METHODS Articles and reviews in the field of AD and gut microbiota from January 1, 1988 to October 20, 2024 were obtained from the Web of Science Core Collection database. Biblioshiny was utilized for evaluating and visualizing the core authors, journals, countries, documents, trend topics, and hotspots in this field. RESULTS Among 1672 documents, it indicated that the number of annual publications generally increased. The United States had the highest production, impact, and international collaboration. Journal of Allergy and Clinical Immunology was the journal of the maximum publications. Based on keyword co-occurrence and clustering analysis, "stratum-corneum lipids," "probiotics," "prebiotics," "fecal microbiota transplantation," "phage therapy," "short chain fatty-acids," "biologic therapy," and "skin inflammation" represented current trend topics. The pathological and molecular mechanisms and associated therapeutic methods for AD and gut microbiota were the research hotspots. The incorporation of microbiota-based therapies alongside conventional treatments can contribute to better clinical outcomes. CONCLUSION We highlighted that gut microbiota may exacerbate symptoms of AD through various aspects, including immunity, metabolites, and neuroendocrine pathways. More efforts are required to investigate the safety and efficacy of gut microbial management methods for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Fang Liu
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Haipeng Zhang
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Lina Fan
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Qi Yu
- Chengdong College, Northeast Agricultural University, Harbin, China
| | - Siqiao Wang
- Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Luo HY, Fang J, Zhang WH, Chan KC, Chan YM, Dong CX, Li SL, Lyu AP, Xu J. Dissecting the anti-obesity components of ginseng: How ginseng polysaccharides and ginsenosides target gut microbiota to suppress high-fat diet-induced obesity. J Adv Res 2024:S2090-1232(24)00558-7. [PMID: 39672231 DOI: 10.1016/j.jare.2024.12.002] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024] Open
Abstract
INTRODUCTION Ginseng demonstrates therapeutic potential in treating obesity, with both experimental and clinical studies suggesting its anti-obesity effects are mediated by gut microbiota. Nonetheless, the specific chemical components responsible for this effect remain largely unidentified. OBJECTIVES This study aims to investigate the anti-obesity effects and mechanisms of ginseng polysaccharides (GP) and ginsenosides (GS), the primary chemical components of ginseng, with a focus on their impact on gut microbiota. METHODS The impact of GP and GS on high-fat diet (HFD)-induced obesity was assessed using a mouse model. Molecular mechanisms were explored through a combination of chemical analysis, metagenomics, RT-qPCR, ELISA, and biochemical assays. RESULTS GP or GS administration effectively prevented adiposity in HFD-fed mice, and both effects were mediated by gut microbiota. Chemical analysis revealed diverse glycosyl groups in GP and GS. Metagenomics data suggested that GP-enriched species, e.g., Bacteroides stercorirosoris and Clostridiales bacterium encoded carbohydrate-active enzymes GH35, GH43 and PL9_1, while GS-enriched Sulfurospirillum halorespirans encoded GH16_5. These enzymes facilitated the utilization of glycosyl groups in GP and GS, selectively stimulating bacterial growth and reshaping the gut microbiota. Furthermore, bacterial species enriched by GP or GS encoded specific functional genes involved in short-chain fatty acid (SCFA) synthesis (K00625 and K00925 for GP; K18118, K00100, and K18122 for GS) and intestinal gluconeogenesis (IGN) (K01678, K00024, and K01596 for GP; K18118 and K00278 for GS). Consequently, the SCFA-GLP-1/PYY signaling and IGN were activated by both GP and GS to ameliorate obesity phenotypes. CONCLUSION GP and GS, containing diverse glycosyl groups, selectively stimulate specific gut bacteria, triggering mechanisms involved in SCFA-GLP-1/PYY signaling and IGN activation to reduce adiposity in HFD-fed mice. The study enhances understanding of the chemical components crucial for the gut microbiota-mediated anti-obesity effect of ginseng. The mechanistic understanding provides valuable insights for developing ginseng-based drugs or health products to combat obesity.
Collapse
Affiliation(s)
- Han-Yan Luo
- Institute of Systems Medicine and Health Science, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong; Research Centre for Standardization of Chinese Medicines, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong
| | - Jing Fang
- Research Centre for Standardization of Chinese Medicines, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong
| | - Wei-Hao Zhang
- Institute of Systems Medicine and Health Science, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong; Research Centre for Standardization of Chinese Medicines, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong
| | - Kam-Chun Chan
- Institute of Systems Medicine and Health Science, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong; Research Centre for Standardization of Chinese Medicines, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong
| | - Yui-Man Chan
- Institute of Systems Medicine and Health Science, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong; Research Centre for Standardization of Chinese Medicines, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong
| | - Cai-Xia Dong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis and Metabolomics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China.
| | - Ai-Ping Lyu
- Institute of Systems Medicine and Health Science, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong.
| | - Jun Xu
- Institute of Systems Medicine and Health Science, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong; Research Centre for Standardization of Chinese Medicines, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong; Department of Pharmaceutical Analysis and Metabolomics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
3
|
Chen T, Wang N, Hao Y, Fu L. Fecal microbiota transplantation from postmenopausal osteoporosis human donors accelerated bone mass loss in mice. Front Cell Infect Microbiol 2024; 14:1488017. [PMID: 39703374 PMCID: PMC11655470 DOI: 10.3389/fcimb.2024.1488017] [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: 08/29/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
Objectives To investigate the effect of gut microbiota from postmenopausal osteoporosis patients on bone mass in mice. Methods Fecal samples were collected from postmenopausal women with normal bone mass (Con, n=5) and postmenopausal women with osteoporosis (Op, n=5). Microbial composition was identified by shallow shotgun sequencing. Then fecal samples were transplanted into pseudo-sterile mice previously treated with antibiotics for 4 weeks. These mice were categorized into two groups: the Vehicle group (n=7) received fecal samples from individuals with normal bone mass, and the FMT group (n=7) received fecal samples from individuals with osteoporosis. After 8 weeks, bone mass, intestinal microbial composition, intestinal permeability and inflammation were assessed, followed by a correlation analysis. Results The bone mass was significantly reduced in the FMT group. Microbiota sequencing showed that Shannon index (p < 0.05) and Simpson index (p < 0.05) were significantly increased in Op groups, and β diversity showed significant differences. the recipient mice were similar. linear discriminant analysis effect size (LEfSe) analysis of mice showed that Halobiforma, Enterorhabdus, Alistipes, and Butyricimonas were significantly enriched in the FMT group. Lachnospiraceae and Oscillibacter were significantly enriched in the Vehicle group. H&E staining of intestinal tissues showed obvious intestinal mucosal injury in mice. Intestinal immunohistochemistry showed that the expression of Claudin and ZO-1 in the intestinal tissue of the FMT group mice was decreased. The FITC-Dextran (FD-4) absorption rate and serum soluble CD14 (sCD14) content were increased in FMT mice. Correlation analysis showed that these dominant genera were significantly associated with bone metabolism and intestinal permeability, and were associated with the enrichment of specific enzymes. Serum and bone tissue inflammatory cytokines detection showed that the expression of TNF-α and IL-17A in the FMT group were significantly increased. Conclusion Overall, our findings suggested gut microbiota from postmenopausal osteoporosis patients accelerate bone mass loss in mice. Aberrant gut microbiota might play a causal role in the process of bone mass loss mediated by inflammation after the destruction of the intestinal barrier.
Collapse
Affiliation(s)
- Tinglong Chen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| | - Ning Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| | - Yongqiang Hao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| | - Lingjie Fu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| |
Collapse
|
4
|
Hu T, Zhu Y, Zhou X, Ye M, Wang X, Lu C, Wang Y. Baicalein ameliorates SEB-induced acute respiratory distress syndrome in a microbiota-dependent manner. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156049. [PMID: 39326141 DOI: 10.1016/j.phymed.2024.156049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/21/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is characterized by sudden and extensive pulmonary inflammation, with a mortality rate of approximately 40 %. Presently, there is no effective treatment to prevent or reverse its severe consequences. Baicalein (BAI) is a natural vicinal trihydroxyflavone and has been identified as the core quality marker of Scutellariae baicalensis for its effect on lung inflammation. However, its oral bioavailability is limited. The majority of studies that investigate BAI's in vivo mechanisms use injection techniques. Currently, there is no clear understanding of the mechanisms by which low-bioavailable BAI functions orally. PURPOSE This study aimed to evaluate the efficiency of BAI in ARDS mice and its underlying mechanisms. STUDY DESIGN AND METHODS Behavioral experiments, histological analysis, immunofluorescence staining, flow cytometry of immune cells, qRT-PCR, and ELISA analysis were performed to evaluate the efficiency of BAI in ARDS mice. Lung tissues transcriptomic-based analyses were performed to detect the differentially expressed genes and biological pathways. Fecal samples were subjected to microbial 16S rRNA analysis and untargeted metabolomics analysis in order to identify the specific flora and metabolites associated with BAI. Furthermore, antibiotic cocktail treatment and fecal microbiota transplantation were used to elucidate the gut microbiota-mediated effects on ARDS. RESULTS In our study, we first find that oral administration of BAI effectively mitigates staphylococcal enterotoxin B-induced ARDS. BAI can alleviate gut dysbiosis and regulate the Toll-like signaling pathway and amino acid metabolism. The protective effects of BAI against ARDS are gut microbiota dependent. Modulation of gut microbiota increases the production of short-chain fatty acids and enhances lung barrier function, which is consistent with the therapeutic interventions with BAI. Notably, BAI greatly enriches the abundance of Prevotellaceae, a butyrate-producing bacterial family, exhibiting a positive correlation with key differentially expressed genes in the TLR4/MyD88 signaling cascades. CONCLUSION BAI emerges as a potential prebiotic agent to attenuate ARDS, and targeting specific microbial species may offer an innovative therapeutic approach to investigate other flavonoids with limited bioavailability.
Collapse
Affiliation(s)
- Tingting Hu
- Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Ying Zhu
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341001, PR China
| | - Xiang Zhou
- Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Miaoyun Ye
- Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Xuecheng Wang
- Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Chen Lu
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341001, PR China
| | - Yaqi Wang
- Jiangxi University of Chinese Medicine, Nanchang 330004, PR China.
| |
Collapse
|
5
|
Munteanu C, Onose G, Rotariu M, Poștaru M, Turnea M, Galaction AI. Role of Microbiota-Derived Hydrogen Sulfide (H2S) in Modulating the Gut–Brain Axis: Implications for Alzheimer’s and Parkinson’s Disease Pathogenesis. Biomedicines 2024; 12:2670. [DOI: 10.3390/biomedicines12122670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Microbiota-derived hydrogen sulfide (H2S) plays a crucial role in modulating the gut–brain axis, with significant implications for neurodegenerative diseases such as Alzheimer’s and Parkinson’s. H2S is produced by sulfate-reducing bacteria in the gut and acts as a critical signaling molecule influencing brain health via various pathways, including regulating inflammation, oxidative stress, and immune responses. H2S maintains gut barrier integrity at physiological levels and prevents systemic inflammation, which could impact neuroinflammation. However, as H2S has a dual role or a Janus face, excessive H2S production, often resulting from gut dysbiosis, can compromise the intestinal barrier and exacerbate neurodegenerative processes by promoting neuroinflammation and glial cell dysfunction. This imbalance is linked to the early pathogenesis of Alzheimer’s and Parkinson’s diseases, where the overproduction of H2S exacerbates beta-amyloid deposition, tau hyperphosphorylation, and alpha-synuclein aggregation, driving neuroinflammatory responses and neuronal damage. Targeting gut microbiota to restore H2S homeostasis through dietary interventions, probiotics, prebiotics, and fecal microbiota transplantation presents a promising therapeutic approach. By rebalancing the microbiota-derived H2S, these strategies may mitigate neurodegeneration and offer novel treatments for Alzheimer’s and Parkinson’s diseases, underscoring the critical role of the gut–brain axis in maintaining central nervous system health.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania
| | - Mădălina Poștaru
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania
| |
Collapse
|
6
|
Wang T, Wu H, Shi X, Dai M, Liu Y. Aminoadipic acid aggravates atherosclerotic vascular inflammation through ROS/TXNIP/NLRP3 pathway, a harmful microbial metabolite reduced by paeonol. Int J Biochem Cell Biol 2024; 177:106678. [PMID: 39490917 DOI: 10.1016/j.biocel.2024.106678] [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: 07/15/2024] [Revised: 09/26/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
AIM Our previous study has found a differential microbial metabolite in atherosclerosis (AS) mice, aminoadipic acid (AAA), which was considered as a potential harmful metabolite. However, whether it can promote AS vascular inflammation and its mechanisms remain unclear. Paeonol (Pae) plays an anti-AS role by regulating the metabolic profile, but whether Pae exerts its antiatherogenic effect by reducing serum AAA levels is unknown. RESULTS The clinical trial results showed that the AS patients' serum AAA levels were higher than those healthy people'. Besides, AAA supplementation could increase aortic plaque size, serum inflammatory cytokines levels and liver malondialdehyde, superoxide dismutase levels in AS mice. Moreover, after AAA stimulation, the ROS levels and ASC, TXNIP, NLRP3 and caspase-1 proteins levels were increased in HUVECs, which could be reversed by antioxidant NAC and NLRP3 inhibitor. Pae significantly reduced the plaque size in the aorta, improved blood lipid levels and decreased serum inflammation factor levels in AS mice. Simultaneously, Pae could reduce the serum AAA levels of AS mice through the gut microbiota transmission. Finally, Pae inhibited NLRP3 inflammasome activation in aortas of AS mice. Broad-spectrum antibiotics could weaken the inhibitory effect of Pae on NLRP3 inflammasome. CONCLUSION Our study clarified that AAA could promote AS vascular inflammation via activating the ROS/TXNIP/NLRP3 pathway. Pae could inhibit AS development by reducing serum AAA levels in a microbiota-dependent manner. Taken together, we proposed that AAA could be served as a potential biomarker for AS clinical diagnosis and provided a new treatment strategy for AS.
