1
|
Shi L, Wang W, Jing C, Hu J, Liao X. Berberine and health outcomes: an overview of systematic reviews. BMC Complement Med Ther 2025; 25:147. [PMID: 40269802 DOI: 10.1186/s12906-025-04872-4] [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: 08/05/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Berberine is an isoquinoline alkaloid isolated from Chinese herb coptis chinensis and other berberis plants which can be used to treat a wide range of chronic diseases. However, the current research evidence on the therapeutic effects of berberine has not been summarized. We aimed to synthesize the current evidence on the systematic review (SRs) of berberine for the treatment of diverse conditions. METHODS A comprehensive search of the Cochrane Library, PubMed, EMBASE, Web of Science, CNKI, Wanfang, VIP, and SinoMed was performed from the database inception to April 11, 2024. SRs on berberine were included and evaluated. The methodological quality and the reporting quality of each SR were assessed using the AMSTAR-2 tool and PRISMA checklist, respectively. The quality of evidence was appraised based on the GRADE. RESULTS Fifty-four SRs were included and analyzed. Overall, associations were found between berberine and 70 health outcomes concerned with 9 diseases. Berberine has improved most outcomes of these diseases: 78% (25/32) cardiovascular disease outcomes, 92.59% (25/27) type 2 diabetes mellitus outcomes, 94.74% (18/19) gastrointestinal disorders outcomes, 72.22% (13/18) polycystic ovary syndrome (PCOS) outcomes, 86.67% (13/15) non-alcoholic fatty liver disease (NAFLD) outcomes, 92.31% (12/13) schizophrenia outcomes, 90.91% (10/11) metabolic syndrome outcomes, 57.14% (4/7) obesity outcomes, and 100.00% (6/6) dyslipidemia outcomes. There was a high overlap of primary studies (CCA > 15%) in the SRs of PCOS, NAFLD, obesity, and schizophrenia. Only one SR was rated as high quality while eight SRs were rated as low quality and forty-five SRs as very low quality according to AMSTAR-2. Regarding the reporting quality, Item 14, 15, 21, and 22 were poorly reported for the included SRs in terms of PRSMA assessment. For GRADE, eight outcomes were rated as high quality evidence, twenty-two outcomes were rated as moderate quality, and 110 outcomes were rated as low quality. CONCLUSION Current evidence suggests that berberine has beneficial effects on a range of health outcomes for people with chronic diseases. Specifically, berberine significantly improves type 2 diabetes, gastrointestinal disorders, schizophrenia, metabolic syndrome, and dyslipidemia outcomes. However, caution is needed considering the shortcomings in the quality of the relevant system reviews included.
Collapse
Affiliation(s)
- Lanjun Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Wenya Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Chengyang Jing
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Jing Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, China.
| | - Xing Liao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China.
| |
Collapse
|
2
|
Zhang M, Feng C, Zhang B, Yin Y, Chen J, Liu H, Farag MA, Mamadalieva NZ, Li N, Sun J, Sun S, Liu C. In vitro and in vivo immune-enhancing effects of punicic acid and the action mechanisms as revealed via microbiome and lipid profiling. Food Funct 2025; 16:3120-3133. [PMID: 40159912 DOI: 10.1039/d4fo05023a] [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: 04/02/2025]
Abstract
Punicic acid (PA) is a chief component of pomegranate seed oil with several health benefits. In this study, the in vitro immunomodulatory activity of PA was assessed using RAW264.7 cells, revealing that PA activated the macrophages, facilitated the concentration of immune-related cytokines and enzymes, and regulated the immune-related NF-κB and MAPK signaling pathways. Further, the in vivo immune-enhancing effect of PA was evaluated with the cyclophosphamide (CTX)-induced immune-compromised mouse model with 16S rDNA amplicon sequencing and relative quantification of lipidome. Results indicated that high doses of PA (200 mg kg-1) remarkably restored CTX-induced immune injury by enhancing the innate and adaptive immunity to stimulate the secretion of immune-related factors. In addition, PA improved gut microbiota dysbiosis and ameliorated lipid metabolism disorders. Our research provides a theoretical basis for the exploitation of PA as a functional component with immune-enhancing effects and adds to the potential health uses of pomegranate seed oil.