Collapse
Affiliation(s)
- Tian Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China
| | - Hongfei Wu
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China
| | - Xiaoyan Shi
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China
| | - Min Dai
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China
| | - Yarong Liu
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei Anhui, 230012, China.
| |
Collapse
|
7
|
Zhang K, Zhang L, Jian Y, Tang X, Han M, Pu Z, Zhang Y, Zhou P. Early-Life Milk α S1-Casein Allergy Induces the Activation of Astrocytes in Mice and Leads to Stress Vulnerability in Adulthood. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23493-23510. [PMID: 39387175 DOI: 10.1021/acs.jafc.4c05425] [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: 10/12/2024]
Abstract
In recent years, the incidence of food allergies in children has been increasing annually, significantly affecting the quality of life for patients and their families. It has long been suspected that childhood allergies might potentially lead to behavioral and psychological issues in adulthood, but the specific connection remains unclear. In this study, we established a model of young mice allergic to milk αS1-casein, conducted behavioral tests, and employed transcriptomics, immunohistochemistry, Golgi staining, and fecal microbiota transplantation to explore the link between early life allergies and adult psychological problems. The results showed that early life milk protein allergy significantly increased intestinal epithelial permeability in mice, leading to the translocation of gut microbiota metabolites. This process subsequently activated astrocyte lysosomes via SLC15a3, making astrocytes more susceptible. This susceptibility caused mice with early life milk protein allergy to have more activated astrocytes and excessive dendritic spine phagocytosis (normal group: 5.4 ± 1.26 spines/10 μm, allergy group: 3.2 ± 0.92 spines/10 μm) under acute stress in adulthood, leading to anxiety and depressive behaviors.
Collapse
Affiliation(s)
- Kai Zhang
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lina Zhang
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yaqiong Jian
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xue Tang
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mengyu Han
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhiping Pu
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yiqian Zhang
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peng Zhou
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
8
|
Yuan Y, Li L, Wang J, Myagmar BO, Gao Y, Wang H, Wang Z, Zhang C, Zhang X. Gut microbiota-derived acetate promotes long-term recovery through angiogenesis guided by lymphatic ingrowth in older adults with stroke. Front Neurosci 2024; 18:1398913. [PMID: 39371609 PMCID: PMC11450648 DOI: 10.3389/fnins.2024.1398913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/20/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction Ischemic stroke is a leading cause of morbidity and mortality in older adults. Therefore, in this study, we sought to understand the interplay between the microbiota, gut, and brain in the context of stroke in older adults. Objective To determine whether gut microbiota from younger individuals promotes recovery through angiogenesis in both elderly stroke patients and aged stroke mice, we explored the changes in gut microbiota and the correlation between short-chain fatty acids (SCFAs) and angiogenesis in the aged stroke population. Then, we altered the gut microbiome in aged mice by transplanting microbiota from younger donors before inducing experimental stroke to explore the mechanism by which gut microbiota-derived SCFAs promote angiogenesis. Methods Part I: We conducted a single-center, double-blind trial to compare gut microbiota diversity and SCFA levels in fecal samples from older stroke patients with those from younger stroke patients. Additionally, we measured levels of vascular endothelial growth factor (VEGF) and VEGFC levels in plasma to assess their correlation with SCFA levels. Part II: We performed fecal microbiota transplantation (FMT) 3 days before inducing ischemic stroke in aged male mice (16-18) via distal middle cerebral artery occlusion (dMCAO). The FMT was conducted using gut microbiomes from either young donors (2-3 months) or aged donors (16-18 months). Results In older stroke patients, gut microbiota diversity was significantly reduced compared to that in younger stroke patients. Furthermore, levels of acetate, a bacterially derived SCFA, were lower and positively correlated with angiogenesis markers (VEGF and VEGF-C). In aged stroke mice, transplantation of young microbiota improved stroke outcomes by promoting angiogenesis, which was facilitated by lymphatic ingrowth into the cortex. This protective effect was linked to gut microbiota-derived acetate, which enhanced lymphangiogenesis by replenishing acetyl coenzyme A. Conclusions (a) Gut microbiota-derived acetate promotes angiogenesis post-stroke and (b) lymphatic ingrowth into the cerebral cortex was observed in post-dMCAO mice. These findings suggest that selectively promoting SCFA-producing bacteria, particularly acetate-producers, could be a promising therapeutic strategy to reduce functional impairments in older stroke subjects.
Collapse
Affiliation(s)
- Yujia Yuan
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Linlin Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jingjing Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bat-Otgon Myagmar
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuxiao Gao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huan Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhao Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
- Hebei Vascular Homeostasis Key Laboratory for Neurology, Shijiazhuang, Hebei, China
| |
Collapse
|
9
|
Zhai Z, Yang Y, Chen S, Wu Z. Long-Term Exposure to Polystyrene Microspheres and High-Fat Diet-Induced Obesity in Mice: Evaluating a Role for Microbiota Dysbiosis. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:97002. [PMID: 39226184 PMCID: PMC11370995 DOI: 10.1289/ehp13913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Microplastics (MPs) have become a global environmental problem, emerging as contaminants with potentially alarming consequences. However, long-term exposure to polystyrene microspheres (PS-MS) and its effects on diet-induced obesity are not yet fully understood. OBJECTIVES We aimed to investigate the effect of PS-MS exposure on high-fat diet (HFD)-induced obesity and underlying mechanisms. METHODS In the present study, C57BL/6J mice were fed a normal diet (ND) or a HFD in the absence or presence of PS-MS via oral administration for 8 wk. Antibiotic depletion of the microbiota and fecal microbiota transplantation (FMT) were performed to assess the influence of PS-MS on intestinal microbial ecology. We performed 16S rRNA sequencing to dissect microbial discrepancies and investigated the dysbiosis-associated intestinal integrity and inflammation in serum. RESULTS Compared with HFD mice, mice fed the HFD with PS-MS exhibited higher body weight, liver weight, metabolic dysfunction-associated steatotic liver disease (MASLD) activity scores, and mass of white adipose tissue, as well as higher blood glucose and serum lipid concentrations. Furthermore, 16S rRNA sequencing of the fecal microbiota revealed that mice fed the HFD with PS-MS had greater α -diversity and greater relative abundances of Lachnospiraceae, Oscillospiraceae, Bacteroidaceae, Akkermansiaceae, Marinifilaceae, Deferribacteres, and Desulfovibrio, but lower relative abundances of Atopobiaceae, Bifidobacterium, and Parabacteroides. Mice fed the HFD with PS-MS exhibited lower expression of MUC2 mucin and higher levels of lipopolysaccharide and inflammatory cytokines [tumor necrosis factor-α (TNF-α ), interleukin-6 (IL-6), IL-1β , and IL-17A] in serum. Correlation analyses revealed that differences in the microbial flora of mice exposed to PS-MS were associated with obesity. Interestingly, microbiota-depleted mice did not show the same PS-MS-associated differences in Muc2 and Tjp1 expression in the distal colon, expression of inflammatory cytokines in serum, or obesity outcomes between HFD and HFD + PS-MS. Importantly, transplantation of feces from HFD + PS-MS mice to microbiota-depleted HFD-fed mice resulted in a lower expression of mucus proteins, higher expression of inflammatory cytokines, and obesity outcomes, similar to the findings in HFD + PS-MS mice. CONCLUSIONS Our findings provide a new gut microbiota-driven mechanism for PS-MS-induced obesity in HFD-fed mice, suggesting the need to reevaluate the adverse health effects of MPs commonly found in daily life, particularly in susceptible populations. https://doi.org/10.1289/EHP13913.
Collapse
Affiliation(s)
- Zhian Zhai
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Food Science and Nutrition, Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Zhenlong Wu
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Hunt A, Drwiega E, Wang Y, Danziger L. A review of fecal microbiota, live-jslm for the prevention of recurrent Clostridioides difficile infection. Am J Health Syst Pharm 2024; 81:e402-e411. [PMID: 38470061 DOI: 10.1093/ajhp/zxae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Indexed: 03/13/2024] Open
Abstract
PURPOSE To review the composition, preparation, proposed mechanism of action, safety, efficacy, and current place in therapy of Rebyota (fecal microbiota, live-jslm). SUMMARY As the first agent in a new class of drugs, live biotherapeutic products (LBPs), fecal microbiota, live-jslm offers another therapeutic approach for the prevention of recurrent Clostridioides difficile infection (rCDI). LBPs are given following antibiotic therapy for C. difficile to reintroduce certain bacteria present in the normal microbiome, as a means to reconstitute the microbiome of infected individuals. This review provides a summary of phase 2 and 3 clinical trials, product information, discussion of data limitations, and recommendations for place in therapy. High efficacy rates compared to placebo with sustained response up to 24 months after administration have been reported. The majority of adverse events identified were mild to moderate without significant safety signals. CONCLUSION Fecal microbiota, live-jslm has consistently been shown in randomized trials to be safe and effective in reducing rCDI. Its approval marks the culmination of decades of work to identify, characterize, and refine the intestinal microbiome to create pharmaceutical products.
Collapse
Affiliation(s)
- Aaron Hunt
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| | - Emily Drwiega
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| | - Yifan Wang
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| | - Larry Danziger
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| |
Collapse
|
11
|
Kiełbik P, Witkowska-Piłaszewicz O. The Relationship between Canine Behavioral Disorders and Gut Microbiome and Future Therapeutic Perspectives. Animals (Basel) 2024; 14:2048. [PMID: 39061510 PMCID: PMC11273744 DOI: 10.3390/ani14142048] [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: 06/18/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Canine behavioral disorders have become one of the most common concerns and challenging issues among dog owners. Thus, there is a great demand for knowledge about various factors affecting dogs' emotions and well-being. Among them, the gut-brain axis seems to be particularly interesting, especially since in many instances the standard treatment or behavioral therapies insufficiently improve animal behavior. Therefore, to face this challenge, the search for novel therapeutic methods is highly required. Existing data show that mammals' gut microbiome, immune system, and nervous system are in continuous communication and influence animal physiology and behavior. This review aimed to summarize and discuss the most important scientific evidence on the relationship between mental disorders and gut microbiota in dogs, simultaneously presenting comparable outcomes in humans and rodent models. A comprehensive overview of crucial mechanisms of the gut-brain axis is included. This refers especially to the neurotransmitters crucial for animal behavior, which are regulated by the gut microbiome, and to the main microbial metabolites-short-chain fatty acids (SCFAs). This review presents summarized data on gut dysbiosis in relation to the inflammation process within the organism, as well as the activation of the hypothalamic-pituitary-adrenal (HPA) axis. All of the above mechanisms are presented in this review in strict correlation with brain and/or behavioral changes in the animal. Additionally, according to human and laboratory animal studies, the gut microbiome appears to be altered in individuals with mental disorders; thus, various strategies to manipulate the gut microbiota are implemented. This refers also to the fecal microbiome transplantation (FMT) method, based on transferring the fecal matter from a donor into the gastrointestinal tract of a recipient in order to modulate the gut microbiota. In this review, the possible effects of the FMT procedure on animal behavioral disorders are discussed.
Collapse
Affiliation(s)
- Paula Kiełbik
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | | |
Collapse
|
12
|
Missong H, Joshi R, Khullar N, Thareja S, Navik U, Bhatti GK, Bhatti JS. Nutrient-epigenome interactions: Implications for personalized nutrition against aging-associated diseases. J Nutr Biochem 2024; 127:109592. [PMID: 38325612 DOI: 10.1016/j.jnutbio.2024.109592] [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: 10/15/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Aging is a multifaceted process involving genetic and environmental interactions often resulting in epigenetic changes, potentially leading to aging-related diseases. Various strategies, like dietary interventions and calorie restrictions, have been employed to modify these epigenetic landscapes. A burgeoning field of interest focuses on the role of microbiota in human health, emphasizing system biology and computational approaches. These methods help decipher the intricate interplay between diet and gut microbiota, facilitating the creation of personalized nutrition strategies. In this review, we analysed the mechanisms related to nutritional interventions while highlighting the influence of dietary strategies, like calorie restriction and intermittent fasting, on microbial composition and function. We explore how gut microbiota affects the efficacy of interventions using tools like multi-omics data integration, network analysis, and machine learning. These tools enable us to pinpoint critical regulatory elements and generate individualized models for dietary responses. Lastly, we emphasize the need for a deeper comprehension of nutrient-epigenome interactions and the potential of personalized nutrition informed by individual genetic and epigenetic profiles. As knowledge and technology advance, dietary epigenetics stands on the cusp of reshaping our strategy against aging and related diseases.
Collapse
Affiliation(s)
- Hemi Missong
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Riya Joshi
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
13
|
Chen C, Xu JL, Gu ZC, Zhou SS, Wei GL, Gu JL, Ma HL, Feng YQ, Song ZW, Yan ZP, Deng S, Ding R, Li SL, Huo JG. Danggui Sini decoction alleviates oxaliplatin-induced peripheral neuropathy by regulating gut microbiota and potentially relieving neuroinflammation related metabolic disorder. Chin Med 2024; 19:58. [PMID: 38584284 PMCID: PMC10999090 DOI: 10.1186/s13020-024-00929-7] [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: 07/05/2023] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Danggui Sini decoction (DSD), a traditional Chinese medicine formula, has the function of nourishing blood, warming meridians, and unblocking collaterals. Our clinical and animal studies had shown that DSD can effectively protect against oxaliplatin (OXA)-induced peripheral neuropathy (OIPN), but the detailed mechanisms remain uncertain. Multiple studies have confirmed that gut microbiota plays a crucial role in the development of OIPN. In this study, the potential mechanism of protective effect of DSD against OIPN by regulating gut microbiota was investigated. METHODS The neuroprotective effects of DSD against OIPN were examined on a rat model of OIPN by determining mechanical allodynia, biological features of dorsal root ganglia (DRG) as well as proinflammatory indicators. Gut microbiota dysbiosis was characterized using 16S rDNA gene sequencing and metabolism disorders were evaluated using untargeted and targeted metabolomics. Moreover the gut microbiota mediated mechanisms were validated by antibiotic intervention and fecal microbiota transplantation. RESULTS DSD treatment significantly alleviated OIPN symptoms by relieving mechanical allodynia, preserving DRG integrity and reducing proinflammatory indicators lipopolysaccharide (LPS), IL-6 and TNF-α. Besides, DSD restored OXA induced intestinal barrier disruption, gut microbiota dysbiosis as well as systemic metabolic disorders. Correlation analysis revealed that DSD increased bacterial genera such as Faecalibaculum, Allobaculum, Dubosiella and Rhodospirillales_unclassified were closely associated with neuroinflammation related metabolites, including positively with short-chain fatty acids (SCFAs) and sphingomyelin (d18:1/16:0), and negatively with pi-methylimidazoleacetic acid, L-glutamine and homovanillic acid. Meanwhile, antibiotic intervention apparently relieved OIPN symptoms. Furthermore, fecal microbiota transplantation further confirmed the mediated effects of gut microbiota. CONCLUSION DSD alleviates OIPN by regulating gut microbiota and potentially relieving neuroinflammation related metabolic disorder.
Collapse
Affiliation(s)
- Chen Chen
- Department of Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
- Department of Oncology, Yancheng TCM Hospital, Yancheng, 224001, Jiangsu, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jian-Lin Xu
- Department of Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
- Department of Oncology, Yancheng TCM Hospital, Yancheng, 224001, Jiangsu, China
| | - Zhan-Cheng Gu
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, 215399, China
| | - Shan-Shan Zhou
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Guo-Li Wei
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, 211299, Jiangsu, China
| | - Jia-Lin Gu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hai-Long Ma
- Department of Paediatrics, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
| | - Yan-Qi Feng
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zi-Wei Song
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zhan-Peng Yan
- Clinical Research Department of Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Shan Deng
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Rong Ding
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| | - Jie-Ge Huo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| |
Collapse
|
14
|
Huang TQ, Chen YX, Zeng SL, Lin Y, Li F, Jiang ZM, Liu EH. Bergenin Alleviates Ulcerative Colitis By Decreasing Gut Commensal Bacteroides vulgatus-Mediated Elevated Branched-Chain Amino Acids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3606-3621. [PMID: 38324392 DOI: 10.1021/acs.jafc.3c09448] [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: 02/09/2024]
Abstract
Ulcerative colitis is closely associated with the dysregulation of gut microbiota. There is growing evidence that natural products may improve ulcerative colitis by regulating the gut microbiota. In this research, we demonstrated that bergenin, a naturally occurring isocoumarin, significantly ameliorates colitis symptoms in dextran sulfate sodium (DSS)-induced mice. Transcriptomic analysis and Caco-2 cell assays revealed that bergenin could ameliorate ulcerative colitis by inhibiting TLR4 and regulating NF-κB and mTOR phosphorylation. 16S rRNA sequencing and metabolomics analyses revealed that bergenin could improve gut microbiota dysbiosis by decreasing branched-chain amino acid (BCAA) levels. BCAA intervention mediated the mTOR/p70S6K signaling pathway to exacerbate the symptoms of ulcerative colitis in mice. Notably, bergenin greatly decreased the symbiotic bacteria Bacteroides vulgatus (B. vulgatus), and the gavage of B. vulgatus increased BCAA concentrations and aggravated the symptoms of ulcerative colitis in mice. Our findings suggest that gut microbiota-mediated BCAA metabolism plays a vital role in the protective effect of bergenin on ulcerative colitis, providing novel insights for ulcerative colitis prevention through manipulation of the gut microbiota.
Collapse
Affiliation(s)
- Tian-Qing Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yu-Xin Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Su-Ling Zeng
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Yang Lin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zheng-Meng Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
- College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China
| | - E-Hu Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
- College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
15
|
Pandey H, Jain D, Tang DWT, Wong SH, Lal D. Gut microbiota in pathophysiology, diagnosis, and therapeutics of inflammatory bowel disease. Intest Res 2024; 22:15-43. [PMID: 37935653 PMCID: PMC10850697 DOI: 10.5217/ir.2023.00080] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/23/2023] [Accepted: 08/27/2023] [Indexed: 11/09/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease, which is thought to be an interplay between genetic, environment, microbiota, and immune-mediated factors. Dysbiosis in the gut microbial composition, caused by antibiotics and diet, is closely related to the initiation and progression of IBD. Differences in gut microbiota composition between IBD patients and healthy individuals have been found, with reduced biodiversity of commensal microbes and colonization of opportunistic microbes in IBD patients. Gut microbiota can, therefore, potentially be used for diagnosing and prognosticating IBD, and predicting its treatment response. Currently, there are no curative therapies for IBD. Microbiota-based interventions, including probiotics, prebiotics, synbiotics, and fecal microbiota transplantation, have been recognized as promising therapeutic strategies. Clinical studies and studies done in animal models have provided sufficient evidence that microbiota-based interventions may improve inflammation, the remission rate, and microscopic aspects of IBD. Further studies are required to better understand the mechanisms of action of such interventions. This will help in enhancing their effectiveness and developing personalized therapies. The present review summarizes the relationship between gut microbiota and IBD immunopathogenesis. It also discusses the use of gut microbiota as a noninvasive biomarker and potential therapeutic option.
Collapse
Affiliation(s)
| | | | - Daryl W. T. Tang
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Sunny H. Wong
- Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Devi Lal
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| |
Collapse
|
16
|
Li Z, Zhang X, Wu H, Ma Z, Liu X, Ma J, Zhang D, Sheng L, Chen X, Zhang S. Hydrangea paniculata coumarins attenuate experimental membranous nephritis by bidirectional interactions with the gut microbiota. Commun Biol 2023; 6:1189. [PMID: 37993541 PMCID: PMC10665342 DOI: 10.1038/s42003-023-05581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023] Open
Abstract
Coumarins isolated from Hydrangea paniculata (HP) had a renal protective effect in experimental membranous nephritis (MN), but the mechanisms are not clear. Currently, we investigate whether the modulation of gut dysbiosis by HP contributes to its renal protection. Experimental MN rats were treated with HP for six weeks. Fecal 16S rDNA sequencing and metabolomics were performed. Fecal microbiota transplantation (FMT) was used for the evaluation study. The results demonstrate that deteriorated renal function and gut dysbiosis are found in MN rats, as manifested by a higher Firmicutes/Bacteroidetes ratio and reduced diversity and richness, but both changes were reversed by HP treatment. Reduced gut dysbiosis is correlated with improved colonic integrity and lower endotoxemia in HP-treated rats. HP normalized the abnormal level of fecal metabolites by increasing short-chain fatty acid production and hindering the production of uremic toxin precursors. FMT of HP-treated feces to MN animals moderately reduced endotoxemia and albuminuria. Moreover, major coumarins in HP were only biotransformed into more bioactive 7-hydroxycoumarin by gut microbiota, which strengthened the effect of HP in vivo. Depletion of the gut microbiota partially abolished its renal protective effect. In conclusion, the bidirectional interaction between HP and the gut microbiota contributes to its beneficial effect.
Collapse
Affiliation(s)
- Zhaojun Li
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Xingguang Zhang
- Department of Endocrinology, The seventh medical center of Chinese PLA General Hospital, Beijing, 100070, China
| | - Haijie Wu
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Zhiling Ma
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Xikun Liu
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Jie Ma
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Dongming Zhang
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Li Sheng
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China.
| | - Xiaoguang Chen
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China.
| | - Sen Zhang
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China.
| |
Collapse
|
17
|
Diaz-Marugan L, Kantsjö JB, Rutsch A, Ronchi F. Microbiota, diet, and the gut-brain axis in multiple sclerosis and stroke. Eur J Immunol 2023; 53:e2250229. [PMID: 37470461 DOI: 10.1002/eji.202250229] [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: 02/04/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/21/2023]
Abstract
Intestinal microbiota can influence the phenotype and function of immune cell responses through the dissemination of bacterial antigens or metabolites. Diet is one of the major forces shaping the microbiota composition and metabolism, contributing to host homeostasis and disease susceptibility. Currently, nutrition is a complementary and alternative approach to the management of metabolic and neurological diseases and cancer. However, the knowledge of the exact mechanism of action of diet and microbiota on the gut-brain communication is only developing in recent years. Here, we reviewed the current knowledge on the effect of diet and microbiota on the gut-brain axis in patients with two different central nervous system diseases, multiple sclerosis and stroke. We have also highlighted the open questions in the field that we believe are important to address to gain a deeper understanding of the mechanisms by which diet can directly or indirectly affect the host via the microbiota. We think this will open up new approaches to the treatment, diagnosis, and monitoring of various diseases.
Collapse
Affiliation(s)
- Laura Diaz-Marugan
- Charité - Universitaetsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH): Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Berlin, Germany
- Departamento de Medicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Johan B Kantsjö
- Charité - Universitaetsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH): Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Berlin, Germany
| | - Andrina Rutsch
- Charité - Universitaetsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH): Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Berlin, Germany
| | - Francesca Ronchi
- Charité - Universitaetsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH): Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Berlin, Germany
| |
Collapse
|
18
|
Marsool MDM, Vora N, Marsool ADM, Pati S, Narreddy M, Patel P, Gadam S, Prajjwal P. Ulcerative colitis: Addressing the manifestations, the role of fecal microbiota transplantation as a novel treatment option and other therapeutic updates. Dis Mon 2023; 69:101606. [PMID: 37357103 DOI: 10.1016/j.disamonth.2023.101606] [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] [Indexed: 06/27/2023]
Abstract
The prevalence and incidence of Ulcerative Colitis (UC), a recurrent and remitting inflammatory condition, are rising. Any part of the colon may be affected, beginning with inflammation of the mucosa in the rectum and continuing proximally continuously. Bloody diarrhea, tenesmus, fecal urgency, and stomach pain are typical presenting symptoms. Many patients present with extraintestinal manifestations (EIMs) including musculoskeletal, ocular, renal, hepatobiliary, and dermatological presentation, among others. Most cases are treated with pharmacological therapy including mesalazine and glucocorticoids. Fecal microbiota transplantation (FMT) is a novel procedure that is increasingly being used to treat UC, however, its use yet remains controversial because of uncertain efficacy. FMT can lower gut permeability and consequently disease severity by boosting short-chain fatty acids production, helping in epithelial barrier integrity preservation. Upadacitinib (JAK Kinase inhibitor) is another newer treatment option, which is an FDA-approved drug that is being used to treat UC. This review article provides a comprehensive review of the EIMs of UC, the role of FMT along with various recent clinical trials pertaining to FMT as well as other diagnostic and therapeutic updates.