Collapse
Affiliation(s)
- Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Caiyun Feng
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China
| | - Bo Zhang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China
| | - Yanlei Yin
- Shandong Institute of Pomology, Tai'an, 271000, China
| | - Jinlong Chen
- Work Station of Forest Fruit Industry in Kashi, Kashi, 844000, PR China
| | - Haoran Liu
- JiMei One Health Industry (Shandong) Co., Ltd, Zaozhuang, 277300, PR China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nilufar Z Mamadalieva
- Institute of the Chemistry of Plant Substances of the Academy Sciences of Uzbekistan, Tashkent, 100170, Uzbekistan
| | - Ningyang Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, PR China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Shutao Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- Shandong Aojing Biotechnology Co., Ltd, Jining, 273500, PR China
| |
Collapse
|
3
|
Fu B, Zhao X, Khan M, Jiang Y, Li W, Mushtaq M, Danzeng B, Ni X, Azeem Z, Shao Q, Xue B, Ouyang Y. Cecum microbiota composition, fermentation characteristics, and immunometabolic biomarkers of Yunshang black goat fed varying dietary energy and protein levels. Front Microbiol 2025; 16:1523586. [PMID: 39967730 PMCID: PMC11832493 DOI: 10.3389/fmicb.2025.1523586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Ruminants including goats have diverse microcosms of microbiota involved in diet digestion, absorption, and assimilation. Moreover, it is well known that changes in dietary regimens including nutrient levels result in varied gut microbiota composition, and ultimately, the performance and health of these animals. Methods The current study examined the effects of varying dietary energy and protein levels on the cecal fermentation, immune biomarkers, and microbiota characteristics of 80 male Yunshan Black Goats (6 months, ~35.82 ± 2.79 kg), divided into four diets: 1) High Energy-High Protein (HEHP), 2) High Energy-Low Protein (HELP), 3) Low Energy-High Protein (LEHP), and 4) Low Energy-Low Protein (LELP). Twenty goats (five from each treatment group) were randomly slaughtered after a 50-day feeding trial, and cecal digesta and tissue were sampled for microbial analysis. Results The cecal content revealed that the high-energy groups (HEHP, HELP) had lower pH levels than the LEHP group (p < 0.05) and significantly higher valeric and isovaleric acid concentrations in HEHP. Although species richness (Chao1 index) remained consistent, the HEHP group showed higher diversity (Shannon and Simpson indices) than LEHP (p < 0.05). Dominant phyla included Bacteroidetes and Firmicutes; LEHP and LELP had significantly higher Bacteroidetes abundance than HELP, while HELP had higher Firmicutes abundance than LEHP (p < 0.05). Verrucomicrobia abundance was lower in LEHP than in HELP and LELP (p < 0.05). At the genus level, 311 genera were identified, with Clostridium, Prevotella, unidentified_BS11, and others showing significant variation. The HELP group had lower unidentified_BS11 than LEHP and LELP, and higher unidentified_Ruminococcaceae, Clostridium, and Lachnospiraceae than LEHP (p < 0.05). VFA metabolism, absorption, cytokine expression, and tight junction protein mRNA in cecal tissue were also analyzed. Genes like MCT-1 and SLC16A4, linked to VFA absorption, positively correlated with Paludibacter, which was associated with immune markers (TLR-3, TLR-4, IFN-γ) and Occludin expression. In contrast, VFA-related genes and tight junction proteins negatively correlated with unidentified Fibrobacterales, suggesting a microbial role in adaptive immunity. Conclusion This study demonstrated that dietary energy and protein levels significantly influenced cecal fermentation, immune biomarkers, and microbiota composition in Yunshan Black Goats.