Collapse
Affiliation(s)
| | - Neel Vora
- B. J. Medical College, Ahmedabad, India
| | | | - Shefali Pati
- St George's University, School of Medicine, Grenada
| | | | - Parth Patel
- Pramukhswami Medical College, Karamsad, India
| | | | | |
Collapse
|
19
|
Guo B, Zhang J, Zhang W, Chen F, Liu B. Gut microbiota-derived short chain fatty acids act as mediators of the gut-brain axis targeting age-related neurodegenerative disorders: a narrative review. Crit Rev Food Sci Nutr 2023; 65:265-286. [PMID: 37897083 DOI: 10.1080/10408398.2023.2272769] [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] [Indexed: 10/29/2023]
Abstract
Neurodegenerative diseases associated with aging are often accompanied by cognitive decline and gut microbiota disorder. But the impact of gut microbiota on these cognitive disturbances remains incompletely understood. Short chain fatty acids (SCFAs) are major metabolites produced by gut microbiota during the digestion of dietary fiber, serving as an energy source for gut epithelial cells and/or circulating to other organs, such as the liver and brain, through the bloodstream. SCFAs have been shown to cross the blood-brain barrier and played crucial roles in brain metabolism, with potential implications in mediating Alzheimer's disease (AD) and Parkinson's disease (PD). However, the underlying mechanisms that SCFAs might influence psychological functioning, including affective and cognitive processes and their neural basis, have not been fully elucidated. Furthermore, the dietary sources which determine these SCFAs production was not thoroughly evaluated yet. This comprehensive review explores the production of SCFAs by gut microbiota, their transportation through the gut-brain axis, and the potential mechanisms by which they influence age-related neurodegenerative disorders. Also, the review discusses the importance of dietary fiber sources and the challenges associated with harnessing dietary-derived SCFAs as promoters of neurological health in elderly individuals. Overall, this study suggests that gut microbiota-derived SCFAs and/or dietary fibers hold promise as potential targets and strategies for addressing age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Bingbing Guo
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jingyi Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Weihao Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Feng Chen
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| | - Bin Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
20
|
Zhou Y, Jia Y, Xu N, Tang L, Chang Y. Auricularia auricula-judae (Bull.) polysaccharides improve obesity in mice by regulating gut microbiota and TLR4/JNK signaling pathway. Int J Biol Macromol 2023; 250:126172. [PMID: 37558018 DOI: 10.1016/j.ijbiomac.2023.126172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/20/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Obesity has emerged as a crucial factor impacting people's lives, and gut microbiota disorders contribute to its development and progression. Auricularia auricula-judae (Bull.) polysaccharides (AAPs), a traditional functional food in Asia, exhibit potential anti-obesity effects. However, the specific mechanism still needs to be further confirmed. This study investigated the beneficial effects and specific mechanisms of AAPs on obesity. Firstly, AAPs showed significant improvements in overweight, insulin resistance, glucose and lipid metabolism disorders, and liver damage in obese mice. Additionally, AAPs ameliorated gut microbiota disorders, promoting the proliferation of beneficial bacteria like Lactobacillus and Roseburia, resulting in increased levels of SCFAs, folate, and cobalamin. Simultaneously, AAPs inhibited the growth of harmful bacteria, thereby protecting intestinal barrier function, improving endotoxemia, and decreasing the levels of inflammatory factors such as TNF-α and IL-6. Furthermore, AAPs can inhibit the TLR4/JNK signaling pathway while promoting the activation of AKT and AMPK. Importantly, our study underscored the pivotal role of gut microbiota in the anti-obesity effects of AAPs, as evidenced by fecal microbiota transplantation experiments. In conclusion, our findings elucidated that AAPs improve obesity by regulating gut microbiota and TLR4/JNK signaling pathway, offering novel perspectives for further conclusion the anti-obesity potential of AAPs.
Collapse
Affiliation(s)
- Yingjun Zhou
- East China University of Science and Technology, People's Republic of China
| | - Yuezhong Jia
- East China University of Science and Technology, People's Republic of China
| | - Nuo Xu
- East China University of Science and Technology, People's Republic of China
| | - Lihua Tang
- East China University of Science and Technology, People's Republic of China
| | - Yaning Chang
- East China University of Science and Technology, People's Republic of China.
| |
Collapse
|
21
|
Liu Y, Wu H, Wang T, Shi X, He H, Huang H, Yang Y, Dai M. Paeonol reduces microbial metabolite α-hydroxyisobutyric acid to alleviate the ROS/TXNIP/NLRP3 pathway-mediated endothelial inflammation in atherosclerosis mice. Chin J Nat Med 2023; 21:759-774. [PMID: 37879794 DOI: 10.1016/s1875-5364(23)60506-0] [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: 08/12/2023] [Indexed: 10/27/2023]
Abstract
Gut microbiota dysbiosis is an avenue for the promotion of atherosclerosis (AS) and this effect is mediated partly via the circulating microbial metabolites. More microbial metabolites related to AS vascular inflammation, and the mechanisms involved need to be clarified urgently. Paeonol (Pae) is an active compound isolated from Paeonia suffruticoas Andr. with anti-AS inflammation effect. However, considering the low oral bioavailability of Pae, it is worth exploring the mechanism by which Pae reduces the harmful metabolites of the gut microbiota to alleviate AS. In this study, ApoE-/- mice were fed a high-fat diet (HFD) to establish an AS model. AS mice were administrated with Pae (200 or 400 mg·kg-1) by oral gavage and fecal microbiota transplantation (FMT) was conducted. 16S rDNA sequencing was performed to investigate the composition of the gut microbiota, while metabolomics analysis was used to identify the metabolites in serum and cecal contents. The results indicated that Pae significantly improved AS by regulating gut microbiota composition and microbiota metabolic profile in AS mice. We also identified α-hydroxyisobutyric acid (HIBA) as a harmful microbial metabolite reduced by Pae. HIBA supplementation in drinking water promoted AS inflammation in AS mice. Furthermore, vascular endothelial cells (VECs) were cultured and stimulated by HIBA. We verified that HIBA stimulation increased intracellular ROS levels, thereby inducing VEC inflammation via the TXNIP/NLRP3 pathway. In sum, Pae reduces the production of the microbial metabolite HIBA, thus alleviating the ROS/TXNIP/NLRP3 pathway-mediated endothelial inflammation in AS. Our study innovatively confirms the mechanism by which Pae reduces the harmful metabolites of gut microbiota to alleviate AS and proposes HIBA as a potential biomarker for AS clinical judgment.
Collapse
Affiliation(s)
- Yarong Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei 230012, China
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei 230012, China
| | - Tian Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xiaoyan Shi
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Hai He
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Hanwen Huang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yulong Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Min Dai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
22
|
Boicean A, Bratu D, Fleaca SR, Vasile G, Shelly L, Birsan S, Bacila C, Hasegan A. Exploring the Potential of Fecal Microbiota Transplantation as a Therapy in Tuberculosis and Inflammatory Bowel Disease. Pathogens 2023; 12:1149. [PMID: 37764957 PMCID: PMC10535282 DOI: 10.3390/pathogens12091149] [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: 07/29/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
This review explores the potential benefits of fecal microbiota transplantation (FMT) as an adjunct treatment in tuberculosis (TB), drawing parallels from its efficacy in inflammatory bowel disease (IBD). FMT has shown promise in restoring the gut microbial balance and modulating immune responses in IBD patients. Considering the similarities in immunomodulation and dysbiosis between IBD and TB, this review hypothesizes that FMT may offer therapeutic benefits as an adjunct therapy in TB. Methods: We conducted a systematic review of the existing literature on FMT in IBD and TB, highlighting the mechanisms and potential implications of FMT in the therapeutic management of both conditions. The findings contribute to understanding FMT's potential role in TB treatment and underscore the necessity for future research in this direction to fully leverage its clinical applications. Conclusion: The integration of FMT into the comprehensive management of TB could potentially enhance treatment outcomes, reduce drug resistance, and mitigate the side effects of conventional therapies. Future research endeavors should focus on well-designed clinical trials to develop guidelines concerning the safety and short- and long-term benefits of FMT in TB patients, as well as to assess potential risks.
Collapse
Affiliation(s)
- Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Dan Bratu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Sorin Radu Fleaca
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Gligor Vasile
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (G.V.); (L.S.)
| | - Leeb Shelly
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (G.V.); (L.S.)
| | - Sabrina Birsan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Ciprian Bacila
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Adrian Hasegan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| |
Collapse
|
23
|
Palandurkar GS, Kumar S. Biofilm's Impact on Inflammatory Bowel Diseases. Cureus 2023; 15:e45510. [PMID: 37868553 PMCID: PMC10585119 DOI: 10.7759/cureus.45510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
The colon has a large surface area covered with a thick mucus coating. Colon's biomass consists of about 1,012 colony-forming units per gram of feces and 500-1,000 distinct bacterial species. The term inflammatory bowel disease (IBD) indicates the collection of intestinal illnesses in which the digestive system (esophagus, large intestine, mouth, stomach, and small intestine) experiences persistent inflammation. IBD development is influenced by environmental (infections, stress, and nutrition) and genetic factors. The microbes present in gut microbiota help maintain intestinal homeostasis and support immune and epithelial cell growth, differentiation, as well as proliferation. It has been discovered that a variety of variables and microorganisms are crucial for the development of biofilms and mucosal colonization during IBD. An extracellular matrix formed by bacteria supports biofilm production in our digestive system and harms the host's immunological response. Irritable bowel syndrome (IBS) and IBD considerably affect human socioeconomic well-being and the standard of living. IBD is a serious public health issue, affecting millions of people across the globe. The gut microbiome may significantly influence IBS pathogenesis, even though few diagnostic and treatment options are available. As a result, current research focuses more on disrupting biofilm in IBD patients and stresses primarily on drugs that help improve the quality of life for human well-being. We evaluate studies on IBD and bacterial biofilm to add fresh insights into the existing state of knowledge of biofilm formation in IBD, incidence of IBD patients, molecular level of investigations, bacteria that are involved in the formation of biofilm, and present and down the line regimens and probiotics. Planning advanced ways to control and eradicate bacteria in biofilms should be the primary goal to add fresh insights into generating innovative diagnostic and alternative therapy options for IBD.
Collapse
Affiliation(s)
- Gopal S Palandurkar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunil Kumar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
24
|
Rossier L, Matter C, Burri E, Galperine T, Hrúz P, Juillerat P, Schoepfer A, Vavricka SR, Zahnd N, Décosterd N, Seibold F. Swiss expert opinion: current approaches in faecal microbiota transplantation in daily practice. Swiss Med Wkly 2023; 153:40100. [PMID: 37769622 DOI: 10.57187/smw.2023.40100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
INTRODUCTION Faecal microbiota transplantation (FMT) is an established therapy for recurrent C. difficile infection, and recent studies have reported encouraging results of FMT in patients with ulcerative colitis. Few international consensus guidelines exist for this therapy, and thus FMT policies and practices differ among European countries. As of 2019, stool transplants are considered a non-standardised medicinal product in Switzerland, and a standardised production process requires authorisation by the Swiss Agency for Therapeutic Products. This authorisation leads to prolonged administrative procedures and increasing costs, which reduces treatment accessibility. In particular, patients with ulcerative colitis in Switzerland can only benefit from FMT off-label, even though it is a valid therapeutic option. Therefore, this study summarised the available data on FMT and established a framework for the standardised use of FMT. METHODS A panel of Swiss gastroenterologists with a special interest in inflammatory bowel disease was established to identify the current key issues of FMT. After a comprehensive review of the literature, statements were formulated about FMT indications, donor screening, stool transplant preparation and administration, and safety aspects. The panel then voted on the statements following the Delphi process; the statements were reformulated and revoted until a consensus was reached. The manuscript was then reviewed by an infectiologist (the head of Lausanne's FMT centre). RESULTS The established statements are summarised in the supplementary tables in the appendix to this paper. The working group hopes these will help standardise FMT practice in Switzerland and contribute to making faecal microbiota transplantation a safe and accessible treatment for patients with recurrent C. difficile infections and selected patients with ulcerative colitis, as well as other indications in the future.
Collapse
Affiliation(s)
- Laura Rossier
- Intesto - Gastroenterology practice and Crohn-colitis Center, Bern, Switzerland
| | - Christoph Matter
- Intesto - Gastroenterology practice and Crohn-colitis Center, Bern, Switzerland
| | - Emanuel Burri
- Department of Gastroenterology and Hepatology, University Medical Clinic, Baselland Canton Hospital, Liestal, Switzerland
| | - Tatiana Galperine
- Fecal microbiota transplantation center, Department of infectious disease, Lausanne University Hospital, Lausanne, Switzerland
| | - Petr Hrúz
- Clarunis, Department of Gastroenterology, St Clara hospital and University hospital Basel, Basel, Switzerland
| | - Pascal Juillerat
- GastroGeb - Gastroenterology practice and Crohn-colitis Center, Lausanne - Bulle, Switzerland
| | - Alain Schoepfer
- Department of Gastroenterology, Lausanne University Hospital, Lausanne, Switzerland
| | - Stephan R Vavricka
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Natalie Décosterd
- Intesto - Gastroenterology practice and Crohn-colitis Center, Bern, Switzerland
| | - Frank Seibold
- Intesto - Gastroenterology practice and Crohn-colitis Center, Bern, Switzerland
| |
Collapse
|
25
|
Boicean A, Bratu D, Bacila C, Tanasescu C, Fleacă RS, Mohor CI, Comaniciu A, Băluță T, Roman MD, Chicea R, Cristian AN, Hasegan A, Birsan S, Dura H, Mohor CI. Therapeutic Perspectives for Microbiota Transplantation in Digestive Diseases and Neoplasia-A Literature Review. Pathogens 2023; 12:766. [PMID: 37375456 PMCID: PMC10302701 DOI: 10.3390/pathogens12060766] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
In a mutually beneficial connection with its host, the gut microbiota affects the host's nutrition, immunity, and metabolism. An increasing number of studies have shown links between certain types of disease and gut dysbiosis or specific microorganisms. Fecal microbiota transplantation (FMT) is strongly advised for the treatment of recurrent or resistant Clostridium difficile infection (CDI) due to its outstanding clinical effectiveness against CDI. The therapeutic potential of FMT for other disorders, particularly inflammatory bowel diseases and malignancies, is currently gaining more and more attention. We summarized the most recent preclinical and clinical evidence to show the promise of FMT in the management of cancer as well as complications related to cancer treatment after reviewing the most recent research on the gut microbiota and its relationship to cancer.
Collapse
Affiliation(s)
- Adrian Boicean
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Dan Bratu
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Ciprian Bacila
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
| | - Ciprian Tanasescu
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Radu Sorin Fleacă
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Calin Ilie Mohor
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Andra Comaniciu
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
| | - Teodora Băluță
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
| | - Mihai Dan Roman
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Radu Chicea
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Nicolae Cristian
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Hasegan
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Sabrina Birsan
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Horațiu Dura
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Cosmin Ioan Mohor
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| |
Collapse
|
26
|
Dallas JW, Warne RW. Captivity and Animal Microbiomes: Potential Roles of Microbiota for Influencing Animal Conservation. MICROBIAL ECOLOGY 2023; 85:820-838. [PMID: 35316343 DOI: 10.1007/s00248-022-01991-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/07/2022] [Indexed: 05/04/2023]
Abstract
During the ongoing biodiversity crisis, captive conservation and breeding programs offer a refuge for species to persist and provide source populations for reintroduction efforts. Unfortunately, captive animals are at a higher disease risk and reintroduction efforts remain largely unsuccessful. One potential factor in these outcomes is the host microbiota which includes a large diversity and abundance of bacteria, fungi, and viruses that play an essential role in host physiology. Relative to wild populations, the generalized pattern of gut and skin microbiomes in captivity are reduced alpha diversity and they exhibit a significant shift in community composition and/or structure which often correlates with various physiological maladies. Many conditions of captivity (antibiotic exposure, altered diet composition, homogenous environment, increased stress, and altered intraspecific interactions) likely lead to changes in the host-associated microbiome. To minimize the problems arising from captivity, efforts can be taken to manipulate microbial diversity and composition to be comparable with wild populations through methods such as increasing dietary diversity, exposure to natural environmental reservoirs, or probiotics. For individuals destined for reintroduction, these strategies can prime the microbiota to buffer against novel pathogens and changes in diet and improve reintroduction success. The microbiome is a critical component of animal physiology and its role in species conservation should be expanded and included in the repertoire of future management practices.
Collapse
Affiliation(s)
- Jason W Dallas
- Department of Biological Sciences, Southern Illinois University, 1125 Lincoln Drive, Carbondale, IL, 62901, USA.
| | - Robin W Warne
- Department of Biological Sciences, Southern Illinois University, 1125 Lincoln Drive, Carbondale, IL, 62901, USA
| |
Collapse
|
27
|
Boicean A, Birlutiu V, Ichim C, Anderco P, Birsan S. Fecal Microbiota Transplantation in Inflammatory Bowel Disease. Biomedicines 2023; 11:biomedicines11041016. [PMID: 37189634 DOI: 10.3390/biomedicines11041016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Inflammatory bowel diseases represent a complex array of diseases of incompletely known etiology that led to gastrointestinal tract chronic inflammation. In inflammatory bowel disease, a promising method of treatment is represented by fecal microbiota transplantation (FMT), FMT has shown its increasing effectiveness and safety in recent years for recurrent CDI; moreover, it showed real clinical benefits in treating SARS-CoV-2 and CDI co-infection. Crohn’s disease and ulcerative colitis are characterized by immune dysregulation, resulting in digestive tract damage caused by immune responses. Most current therapeutic strategies are associated with high costs and many adverse effects by directly targeting the immune response, so modifying the microbial environment by FMT offers an alternative approach that could indirectly influence the host’s immune system in a safe way. Studies outline the endoscopic and clinical improvements in UC and CD in FMT patients versus control groups. This review outlines the multiple benefits of FMT in the case of IBD by improving patients unbalanced gut, therefore improving endoscopic and clinical symptomatology. We aim to emphasize the clinical importance and benefits of FMT in order to prevent flares or complications of IBD and to highlight that further validation is needed for establishing a clinical protocol for FMT in IBD.
Collapse
|
28
|
Liu YY. Controlling the human microbiome. Cell Syst 2023; 14:135-159. [PMID: 36796332 PMCID: PMC9942095 DOI: 10.1016/j.cels.2022.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/18/2022] [Accepted: 12/21/2022] [Indexed: 02/17/2023]
Abstract
We coexist with a vast number of microbes that live in and on our bodies. Those microbes and their genes are collectively known as the human microbiome, which plays important roles in human physiology and diseases. We have acquired extensive knowledge of the organismal compositions and metabolic functions of the human microbiome. However, the ultimate proof of our understanding of the human microbiome is reflected in our ability to manipulate it for health benefits. To facilitate the rational design of microbiome-based therapies, there are many fundamental questions to be addressed at the systems level. Indeed, we need a deep understanding of the ecological dynamics associated with such a complex ecosystem before we rationally design control strategies. In light of this, this review discusses progress from various fields, e.g., community ecology, network science, and control theory, that are helping us make progress toward the ultimate goal of controlling the human microbiome.
Collapse
Affiliation(s)
- Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| |
Collapse
|
29
|
Wang Z, Yao W, Sun Y, Han Y, Chen X, Gong P, Zhai P, Pei S, Xie J, Ba Q, Wang H. Eucommia Bark/Leaf Extract Improves Lipid Metabolism Disorders by Affecting Intestinal Microbiota and Microbiome-Host Interaction in HFD Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3297-3314. [PMID: 36753681 DOI: 10.1021/acs.jafc.2c07239] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Eucommia bark contains many bioactive compounds and has anti-hyperlipidemic effects. However, due to the slow growth rate of the plant, there is a limited supply of this resource. Studies have demonstrated that Eucommia leaves contain active ingredients similar to those of Eucommia bark and also have anti-hyperlipidemic effects. It is not currently clear whether Eucommia leaf can be used as a substitute for Eucommia bark. Furthermore, their mechanism of action for anti-hyperlipidemia by improving the structure of the gut microbiota is also unclear. We aimed to determine the composition of the active ingredients in EBE and ELE by HPLC, establish an HFD-induced hyperlipidemia model, and combine fecal microbiota transplantation (FMT) experiments to investigate the mechanism of EBE/ELE anti-hyperlipidemia by modifying the structure of intestinal microbiota, as well as to compare the effects of EBE and ELE. Our results showed that EBE and ELE contained similar active ingredients and significantly alleviated lipid metabolism disorders and blood glucose levels in the HFD-induced hyperlipidemia model. In this study, EBE and ELE significantly reduced the relative abundance of Desulfovibrionaceae and Erysipelotrichaceae and significantly increased the relative abundance of Ruminococcaceae. They also promoted the production of short-chain fatty acids (SCFAs) and activated the gene expression of the SCFA receptors G protein-coupled receptor 41 (GPR41) and GPR43. In addition, EBE and ELE can significantly increase the expression of the fasting-induced adipose factor (Fiaf) gene in the colon and inhibit the secretion of lipoprotein lipase (LPL) in the liver, thereby inhibiting triglyceride (TG) synthesis. They also significantly activate the expression of GPR41 and GPR43 genes in the epididymal fat tissue, leading to reduced lipid accumulation in adipocytes. These effects on the target genes were associated with changes in the abundance of Desulfovibrionaceae, Erysipelotrichaceae, and Ruminococcaceae bacteria in the intestinal microbiota. Thus, regulating the relative abundance of these microbes may serve as prospective targets for EBE/ELE to influence the Fiaf-LPL gut-liver axis and the SCFAs-GPR41/GPR43 gut-fat axis. In addition, there was no significant difference in the anti-hyperlipidemic effects of ELE and EBE, suggesting that Eucommia leaf may be a suitable alternative to Eucommia bark for managing hyperlipidemia by regulating the structure of the intestinal microbiota. These findings suggest that Eucommia leaves have great potential for development as a functional food with lipid-lowering properties.
Collapse
Affiliation(s)
- Zhineng Wang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Wenbo Yao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Ying Sun
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Yewen Han
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Xuefeng Chen
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Pin Gong
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Pengtao Zhai
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Shuya Pei
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Jianwu Xie
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Qian Ba
- Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Hui Wang
- Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
30
|
Wang Z, Sun Y, Han Y, Chen X, Gong P, Zhai P, Yao W, Ba Q, Wang H. Eucommia bark/leaf extract improves HFD-induced lipid metabolism disorders via targeting gut microbiota to activate the Fiaf-LPL gut-liver axis and SCFAs-GPR43 gut-fat axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154652. [PMID: 36638713 DOI: 10.1016/j.phymed.2023.154652] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/25/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND The bark of Eucommia ulmoides (a perennial deciduous tree termed eucommia hereafter) has anti-hyperlipidemia effects due to its bioactive components. However, the slow growth of eucommia bark leads to a deficit in this resource. Studies have shown that eucommia leaf has bioactive components similar to those of eucommia bark and anti-hyperlipidemia effects. At present, the strength of the anti-hyperlipidemia effect of eucommia bark and eucommia leaf has not been reported. Their interaction with the gut microbiota and the mechanism by which the gut microbiota exerts anti-hyperlipidemia effects are unclear. PURPOSES Through fecal microbiota transplantation (FMT) experiments, this study aimed to investigate the mechanism by which fecal bacteria suspensions containing chlorogenic acid (CGA), eucommia bark extract (EBE), and eucommia leaves extract (ELE) improve high-fat diet (HFD)-induced lipid metabolism disorders. Difference in anti-hyperlipidemia effects between EBE and ELE and exploring an eucommia bark substitute to improve the sustainable utilization of eucommia were also evaluated. RESULTS EBE and ELE contain eight identical bioactive ingredients, and fecal bacteria suspensions containing EBE and ELE significantly improved HFD-induced lipid metabolism disorders and elevated blood glucose levels. The fecal bacteria suspension of healthy mice containing CGA, EBE, and ELE significantly reduced the relative abundance of Erysipelothrichaceae and Ruminococcaceae and promoted short chain fatty acids (SCFAs) production thereby activating the expression of the SCFA. G protein-coupled receptor 43 (GPR43) gene in colon and epididymal fat tissues. In addition, fecal bacteria suspensions of healthy mice containing CGA, EBE, or ELE significantly activated fasting-induced adipose factor (Fiaf) gene expression in colon tissue and inhibited the secretion of lipoprotein lipase (LPL) in liver tissue, thereby inhibiting the synthesis of triglycerides (TG). Changed in the Erysipelotrichaceae and Ruminococcaceae relative abundances were significantly correlated with these target genes. Thus, regulating the abundance of the Erysipelotrichaceae and Ruminococcaceae could serve as a potential target for the role of fecal bacteria suspensions of healthy mice containing CGA, EBE, or ELE in the Fiaf-LPL gut-liver axis and SCFAs-GPR43 gut-fat axis. In addition, regarding HFD-induced lipid metabolism disorders and gut microbiota structural disorders, we found no significant difference between ELE and EBE. CONCLUSIONS Our FMT experiments evidenced that EBE and ELE improve lipid metabolism disorders by regulating the gut microbiota, providing a new pathway for treating hyperlipidemia using eucommia dietary therapy. There was no significant difference in the anti-hyperlipidemia effects of ELE and EBE; thus, eucommia leaf could replace eucommia bark in traditional Chinese medicine, so as to achieve a sustainable utilization of eucommia resources.