Collapse
Affiliation(s)
- Binlong Fu
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Xiaoqi Zhao
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Muhammad Khan
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Yanting Jiang
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Weijuan Li
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Maida Mushtaq
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Baiji Danzeng
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Xiaojun Ni
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Zobia Azeem
- Department of Zoology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Qingyong Shao
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| | - Bai Xue
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yina Ouyang
- Yunnan Animal Sciences and Veterinary Institute, Kunming, China
| |
Collapse
|
4
|
Yasmeen F, Pirzada RH, Ahmad B, Choi B, Choi S. Understanding Autoimmunity: Mechanisms, Predisposing Factors, and Cytokine Therapies. Int J Mol Sci 2024; 25:7666. [PMID: 39062908 PMCID: PMC11277571 DOI: 10.3390/ijms25147666] [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: 06/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Autoimmunity refers to an organism's immune response against its own healthy cells, tissues, or components, potentially leading to irreversible damage to vital organs. Central and peripheral tolerance mechanisms play crucial roles in preventing autoimmunity by eliminating self-reactive T and B cells. The disruption of immunological tolerance, characterized by the failure of these mechanisms, results in the aberrant activation of autoreactive lymphocytes that target self-tissues, culminating in the pathogenesis of autoimmune disorders. Genetic predispositions, environmental exposures, and immunoregulatory disturbances synergistically contribute to the susceptibility and initiation of autoimmune pathologies. Within the realm of immune therapies for autoimmune diseases, cytokine therapies have emerged as a specialized strategy, targeting cytokine-mediated regulatory pathways to rectify immunological imbalances. Proinflammatory cytokines are key players in inducing and propagating autoimmune inflammation, highlighting the potential of cytokine therapies in managing autoimmune conditions. This review discusses the etiology of autoimmune diseases, current therapeutic approaches, and prospects for future drug design.
Collapse
Affiliation(s)
- Farzana Yasmeen
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Rameez Hassan Pirzada
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Bilal Ahmad
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Bogeum Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| |
Collapse
|
5
|
Majumder S, Kiritkumar Makwana R, Shetty V, Mukherjee S, Narayan P. Cardiovascular diseases and the heart-gut cross talk. Indian Heart J 2024; 76:94-100. [PMID: 38070671 PMCID: PMC11143509 DOI: 10.1016/j.ihj.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/20/2023] [Accepted: 12/02/2023] [Indexed: 06/01/2024] Open
Abstract
The purpose of this narrative review is to provide a comprehensive overview of current research on heart-gut cross talk and its implications for cardiovascular disease. To uncover relevant preclinical and clinical research examining heart-gut cross talk, a thorough literature search was undertaken utilising electronic databases. The chosen publications were critically examined, and major findings were synthesised to offer a thorough perspective on the subject. We want to synthesise the most recent study findings, explain the underlying mechanisms, and provide potential treatment techniques. By exploring bidirectional connection between the heart and the gut, we shed light on novel future options for the prevention and treatment of cardiovascular diseases. The heart-gut cross talk is an exciting field of study with implications for cardiovascular disease. Understanding the complex connection between the heart and the gastrointestinal tract may lead to the development of novel therapeutic targets and therapies for the prevention and management of cardiovascular diseases. Future research should concentrate on identifying the specific processes driving this crosstalk as well as assessing the efficacy of therapies targeting the gut microbiota and the gut-brain axis in improving cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Varun Shetty
- Department of Cardiac Surgery, Narayana Health, India
| | | | | |
Collapse
|
6
|
Zhu Y, Cai PJ, Dai HC, Xiao YH, Jia CL, Sun AD. Black chokeberry ( Aronia melanocarpa L.) polyphenols attenuate obesity-induced colonic inflammation by regulating gut microbiota and the TLR4/NF-κB signaling pathway in high fat diet-fed rats. Food Funct 2023; 14:10014-10030. [PMID: 37840453 DOI: 10.1039/d3fo02177g] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
This study investigated the potential benefits of black chokeberry polyphenol (BCP) supplementation on lipopolysaccharide (LPS)-stimulated inflammatory response in RAW264.7 cells and obesity-induced colonic inflammation in a high fat diet (HFD)-fed rat model. Our findings demonstrated that BCP treatment effectively reduced the production of nitric oxide (NO) and pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and MCP-1) in LPS-induced RAW264.7 cells and concurrently mitigated oxidative stress by modulating the levels of malondialdehyde (MDA), catalase (CAT), and glutathione peroxidase (GSH-Px) in a dose-dependent manner. Furthermore, BCP supplementation significantly ameliorated HFD-induced obesity, improved glucose tolerance, and reduced systemic inflammation in HFD-fed rats. Notably, BCP treatment suppressed the mRNA expression of pro-inflammatory cytokines and alleviated intestinal barrier dysfunction by regulating the mRNA and protein expression of key tight junction proteins (ZO-1, occludin, and claudin-1), thereby inhibiting colonic inflammation caused by the TLR4/NF-κB signaling pathway. Additionally, BCP treatment altered the composition and function of the gut microbiota, leading to an increase in the total content of short-chain fatty acids (SCFAs), particularly acetic acid, propionic acid, isobutyric acid, and butyric acid. Collectively, our results highlighted the potential of BCP supplementation as a promising prebiotic strategy for treating obesity-induced colonic inflammation.
Collapse
Affiliation(s)
- Yue Zhu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China.
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, P.R. China
| | - Peng-Ju Cai
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China.
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, P.R. China
| | - Han-Chu Dai
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yu-Hang Xiao
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China.
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, P.R. China
| | - Cheng-Li Jia
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China.
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, P.R. China
| | - Ai-Dong Sun
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China.
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, P.R. China
| |
Collapse
|
7
|
Asami S, Tsutsui Y, Yamamoto S, Miyajima M. T-cell deficiency induces deficits in social behavior and dyslipidemia in mice. Biochem Biophys Res Commun 2023; 648:81-86. [PMID: 36739817 DOI: 10.1016/j.bbrc.2023.01.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Patients with neuropsychiatric disorders often exhibit an altered metabolic status. However, the underlying factors that induce behavioral and metabolic dysfunctions remain poorly understood. Therefore, we investigated whether behavioral and metabolic alterations could be induced in immunodeficient conditions. We found that T-cell-deficient Cd3e-/- mice exhibit deficits in social behavior associated with dyslipidemia. Cd3e-/- mice exhibited abnormal social novelty preference, but normal anxiety-like behavior. We also detected decreases in the concentrations of plasma triglyceride and the lipid transporter molecule fatty acid-binding protein 2. Furthermore, the adoptive transfer of T-cells to Cd3e-/- mice ameliorated the deficits in social behavior and recovered plasma triglyceride concentration. Thus, we found that T-cell disruption can induce defects in social behavior and systemic lipid homeostasis in mice. Given these findings, we believe that Cd3e-/- mice represent a useful tool for investigating the mechanisms of causal relationships among immune dysfunction, behavior, and metabolism.
Collapse
Affiliation(s)
- Shohei Asami
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan; Division for Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, 2669, Yamazaki, Noda, 278-0022, Japan
| | - Yumi Tsutsui
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Sachiko Yamamoto
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Michio Miyajima
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan.