Collapse
Affiliation(s)
- Zhineng Wang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Yin Sun
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Yewen Han
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Xuefeng Chen
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Pin Gong
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Pengtao Zhai
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Wenbo Yao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China.
| | - Qian Ba
- Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Experimental Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Hui Wang
- Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Recent Approaches for Downplaying Antibiotic Resistance: Molecular Mechanisms. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5250040. [PMID: 36726844 PMCID: PMC9886476 DOI: 10.1155/2023/5250040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023]
Abstract
Antimicrobial resistance (AMR) is a ubiquitous public health menace. AMR emergence causes complications in treating infections contributing to an upsurge in the mortality rate. The epidemic of AMR in sync with a high utilization rate of antimicrobial drugs signifies an alarming situation for the fleet recovery of both animals and humans. The emergence of resistant species calls for new treatments and therapeutics. Current records propose that health drug dependency, veterinary medicine, agricultural application, and vaccination reluctance are the primary etymology of AMR gene emergence and spread. Recently, several encouraging avenues have been presented to contest resistance, such as antivirulent therapy, passive immunization, antimicrobial peptides, vaccines, phage therapy, and botanical and liposomal nanoparticles. Most of these therapies are used as cutting-edge methodologies to downplay antibacterial drugs to subdue the resistance pressure, which is a featured motive of discussion in this review article. AMR can fade away through the potential use of current cutting-edge therapeutics, advancement in antimicrobial susceptibility testing, new diagnostic testing, prompt clinical response, and probing of new pharmacodynamic properties of antimicrobials. It also needs to promote future research on contemporary methods to maintain host homeostasis after infections caused by AMR. Referable to the microbial ability to break resistance, there is a great ultimatum for using not only appropriate and advanced antimicrobial drugs but also other neoteric diverse cutting-edge therapeutics.
Collapse
|
32
|
Bachour SP, Dalal R, Allegretti JR. The impact of the COVID-19 pandemic on Clostridioides difficile infection and utilization of fecal microbiota transplantation. Therap Adv Gastroenterol 2023; 16:17562848231165581. [PMID: 37091531 PMCID: PMC10107020 DOI: 10.1177/17562848231165581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/06/2023] [Indexed: 04/25/2023] Open
Abstract
Previous research has demonstrated that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) gains cell entry through the angiotensin-converting enzyme 2 receptor, which is abundantly found throughout the gastrointestinal (GI) tract, resulting in a wide array of GI manifestations of coronavirus disease 2019 (COVID-19). By gaining entry into the intestinal epithelial and stromal cells, SARS-CoV-2 has been observed to cause intestinal inflammation and gut dysbiosis. Alterations in gut microbiota are known to be involved in the pathophysiology of Clostridioides difficile infection (CDI). During the initial stages of the COVID-19 pandemic, rates of CDI were similar to historical data despite the increased use of antibiotics. This may be due to increased emphasis on hygiene and protective equipment and reduced C. difficile testing as diarrhea was presumed to be COVID-19 related. Studies also demonstrated additional risk factors for CDI in COVID-19 patients, including length of hospitalization and new abdominal pain during admission. Although not associated with increased mortality, CDI was associated with increased length of hospital stay among patients admitted with COVID-19. Due to fecal viral shedding and concern of oral-fecal transmission of SARS-CoV-2, increased safety regulations were introduced to fecal microbiota transplantation (FMT) leading to reduced rates of this procedure during the COVID-19 pandemic. FMT for recurrent CDI during the COVID-19 pandemic remained highly effective without any reports of SARS-CoV-2 transmission. In addition, limited data show that FMT may be effective in treating COVID-19 and restoring healthy gut microbiota. The goal of this article is to review the impact that the COVID-19 pandemic has had on hospital-acquired CDI and the utilization of FMT.
Collapse
Affiliation(s)
- Salam P. Bachour
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rahul Dalal
- Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Brigham and Women’s Hospital, Boston, MA, USA
| | | |
Collapse
|
33
|
Zhang X, Ishikawa D, Ohkusa T, Fukuda S, Nagahara A. Hot topics on fecal microbiota transplantation for the treatment of inflammatory bowel disease. Front Med (Lausanne) 2022; 9:1068567. [PMID: 36530877 PMCID: PMC9755187 DOI: 10.3389/fmed.2022.1068567] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/21/2022] [Indexed: 11/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal mucosal inflammatory disease with complex etiology. Traditional anti-inflammatory treatment regimens have yielded unsatisfactory results. As research continues to deepen, it has been found that the gut microbiota of patients with IBD is generally altered. The presence of microorganisms in the human gastrointestinal tract is inextricably linked to the regulation of health and disease. Disruption of the microbiotic balance of microbiota in the gastrointestinal tract is called dysbiosis, which leads to disease. Therefore, in recent years, the exploration of therapeutic methods to restore the homeostasis of the gut microbiota has attracted attention. Moreover, the use of the well-established fecal microbiota transplantation (FMT) regimen for the treatment of Clostridioides difficile infection has attracted the interest of IBD researchers. Therefore, there are an increasing number of clinical studies regarding FMT for IBD treatment. However, a series of questions regarding FMT in the treatment of IBD warrants further investigation and discussion. By reviewing published studies, this review explored hot topics such as the efficacy, safety, and administration protocol flow of FMT in the treatment of IBD. Different administration protocols have generally shown reassuring results with significant efficacy and safety. However, the FMT treatment regimen needs to be further optimized. We believe that in the future, individual customized or standard FMT implementation will further enhance the relevance of FMT in the treatment of IBD.
Collapse
Affiliation(s)
- Xiaochen Zhang
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Dai Ishikawa
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
- Department of Regenerative Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Toshifumi Ohkusa
- Department of Microbiota Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Gastroenterology and Hepatology, The Jikei University Kashiwa Hospital, Chiba, Japan
| | - Shinji Fukuda
- Department of Regenerative Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akihito Nagahara
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
- Department of Regenerative Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Abstract
Introduction In the past few years, the advancement of 16S rRNA metagenomic analysis sequencing has enabled assessing the impact of gut microbiota on the development of skin disease. Alopecia areata (AA) is a nonscarring hair loss disorder with an unknown etiopathogenesis, however, it is hypothesised that a combination of genetic and environmental factors might be involved. Although numerous studies have shown that the microbiome plays a key role at the beginning of skin diseases, the link between AA and gut dysbiosis remains unclear. Aim To analyse the intestinal microbiome in patients suffering from AA. Material and methods The study describes the conceivable involvement of gut microbiota in the unclear pathogenesis of AA. We enrolled 25 patients, over 18 years of age with an active state of AA who donated their stool samples. The samples were examined at the human gut microbial community at the species level by metataxonomic analysis of the full-length 16S V3-V4 sequencing. Results The four major genera that constitute the microbiome's core are Lachnoclostridium, Bifidobacterium, Streptococcus, and Eubacterium, as well as three major phyla: Firmicutes, Proteobacteria, and Actinobacteria. Firmicutes and Proteobacteria are overrepresented in the microflora, which might suggest a disturbed microflora. Furthermore, the composition of bacterial communities suggests a loss of overall richness and a decrease in taxonomic diversity across all samples. Conclusions This study describes, for the first time, the characteristics of the gut microbiome in AA patients and may provide new insight into the gut microbiome that may play a role in the development of AA.
Collapse
|
35
|
Wang R, Liu M, Ren G, Luo G, Wang Z, Ge Z, Pu Q, Ren W, Yang S. Zhilong Huoxue Tongyu Capsules' Effects on ischemic stroke: An assessment using fecal 16S rRNA gene sequencing and untargeted serum metabolomics. Front Pharmacol 2022; 13:1052110. [PMID: 36467061 PMCID: PMC9715974 DOI: 10.3389/fphar.2022.1052110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/07/2022] [Indexed: 09/19/2023] Open
Abstract
Zhilong Huoxue Tongyu capsule (ZHTC) is an effective traditional Chinese medicine compound for the treatment of ischemic stroke, which is widely used in clinical ischemic stroke patients. However, it is uncertain whether ZHTC affects ischemic stroke through gut microbiota and serum metabolites. In this study, a rat model of middle cerebral artery occlusion (MCAO) was prepared. By evaluating motor nerve function score, cerebral infarct size, brain tissue damage and intestinal barrier damage, it was found that ZHTC improved stroke-related symptoms in MCAO rats. Using 16S rRNA gene sequencing, fecal microbial transplantation (FMT), untargeted metabolomics, and spearman correlation analysis of gut microbiota and serum metabolites, we found that ZHTC can regulate the abundance of p_Firmicutes, p_Bacteroidota,p_Proteobacteria, g_Prevotella, and g_Lactobacillus, and regulated 23 differential metabolites. Spearman correlation analysis found that Arginine was positively correlated with p_Firmicutes, o_Clostridiales, c_Clostridia, and negatively correlated with p_Bacteroidetes, c_Bacteroidia,o_Bacteroidales; L-Lysine was negatively correlated with f_Christensenellaceae; L-methionine was positively correlated with o_Lactobacillales, f_Lactobacillaceae, and g_Lactobacillus. Altogether, this study shows for the first time that ZHTC can ameliorate ischemic stroke by modulating gut microbiota and metabolic disturbances. This lays the foundation for further revealing the causal relationship between ZHTC, gut dysbiosis, plasma metabolite levels and ischemic stroke, and provides a scientific explanation for the ameliorating effect of ZHTC on ischemic stroke.
Collapse
Affiliation(s)
- Raoqiong Wang
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Mengnan Liu
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Guilin Ren
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Zhichuan Wang
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Zhengxin Ge
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Qingrong Pu
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
36
|
Rehman AU, Khan AI, Xin Y, Liang W. Morchella esculenta polysaccharide attenuate obesity, inflammation and modulate gut microbiota. AMB Express 2022; 12:114. [PMID: 36056976 PMCID: PMC9440975 DOI: 10.1186/s13568-022-01451-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
Edible mushrooms have now been suggested as promising sources of biological functional ingredients and are the subject of the most recent nutrition research and novel functional foods. Polysaccharides from mushrooms exhibit impressive biological effects, notably against obesity. Obesity is a chronic metabolic disorder characterized by chronic inflammation, gut dysbiosis, and hyperpermeability of the colon. Here, we prove that mushrooms Morchella esculenta polysaccharide (MEP) effects on HFD-induced obesity, colonic inflammation, and gut microbiota dysbiosis. Our findings demonstrate MEP supplementation attenuates obesity parameters and reduces inflammation in the colon via regulation of Toll-like receptor 4 (TLR4), nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and inactivation of nuclear factor kappa B (NF-κB). Furthermore, MEP administration restores gut microbiota dysregulation by ameliorating Firmicutes to Bacteroidetes proportion as well as enhancing beneficial bacteria, like Lactobacillus, and inhibiting pathogenic bacteria like Enterococcus. MEP improves gut integrity by increasing tight junction proteins (TJs) and reducing endotoxin levels by controlling Lipopolysaccharide (LPS) in HFD-induced obese mice. These results demonstrated the therapeutic efficacy of MEP in attenuating HFD-induced obesity via regulating inflammatory cascades, ameliorating the gut microbiome, and modulating gut integrity.
Collapse
Affiliation(s)
- Ata Ur Rehman
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Asif Iqbal Khan
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yi Xin
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China.
| | - Wang Liang
- Clinical Stem cell Research Centre, First Affiliated Hospital, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
37
|
Luo L, Luo J, Cai Y, Fu M, Li W, Shi L, Liu J, Dong R, Xu X, Tu L, Yang Y. Inulin-type fructans change the gut microbiota and prevent the development of diabetic nephropathy. Pharmacol Res 2022; 183:106367. [PMID: 35882293 DOI: 10.1016/j.phrs.2022.106367] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
Abstract
Diabetic nephropathy (DN) is the most common cause of end-stage renal disease, and few treatment options that prevent the progressive loss of renal function are available. Studies have shown that dietary fiber intake improves kidney diseases and metabolism-related diseases, most likely through short-chain fatty acids (SCFAs). The present study aimed to examine the protective effects of inulin-type fructans (ITFs) on DN through 16 S rRNA gene sequencing, gas chromatographymass spectrometry (GCMS) analysis and fecal microbiota transplantation (FMT). The results showed that ITFs supplementation protected against kidney damage in db/db mice and regulated the composition of the gut microbiota. Antibiotic treatment and FMT experiments further demonstrated a key role of the gut microbiota in mediating the beneficial effects of ITFs. The ITFs treatment-induced changes in the gut microbiota led to an enrichment of SCFA-producing bacteria, especially the genera Akkermansia and Candidatus Saccharimonas, which increased the fecal and serum acetate concentrations. Subsequently, acetate supplementation improved glomerular damage and renal fibrosis by attenuating mitochondrial dysfunction and reducing toxic glucose metabolite levels. In conclusion, ITFs play a renoprotective role by modulating the gut microbiota and increasing acetate production. Furthermore, acetate mediates renal protection by regulating glucose metabolism, decreasing glycotoxic product levels and improving mitochondrial function.