| |
Collapse
|
8
|
Yao D, Wu M, Dong Y, Ma L, Wang X, Xu L, Yu Q, Zheng X. In vitro fermentation of fructooligosaccharide and galactooligosaccharide and their effects on gut microbiota and SCFAs in infants. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
9
|
Gut Microbiota Mediates Skin Ulceration Syndrome Outbreak by Readjusting Lipid Metabolism in Apostichopus japonicus. Int J Mol Sci 2022; 23:ijms232113583. [DOI: 10.3390/ijms232113583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/13/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
The intestinal tract is the most important location for symbiotes and pathogens, and the microbiota plays a crucial role in affecting the health of the gut and other host organs. Dysbacteriosis in the intestinal system has been proven to be significant in skin ulceration syndrome (SUS) in sea cucumbers. This study investigates whether the gut microbiota and lipid metabolites are relevant to the initiation and progression of SUS in a Vibrio-splendidus-infected sea cucumber model. The tight junction genes were downregulated and the inflammatory factor gene transcriptions were upregulated after V. splendidus infection in the intestinal tissue of the sea cucumber. V. splendidus infection modulated the gut microbiota by interacting with Psychromonas macrocephali, Propionigenium maris, Bacillus cereus, Lutibacter flavus, and Hoeflea halophila. Meanwhile, the metabolites of the long-chain fatty acids in the intestinal tissue, including triglycerides (TG), phosphatidylethanolamines (PE), and phosphatidylglycerols (PG), were altered after V. splendidus infection. V. splendidus engaged in positive interactions with PG and PE and negative interactions with specific TG. These results related to gut microbiota and metabolites can offer practical assistance in the identification of the inflammatory mechanisms related to SUS, and this study may serve as a reference for predicting the disease.
Collapse
|
10
|
Gut Microbiota, the Potential Biological Medicine for Prevention, Intervention and Drug Sensitization to Fight Diseases. Nutrients 2022; 14:nu14204220. [PMID: 36296908 PMCID: PMC9610464 DOI: 10.3390/nu14204220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
As the largest “immune organ” of human beings, the gut microbiota is symbiotic and mutually beneficial with the human host, playing multiple physiological functions. Studies have long shown that dysbiosis of gut microbiota is associated with almost all human diseases, mainly including type II diabetes, cancers, neurodegenerative diseases, autism spectrum disorder, and kidney diseases. As a novel and potential biological medicine for disease prevention, intervention and drug sensitization, the gut microbiota has attracted more and more attention recently. Although the gut microbiota is a comprehensive microbial community, several star bacteria have emerged as possible tools to fight against various diseases. This review aims to elucidate the relevance of gut microbiota dysbiosis with disease occurrence and progression, and mainly summarizes four well-known genera with therapeutic and sensitizing potential, Akkermansia, Bifidobacterium, Lactobacillus and Parabacteroides, thoroughly elucidate their potential value as biological drugs to treat diverse disease.
Collapse
|
11
|
Li Q, Li N, Cai W, Xiao M, Liu B, Zeng F. Fermented natural product targeting gut microbiota regulate immunity and anti-inflammatory activity: A possible way to prevent COVID-19 in daily diet. J Funct Foods 2022; 97:105229. [PMID: 36034155 PMCID: PMC9393180 DOI: 10.1016/j.jff.2022.105229] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Low immune function makes the body vulnerable to being invaded by external bacteria or viruses, causing influenza and inflammation of various organs, and this trend is shifting to the young and middle-aged group. It has been pointed out that natural products fermented by probiotic have benign changes about their active ingredients in some studies, and it have shown strong nutritional value in anti-oxidation, anti-aging, regulating lipid metabolism, anti-inflammatory and improving immunity. In recent years, the gut microbiota plays a key role and has been extensively studied in improving immunity and anti-inflammation activity. By linking the relationship between natural products fermented by probiotic, gut microbiota, immunity, and inflammation, this review presents the modulating effects of probiotics and their fermented natural products on the body, including immunity-enhancing and anti-inflammatory activities by modulating gut microbiota, and it is discussed that the current understanding of its molecular mechanisms. It may become a possible way to prevent COVID-19 through consuming natural products fermented by probiotic in our daily diet.
Collapse
Affiliation(s)
- Quancen Li
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Na Li
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenwen Cai
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Meifang Xiao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Feng Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
12
|
Zhang B, Qin S, Wu Y, Zhang R, Xu Y, Yang C. Rhamnolipids Regulate Lipid Metabolism, Immune Response, and Gut Microbiota in Rats. Front Nutr 2022; 9:886256. [PMID: 35571898 PMCID: PMC9096903 DOI: 10.3389/fnut.2022.886256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Gut microbes influence lipid metabolism and immune responses that are key features of metabolic disorders. This study examined effects of bacterial rhamnolipids (RLS) on lipid metabolism, immune response, and gut microbiota in rats. Methods Twenty-four Sprague-Dawley rats were randomly divided into three groups and gavage-fed for seven weeks with normal saline (NCO group), 50 mg/kg bw RLS (RLS1 group), and 100 mg/kg bw RLS (RLS2 group). Results Compared with those of the NCO group, the RLS1 and RLS2 groups showed significantly decreased fat weight, relative fat weight, and adipocyte size (P < 0.05). Furthermore, RLS1 and RLS2 significantly decreased concentrations of triglycerides, low-density lipoprotein-cholesterol, and non-esterified fatty acids and increased high-density lipoprotein-cholesterol levels (P < 0.05). However, the total cholesterol content among the three groups (P > 0.05) were not significantly different. Serum concentrations of interleukin-1β, interleukin-6, and tumor necrosis factor-α were significantly lower in the RLS2 group than those in the NCO group (P < 0.05). The relative mRNA expression of fatty acid synthase was significantly decreased, while those of carnitine palmitoyltransferase-1, carnitine palmitoyltransferase-2, and peroxisome proliferator-activated receptor-gamma coactivator-1α were significantly increased in the RLS2 group compared with those in the NCO group (P < 0.05). Moreover, the relative abundances of Lactobacillus, Roseburia, Ruminococcus-1, and Parabacteroides were significantly higher in the RLS2 group than those in the NCO group (P < 0.05). Conclusion Our findings suggest that RLS reduces fat deposition, inhibits inflammation, regulates intestinal flora, and promotes the proliferation of beneficial bacteria in rats.
Collapse
Affiliation(s)
- Bing Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Songke Qin
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Yanping Wu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Ruiqiang Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Yinglei Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Caimei Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| |
Collapse
|
13
|
Fernández-García V, González-Ramos S, Martín-Sanz P, Laparra JM, Boscá L. Beyond classic concepts in thyroid homeostasis: Immune system and microbiota. Mol Cell Endocrinol 2021; 533:111333. [PMID: 34048865 DOI: 10.1016/j.mce.2021.111333] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
It has long been known that thyroid hormones have implications for multiple physiological processes and can lead to serious illness when there is an imbalance in its metabolism. The connections between thyroid hormone metabolism and the immune system have been extensively described, as they can participate in inflammation, autoimmunity, or cancer progression. In addition, changes in the normal intestinal microbiota involve the activation of the immune system while triggering different pathophysiological disorders. Recent studies have linked the microbiota and certain bacterial fragments or metabolites to the regulation of thyroid hormones and the general response in the endocrine system. Even if the biology and function of the thyroid gland has attracted more attention due to its pathophysiological importance, there are essential mechanisms and issues related to it that are related to the interplay between the intestinal microbiota and the immune system and must be further investigated. Here we summarize additional information to uncover these relationships, the knowledge of which would help establish new personalized medical strategies.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain.