Collapse
Affiliation(s)
- Liman Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Division of Endocrinology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yueting Cai
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Menglu Fu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenhua Li
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lili Shi
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingrui Liu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruolan Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xizhen Xu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Yan Yang
- Division of Endocrinology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
38
|
Chang SLY, Lin YY, Liu SC, Tsai YS, Lin SW, Chen YL, Chen CC, Ko CY, Chen HT, Chen WC, Tang CH. Oral Administration of Clostridium butyricum GKB7 Ameliorates Signs of Osteoarthritis in Rats. Cells 2022; 11:2169. [PMID: 35883610 PMCID: PMC9323988 DOI: 10.3390/cells11142169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative and painful inflammatory joint disease affecting the cartilage, bone, and synovial membranes, without any effective treatment that targets the underlying mechanisms of OA. Our study evaluated the therapeutic effects of a live probiotic strain, Clostridium butyricum GKB7, administered for 6 weeks to rats with knee OA (KOA) induced by anterior cruciate ligament transection (ACLT) of the right knee. All rats underwent weekly weight-bearing behavioral testing and body weight measurements. At 6 weeks, all rats were sacrificed, and the right hind knees were collected for micro-computed tomography imaging and histopathological and immunohistochemical analyses. Compared with rats in the ACLT-only group, ACLT rats administered the probiotic exhibited dramatic improvements in pain-related behavior from postoperative week 2, had significantly less osseous and cartilaginous damage at week 6, and significantly lower levels of the inflammatory markers interleukin 1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in cartilage and synovium sections. C. butyricum GKB7 appeared to slow or even reverse OA progression and is worth investigating as a novel therapeutic for OA.
Collapse
Affiliation(s)
- Sunny Li-Yun Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (S.L.-Y.C.); (C.-Y.K.)
- School of Medicine, China Medical University, Taichung 404333, Taiwan;
| | - Yen-You Lin
- School of Medicine, China Medical University, Taichung 404333, Taiwan;
| | - Shan-Chi Liu
- Department of Medical Education and Research, Beigang Hospital, China Medical University, Yunlin 651012, Taiwan;
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (Y.-S.T.); (S.-W.L.); (Y.-L.C.)
| | - Shih-Wei Lin
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (Y.-S.T.); (S.-W.L.); (Y.-L.C.)
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (Y.-S.T.); (S.-W.L.); (Y.-L.C.)
| | - Chin-Chu Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei 106617, Taiwan;
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei 104036, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Chih-Yuan Ko
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (S.L.-Y.C.); (C.-Y.K.)
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 404333, Taiwan;
| | - Hsien-Te Chen
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 404333, Taiwan;
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung 404333, Taiwan
| | - Wei-Cheng Chen
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
- Division of Sports Medicine & Surgery, Department of Orthopedic Surgery, MacKay Memorial Hospital, Taipei 104217, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (S.L.-Y.C.); (C.-Y.K.)
- School of Medicine, China Medical University, Taichung 404333, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 404333, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung 40354, Taiwan
| |
Collapse
|
39
|
Elhag DA, Kumar M, Saadaoui M, Akobeng AK, Al-Mudahka F, Elawad M, Al Khodor S. Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response. Int J Mol Sci 2022; 23:ijms23136966. [PMID: 35805965 PMCID: PMC9266456 DOI: 10.3390/ijms23136966] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated inflammation of the gastrointestinal tract with a highly heterogeneous presentation. It has a relapsing and remitting clinical course that necessitates lifelong monitoring and treatment. Although the availability of a variety of effective therapeutic options including immunomodulators and biologics (such as TNF, CAM inhibitors) has led to a paradigm shift in the treatment outcomes and clinical management of IBD patients, some patients still either fail to respond or lose their responsiveness to therapy over time. Therefore, according to the recent Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE-II) recommendations, continuous disease monitoring from symptomatic relief to endoscopic healing along with short- and long-term therapeutic responses are critical for providing IBD patients with a tailored therapy algorithm. Moreover, considering the high unmet need for novel therapeutic approaches for IBD patients, various new modulators of cytokine signaling events (for example, JAK/TYK inhibitors), inhibitors of cytokines (for example IL-12/IL-23, IL-22, IL-36, and IL-6 inhibitors), anti-adhesion and migration strategies (for example, β7 integrin, sphingosine 1-phosphate receptors, and stem cells), as well as microbial-based therapeutics to decolonize the bed buds (for example, fecal microbiota transplantation and bacterial inhibitors) are currently being evaluated in different phases of controlled clinical trials. This review aims to offer a comprehensive overview of available treatment options and emerging therapeutic approaches for IBD patients. Furthermore, predictive biomarkers for monitoring the therapeutic response to different IBD therapies are also discussed.
Collapse
Affiliation(s)
- Duaa Ahmed Elhag
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
| | - Manoj Kumar
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
| | - Marwa Saadaoui
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
| | - Anthony K. Akobeng
- Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar; (A.K.A.); (F.A.-M.); (M.E.)
| | - Fatma Al-Mudahka
- Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar; (A.K.A.); (F.A.-M.); (M.E.)
| | - Mamoun Elawad
- Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar; (A.K.A.); (F.A.-M.); (M.E.)
| | - Souhaila Al Khodor
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
- Correspondence:
| |
Collapse
|
40
|
The Combination of Intestinal Alkaline Phosphatase Treatment with Moderate Physical Activity Alleviates the Severity of Experimental Colitis in Obese Mice via Modulation of Gut Microbiota, Attenuation of Proinflammatory Cytokines, Oxidative Stress Biomarkers and DNA Oxidative Damage in Colonic Mucosa. Int J Mol Sci 2022; 23:ijms23062964. [PMID: 35328382 PMCID: PMC8955215 DOI: 10.3390/ijms23062964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are commonly considered as Crohn's disease and ulcerative colitis, but the possibility that the alterations in gut microbiota and oxidative stress may affect the course of experimental colitis in obese physically exercising mice treated with the intestinal alkaline phosphatase (IAP) has been little elucidated. Mice fed a high-fat-diet (HFD) or normal diet (ND) for 14 weeks were randomly assigned to exercise on spinning wheels (SW) for 7 weeks and treated with IAP followed by intrarectal administration of TNBS. The disease activity index (DAI), grip muscle strength test, oxidative stress biomarkers (MDA, SOD, GSH), DNA damage (8-OHdG), the plasma levels of cytokines IL-2, IL-6, IL-10, IL-12p70, IL-17a, TNF-α, MCP-1 and leptin were assessed, and the stool composition of the intestinal microbiota was determined by next generation sequencing (NGS). The TNBS-induced colitis was worsened in obese sedentary mice as manifested by severe colonic damage, an increase in DAI, oxidative stress biomarkers, DNA damage and decreased muscle strength. The longer running distance and weight loss was observed in mice given IAP or subjected to IAP + SW compared to sedentary ones. Less heterogeneous microbial composition was noticed in sedentary obese colitis mice and this effect disappeared in IAP + SW mice. Absence of Alistipes, lower proportion of Turicibacter, Proteobacteria and Faecalibacterium, an increase in Firmicutes and Clostridium, a decrease in oxidative stress biomarkers, 8-OHdG content and proinflammatory cytokines were observed in IAP + SW mice. IAP supplementation in combination with moderate physical activity attenuates the severity of murine colitis complicated by obesity through a mechanism involving the downregulation of the intestinal cytokine/chemokine network and oxidative stress, the modulation of the gut microbiota and an improvement of muscle strength.
Collapse
|
41
|
Liu L, Fu Q, Li T, Shao K, Zhu X, Cong Y, Zhao X. Gut microbiota and butyrate contribute to nonalcoholic fatty liver disease in premenopause due to estrogen deficiency. PLoS One 2022; 17:e0262855. [PMID: 35108315 PMCID: PMC8809533 DOI: 10.1371/journal.pone.0262855] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) in postmenopausal women has increased significantly. Estrogen plays a very important role in NAFLD, but whether NAFLD in premenopausal women was caused by estrogen deficiency was unknown. Thus, it is of great clinical significance to explore the mechanism of NAFLD in premenopausal women. Gut microbiota and its metabolites short chain fatty acids (SCFA) have been shown to play important roles in the development of NAFLD. In this study, we investigated the impact of gut microbiota and SCFA in NAFLD patients and mice caused by estrogen deficiency. We showed that premenopause NAFLD patients had much lower estrogen levels. Estrogen deficient mice, due to ovariectomy (OVX), suffered more severe liver steatosis with an elevated body weight, abdominal fat weight, serum triglycerides and deterioration in hepatic steatosis. Altered gut microbiota composition and decreased butyrate content were found in NAFLD patients and in OVX mice. Furthermore, fecal microbiota transplantation (FMT) or supplementing with butyrate alleviated NAFLD in OVX mice. The production of antimicrobial peptides (AMP) Reg3ɣ, β-defensins and the expression of intestinal epithelial tight junction, including ZO-1 and Occluding-5, were decreased in the OVX mice compared to control mice. Upregulation of PPAR-ɣ and VLDLR, downregulation of PPAR-ɑ indicated that OVX mice suffered from abnormal lipid metabolism. These data indicate that changes in the gut microbiota and SCFA caused by estrogen reduction, together with a disorder in AMP production and lipid metabolism, promote NAFLD, thus provide SCFAs derived from microbiota as new therapeutic targets for the clinical prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Limin Liu
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Qingsong Fu
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- * E-mail: (XZ); (QF)
| | - Tiantian Li
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Kai Shao
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xing Zhu
- Department of Pathology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Xiaoyun Zhao
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- * E-mail: (XZ); (QF)
| |
Collapse
|
42
|
Michel‐Mata S, Wang X, Liu Y, Angulo MT. Predicting microbiome compositions from species assemblages through deep learning. IMETA 2022; 1:e3. [PMID: 35757098 PMCID: PMC9221840 DOI: 10.1002/imt2.3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 05/13/2023]
Abstract
Microbes can form complex communities that perform critical functions in maintaining the integrity of their environment or their hosts' well-being. Rationally managing these microbial communities requires improving our ability to predict how different species assemblages affect the final species composition of the community. However, making such a prediction remains challenging because of our limited knowledge of the diverse physical, biochemical, and ecological processes governing microbial dynamics. To overcome this challenge, we present a deep learning framework that automatically learns the map between species assemblages and community compositions from training data only, without knowing any of the above processes. First, we systematically validate our framework using synthetic data generated by classical population dynamics models. Then, we apply our framework to data from in vitro and in vivo microbial communities, including ocean and soil microbiota, Drosophila melanogaster gut microbiota, and human gut and oral microbiota. We find that our framework learns to perform accurate out-of-sample predictions of complex community compositions from a small number of training samples. Our results demonstrate how deep learning can enable us to understand better and potentially manage complex microbial communities.
Collapse
Affiliation(s)
- Sebastian Michel‐Mata
- Center for Applied Physics and Advanced TechnologyUniversidad Nacional Autónoma de MéxicoJuriquillaMexico
- Department of Ecology and Evolutionary BiologyPrinceton UniversityPrincetonNew JerseyUSA
| | - Xu‐Wen Wang
- Channing Division of Network Medicine, Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yang‐Yu Liu
- Channing Division of Network Medicine, Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Marco Tulio Angulo
- CONACyT—Institute of MathematicsUniversidad Nacional Autónoma de MéxicoJuriquillaMexico
| |
Collapse
|
43
|
Littleford-Colquhoun BL, Weyrich LS, Hohwieler K, Cristescu R, Frère CH. How microbiomes can help inform conservation: landscape characterisation of gut microbiota helps shed light on additional population structure in a specialist folivore. Anim Microbiome 2022; 4:12. [PMID: 35101152 PMCID: PMC8802476 DOI: 10.1186/s42523-021-00122-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The koala (Phascolarctos cinereus), an iconic yet endangered specialised folivore experiencing widespread decline across Australia, is the focus of many conservation programs. Whilst animal translocation and progressive conservation strategies such as faecal inoculations may be required to bring this species back from the brink of extinction, insight into the variation of host-associated gut microbiota and the factors that shape this variation are fundamental for their success. Despite this, very little is known about the landscape variability and factors affecting koala gut microbial community dynamics. We used large scale field surveys to evaluate the variation and diversity of koala gut microbiotas and compared these diversity patterns to those detected using a population genetics approach. Scat samples were collected from five locations across South East Queensland with microbiota analysed using 16S rRNA gene amplicon sequencing. RESULTS Across the landscape koala gut microbial profiles showed large variability, with location having a large effect on bacterial community composition and bacterial diversity. Certain bacteria were found to be significantly differentially abundant amongst locations; koalas from Noosa showed a depletion in two bacterial orders (Gastranaerophilales and Bacteroidales) which have been shown to provide beneficial properties to their host. Koala gut microbial patterns were also not found to mirror population genetic patterns, a molecular tool often used to design conservation initiatives. CONCLUSIONS Our data shows that koala gut microbiotas are extremely variable across the landscape, displaying complex micro- and macro- spatial variation. By detecting locations which lack certain bacteria we identified koala populations that may be under threat from future microbial imbalance or dysbiosis. Additionally, the mismatching of gut microbiota and host population genetic patterns exposed important population structure that has previously gone undetected across South East Queensland. Overall, this baseline data highlights the importance of integrating microbiota research into conservation biology in order to guide successful conservation programs such as species translocation and the implementation of faecal inoculations.
Collapse
Affiliation(s)
- B. L. Littleford-Colquhoun
- Global Change Ecology, School of Science and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556 Australia
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912 USA
- Institute at Brown for Environment and Society, Brown University, Providence, RI 02912 USA
| | - L. S. Weyrich
- Department of Anthropology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802 USA
- School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005 Australia
| | - K. Hohwieler
- Global Change Ecology, School of Science and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556 Australia
| | - R. Cristescu
- Global Change Ecology, School of Science and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556 Australia
| | - C. H. Frère
- Global Change Ecology, School of Science and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556 Australia
| |
Collapse
|
44
|
Establishment and resilience of transplanted gut microbiota in aged mice. iScience 2022; 25:103654. [PMID: 35024588 PMCID: PMC8733228 DOI: 10.1016/j.isci.2021.103654] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/29/2021] [Accepted: 12/13/2021] [Indexed: 01/04/2023] Open
Abstract
The maintenance of healthy and resilient gut microbiota is critical for the life quality and healthspan of the elderly. Fecal microbiota transplantation (FMT) has been increasingly used to restore healthy gut microbiota. We systemically studied the establishment and resilience of transplanted microbiota after autologous versus heterologous FMT in aged recipients. Gut microbiota of aged mice (20 months old) failed to restore their original diversity and composition over 8 weeks via spontaneous recovery after antibiotics treatment; in contrast, FMT using either autologous or heterologous (2 months old from a different vendor) donors facilitated the recovery successfully, established donor-like microbiota states, and affected host gene expression profile. Furthermore, the transplanted microbiota established by heterologous FMT is not resilient during chemical-induced colonic inflammation, in contrast to that of autologous FMT. Our findings highlighted the need to monitor the long-term stability of transplanted gut microbiota and to perform multiple FMT when necessary. Aged mice microbiota restores slowly after antibiotics treatment Both autologous and heterologous FMT facilitate microbiota restoration in aged mice FMT affects long-term homeostasis of gut metagenome and colon gene expression Established microbiota after heterologous FMT is not resilient against colitis
Collapse
|
45
|
Liu P, Zhou W, Xu W, Peng Y, Yan Y, Lu L, Mi J, Zeng X, Cao Y. The Main Anthocyanin Monomer from Lycium ruthenicum Murray Fruit Mediates Obesity via Modulating the Gut Microbiota and Improving the Intestinal Barrier. Foods 2021; 11:foods11010098. [PMID: 35010223 PMCID: PMC8750395 DOI: 10.3390/foods11010098] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
Anthocyanins have been shown to exert certain antiobesity properties, but the specific relationship between anthocyanin-induced beneficial effects and the gut microbiota remains unclear. Petunidin-3-O-[rhamnopyranosyl-(trans-p-coumaroyl)]-5-O-(β-D-glucopyranoside) (P3G) is the main anthocyanin monomer from the fruit of Lycium ruthenicum Murray. Therefore, in this study, we investigated the antiobesity and remodeling effects of P3G on gut microbiota through a high-fat diet (HFD)-induced obesity mouse model and a fecal microbiota transplantation experiment. P3G was found to reduce body weight gain, fat accumulation, and liver steatosis in HFD-induced obese mice. Moreover, supplementation with P3G alleviated the HFD-induced imbalance in gut microbiota composition, and transferring the P3G-regulated gut microbiota to recipient mice provided comparable protection against obesity. This is the first time evidence is provided that P3G has an antiobesity effect by changing the intestinal microbiota. Our present data highlight a link between P3G intervention and enhancement in gut barrier integrity. This may be a promising option for obesity prevention.
Collapse
Affiliation(s)
- Peiyun Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (P.L.); (W.Z.); (W.X.); (Y.P.)
| | - Wangting Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (P.L.); (W.Z.); (W.X.); (Y.P.)
| | - Weiqi Xu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (P.L.); (W.Z.); (W.X.); (Y.P.)
| | - Yujia Peng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (P.L.); (W.Z.); (W.X.); (Y.P.)
| | - Yamei Yan
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China; (Y.Y.); (L.L.); (J.M.)
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| | - Lu Lu
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China; (Y.Y.); (L.L.); (J.M.)
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| | - Jia Mi
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China; (Y.Y.); (L.L.); (J.M.)
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (P.L.); (W.Z.); (W.X.); (Y.P.)
- Correspondence: (X.Z.); (Y.C.); Tel.: +86-25-84396791 (X.Z.); +86-951-6886783 (Y.C.)
| | - Youlong Cao
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China; (Y.Y.); (L.L.); (J.M.)
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
- Correspondence: (X.Z.); (Y.C.); Tel.: +86-25-84396791 (X.Z.); +86-951-6886783 (Y.C.)
| |
Collapse
|
46
|
Shi Y, Zou Y, Xiong Y, Zhang S, Song M, An X, Liu C, Zhang W, Chen S. Host Gasdermin D restrains systemic endotoxemia by capturing Proteobacteria in the colon of high-fat diet-feeding mice. Gut Microbes 2021; 13:1946369. [PMID: 34275417 PMCID: PMC8288038 DOI: 10.1080/19490976.2021.1946369] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gasdermin D (GSDMD) functions as a key pyroptotic executor through its secreted N-terminal domain (GSDMD-N). However, the functional relevance and mechanistic basis of the precise roles of host colonic GSDMD in high-fat diet (HFD)-induced gut dysbiosis and systemic endotoxemia remain elusive. In this study, we demonstrate that HFD feeding triggers GSDMD-N secretion of both T-lymphocytes and enterocytes in mouse colons. GSDMD deficiency aggravates HFD-induced systemic endotoxemia, gut barrier impairment, and colonic inflammation. More importantly, active GSDMD-N kills the Proteobacteria phylum via directly interacting with Cardiolipin. Mechanistically, we identify that the Glu236 (a known residue for GSDMD protein cleavage) is a bona fide important site for the bacterial recognition of GSDMD. Collectively, our findings explain the mechanism by which colonic GSDMD-N maintains low levels of HFD-induced metabolic endotoxemia. A GSDMD-N mimetic containing an exposed Glu236 site could be an attractive strategy for the treatment of HFD-induced metabolic endotoxemia.
Collapse
Affiliation(s)
- Yujie Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yixin Zou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yonghong Xiong
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shiyao Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,CONTACT Siyu Chen
| |
Collapse
|
47
|
Mortelé O, Britto XB, Christine L, Surbhi MK, Jorens Philippe G, Eveline D, van Nuijs AL, Nina H. Obesity influences the microbiotic biotransformation of chlorogenic acid. J Pharm Biomed Anal 2021; 211:114550. [DOI: 10.1016/j.jpba.2021.114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/24/2021] [Accepted: 12/21/2021] [Indexed: 11/15/2022]
|
48
|
Wu J, Qiu M, Zhang C, Zhang C, Wang N, Zhao F, Lv L, Li J, Lyu-Bu AGA, Wang T, Zhao B, You S, Wu Y, Wang X. Type 3 resistant starch from Canna edulis modulates obesity and obesity-related low-grade systemic inflammation in mice by regulating gut microbiota composition and metabolism. Food Funct 2021; 12:12098-12114. [PMID: 34784410 DOI: 10.1039/d1fo02208c] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Obesity is a most prevalent human health problem. Several studies showed that appropriate modulation of gut microbiota could help reshape the metabolic profile of obese individuals, thereby altering the development of obesity. A nutritional strategy for treating obesity includes prebiotics. Type 3 Resistant Starch from Canna edulis (Ce-RS3) is a dietary fiber that exerts potential effects on the intestinal microbial community; however, the metabolic landscape and anti-obesity mechanism remain unclear. In the present study, obese mice were treated with Ce-RS3, and 16S rRNA gene sequencing and metabolomics were used to measure changes in gut microbiota and fecal metabolic profiles, respectively. At the end of the treatment (13 weeks), we observed slow weight gain in the mice, and pathological damage and inflammation were substantially reduced. Ce-RS3 constructs a healthy gut microbiota structure and can enhance intestinal immunity and reduce metabolic inflammation. Ce-RS3 increased the diversity of gut microbiota with enrichment of Bifidobacterium and Roseburia. Ce-RS3 regulated the systemic metabolic dysbiosis in obese mice and adjusted 26 abnormal metabolites in amino acids and lipids metabolism, many of which are related to the microbiome. More importantly, we found that the anti-obesity effect of Ce-RS3 can be transferred by fecal transplantation. The beneficial effects of Ce-RS3 might derive from gut microbiota changes, which might improve obesity and metabolic inflammation by altering host-microbiota interactions with impacts on the metabolome. In conclusion, Ce-RS3 can be used as a prebiotic with potential value for the treatment of obesity.
Collapse
Affiliation(s)
- Jiahui Wu
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - Minyi Qiu
- Medicament Department, Peking University People's Hospital, Beijing, 100044, China
| | - Chi Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - Caijuan Zhang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Nan Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - Fangyuan Zhao
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - Liqiao Lv
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - Junling Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - A G A Lyu-Bu
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shaowei You
- The Second Affiliated Hospital of Guizhou University of traditional Chinese Medicine, Guizhou, 550003, China
| | - Yuanhua Wu
- The First Affiliated Hospital of Guizhou University of traditional Chinese Medicine, Gouzhou, 550001, China
| | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| |
Collapse
|
49
|
The intestinal flora of patients with GHPA affects the growth and the expression of PD-L1 of tumor. Cancer Immunol Immunother 2021; 71:1233-1245. [PMID: 34647152 PMCID: PMC9016060 DOI: 10.1007/s00262-021-03080-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
Context Pituitary adenoma (PA) is a common intracranial tumor. The evidence indicates that the tumor immune microenvironment (TIME) is associated with PA and that the intestinal flora influences other tumors' growth through interacting with the TIME. However, how the intestinal microbial flora contributes to the development of PA through the immune response is unknown.
Objective and methods Here we used high-throughput Illumina MiSeq sequencing targeting the V3−V4 region of the 16S ribosomal RNA gene to investigate the intestinal flora of patients with growth hormone-secreting pituitary adenoma (GHPA), nonfunctional pituitary adenoma (NFPA), and healthy controls. We determined their effects on tumor growth and the TIME. Fecal microbiota transplantation (FMT) was performed after adoptive transfer via peripheral blood mononuclear cells to tumor-bearing nude mice, which allowed the study of the immune response. Result We discovered differences in the structures and quantities of intestinal flora between patients with GHPA, patients with NFPA, and healthy controls. After FMT, the intestinal flora of GHPA patients promoted the growth of tumors in mouse models. The number of programmed cell death ligand 1 (PD-L1)-positive cells increased in tumor tissues as well as the extent of infiltration of CD8+ cells. Increased numbers of CD3+CD8+ cells and increased levels of sPD-L1 were detected in peripheral blood. Conclusion These findings indicated that the intestinal flora of patients with GHPA promoted tumor growth and that the immune system may mediate this change. Supplementary Information The online version contains supplementary material available at 10.1007/s00262-021-03080-6.
Collapse
|
50
|
Liu C, Wang YL, Yang YY, Zhang NP, Niu C, Shen XZ, Wu J. Novel approaches to intervene gut microbiota in the treatment of chronic liver diseases. FASEB J 2021; 35:e21871. [PMID: 34473374 DOI: 10.1096/fj.202100939r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Recent investigations of gut microbiota have contributed to understanding of the critical role of microbial community in pathophysiology. Dysbiosis not only causes disturbance directly to the gastrointestinal tract but also affects the liver through gut-liver axis. Various types of dysbiosis have been documented in alcoholic liver disease (ALD), nonalcoholic fatty liver disease, autoimmune hepatitis (AIH), primary sclerosing cholangitis, and may be crucial for the initiation, progression, or deterioration to end-stage liver disease. A few microbial species have been identified as the causal factors leading to these chronic illnesses that either do not have clear etiologies or lack effective treatment. Notably, cytolysin-producing Enterococcus faecalis, Klebsiella pneumoniae and Enterococcus gallinarum were defined for ALD, NASH, and AIH, respectively. These groundbreaking discoveries drive a rapid development in innovative therapeutics, such as fecal microbial transplantation and implementation of specific bacteriophages in addition to prebiotics, probiotics, or synbiotics for intervention of dysbiosis. Although most emerging interventions are in preclinical development or early clinical trials, a better delineation of specific dysbiosis in these disorders at metabolic, immunogenic, or molecular levels in establishing particular causal effects aids in modulating or correcting the microbial community which is the part of daily life for human being.
Collapse
Affiliation(s)
- Chang Liu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Yu-Li Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Yong-Yu Yang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Ning-Ping Zhang
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China.,Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, China
| | - Chen Niu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Xi-Zhong Shen
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China.,Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, China
| | - Jian Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China.,Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China.,Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|