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Melchor Fernández Almagro 6, 28029, Madrid, Spain
| | - José M Laparra
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra. Cantoblanco 8, 28049, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain.
| |
Collapse
|
14
|
Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front Immunol 2021; 12:686501. [PMID: 34335588 PMCID: PMC8319742 DOI: 10.3389/fimmu.2021.686501] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE), often considered the prototype of autoimmune diseases, is characterized by over-activation of the autoimmune system with abnormal functions of innate and adaptive immune cells and the production of a large number of autoantibodies against nuclear components. Given the highly complex and heterogeneous nature of SLE, the pathogenesis of this disease remains incompletely understood and is presumed to involve both genetic and environmental factors. Currently, disturbance of the gut microbiota has emerged as a novel player involved in the pathogenesis of SLE. With in-depth research, the understanding of the intestinal bacteria-host interaction in SLE is much more comprehensive. Recent years have also seen an increase in metabolomics studies in SLE with the attempt to identify potential biomarkers for diagnosis or disease activity monitoring. An intricate relationship between gut microbiome changes and metabolic alterations could help explain the mechanisms by which gut bacteria play roles in the pathogenesis of SLE. Here, we review the role of microbiota dysbiosis in the aetiology of SLE and how intestinal microbiota interact with the host metabolism axis. A proposed treatment strategy for SLE based on gut microbiome (GM) regulation is also discussed in this review. Increasing our understanding of gut microbiota and their function in lupus will provide us with novel opportunities to develop effective and precise diagnostic strategies and to explore potential microbiota-based treatments for patients with lupus.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Yang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongkang Wu
- Department of Laboratory Medicine and Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Sun Y, Ni A, Jiang Y, Li Y, Huang Z, Shi L, Xu H, Chen C, Li D, Han Y, Chen J. Effects of Replacing In-feed Antibiotics with Synergistic Organic Acids on Growth Performance, Health, Carcass, and Immune and Oxidative Statuses of Broiler Chickens Under Clostridium perfringens Type A Challenge. Avian Dis 2021; 64:393-400. [PMID: 33205169 DOI: 10.1637/aviandiseases-d-19-00101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/06/2020] [Indexed: 11/05/2022]
Abstract
This study was conducted to investigate the effects of replacing in-feed antibiotics with synergistic organic acids on growth performance, health, carcass, and immune and oxidative statuses of broiler chickens under Clostridium perfringens (CP) type A challenge. Two organic acid products were tested: organic acid 1 (OA1), consisting of butyrate, medium-chain fatty acids, organic acids, and phenolics; and organic acid 2 (OA2), consisting of buffered short-chain fatty acids. Six hundred 1-day-old male Arbor Acres broiler chicks were randomly assigned to one of five treatments: Control 1, basal diet, nonchallenged birds; Control 2, basal diet, with CP challenge; antimicrobial growth promoters (AGP), basal diet supplemented with Aureomycin (chlortetracycline), with CP challenge; OA1, basal diet supplemented with OA1, with CP challenge; and OA1OA2, basal diet supplemented with OA1 and OA2, with CP challenge. Each treatment had eight replicate pens of 15 birds. The experiments lasted for 29 days. The disease challenge was performed on days 15-17, with an oral gavage of 0.5 mL of CP culture (2.0 × 108 colony-forming units [CFU]/mL) for each bird. Body weights (BWs), intestinal lesion scores, immune organ indices, and serum malondialdehyde (MDA) concentrations were measured on days 19, 22, and 29, respectively, in three birds per pen. Carcass characteristics were determined on day 29. No treatment-related differences in mortality were noted before (P = 0.28) or after (P = 0.64) challenge or over the whole study period (days 0-28; P = 0.66). On day 19, the BW of Control 2 was lower than other treatments (P < 0.0001). On day 22, AGP, OA1, and OA1OA2 had higher BW than Control 2 (P = 0.001). The breast muscle yield of OA1 and OA1OA2 was higher than AGP (P < 0.05). The abdominal fat yield of OA1OA2 was lower than AGP and Control 2 (P < 0.05). On day 22, the birds fed OA1OA2 showed lower intestinal lesion scores than OA1 (P < 0.05). No treatment-related differences in immune organ (spleen, thymus, and bursa) indices were noted (P > 0.05). On day 29, the MDA concentration of OA1 and OA1OA2 was lower than those of Control 1 and AGP (P < 0.05). In conclusion, the addition of organic acids may protect broiler chickens from severe intestinal lesions and oxidative stress and may help reduce abdominal fat mass deposition. There is potential for organic acid-based products as alternatives for AGP in preventing necrotic enteritis in broilers.
Collapse
Affiliation(s)
- Yanyan Sun
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Aixin Ni
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Ying Jiang
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Yunlei Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Ziyan Huang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Lei Shi
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Hong Xu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Chao Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Dongli Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Yanming Han
- Trouw Nutrition R & D, Stationsstraat 77, 3811 MH, Amersfoort, the Netherlands
| | - Jilan Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| |
Collapse
|
16
|
Voidarou C, Antoniadou M, Rozos G, Tzora A, Skoufos I, Varzakas T, Lagiou A, Bezirtzoglou E. Fermentative Foods: Microbiology, Biochemistry, Potential Human Health Benefits and Public Health Issues. Foods 2020; 10:E69. [PMID: 33396397 PMCID: PMC7823516 DOI: 10.3390/foods10010069] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
Fermented foods identify cultures and civilizations. History, climate and the particulars of local production of raw materials have urged humanity to exploit various pathways of fermentation to produce a wide variety of traditional edible products which represent adaptations to specific conditions. Nowadays, industrial-scale production has flooded the markets with ferments. According to recent estimates, the current size of the global market of fermented foods is in the vicinity of USD 30 billion, with increasing trends. Modern challenges include tailor-made fermented foods for people with special dietary needs, such as patients suffering from Crohn's disease or other ailments. Another major challenge concerns the safety of artisan fermented products, an issue that could be tackled with the aid of molecular biology and concerns not only the presence of pathogens but also the foodborne microbial resistance. The basis of all these is, of course, the microbiome, an aggregation of different species of bacteria and yeasts that thrives on the carbohydrates of the raw materials. In this review, the microbiology of fermented foods is discussed with a special reference to groups of products and to specific products indicative of the diversity that a fermentation process can take. Their impact is also discussed with emphasis on health and oral health status. From Hippocrates until modern approaches to disease therapy, diet was thought to be of the most important factors for health stability of the human natural microbiome. After all, to quote Pasteur, "Gentlemen, the microbes will have the last word for human health." In that sense, it is the microbiomes of fermented foods that will acquire a leading role in future nutrition and therapeutics.
Collapse
Affiliation(s)
- Chrysa Voidarou
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, School of Agriculture, University of Ioannina, 47132 Arta, Greece; (C.V.); (A.T.); (I.S.)
| | - Maria Antoniadou
- School of Dentistry, National and Kapodistrian University of Athens, 11521 Athens, Greece;
| | - Georgios Rozos
- Laboratory of Microbiology, Biotechnology & Hygiene, Department of Agricultural Development, Democritus University of Thrace, 68200 Orestiada, Greece;
| | - Athina Tzora
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, School of Agriculture, University of Ioannina, 47132 Arta, Greece; (C.V.); (A.T.); (I.S.)
| | - Ioannis Skoufos
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, School of Agriculture, University of Ioannina, 47132 Arta, Greece; (C.V.); (A.T.); (I.S.)
| | - Theodoros Varzakas
- Department of Food Science and Technology, University of the Peloponnese, 24100 Kalamata, Greece
| | - Areti Lagiou
- Department of Public and Community Health, University of West Attika, 11521 Athens, Greece;
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
17
|
Zhang L, Zhou W, Zhan L, Hou S, Zhao C, Bi T, Lu X. Fecal microbiota transplantation alters the susceptibility of obese rats to type 2 diabetes mellitus. Aging (Albany NY) 2020; 12:17480-17502. [PMID: 32920548 PMCID: PMC7521520 DOI: 10.18632/aging.103756] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/06/2020] [Indexed: 01/24/2023]
Abstract
Obesity is one of the susceptibility factors for type 2 diabetes (T2DM), both of which could accelerate the aging of the body and bring many hazards. A causal relationship is present between intestinal microbiota and body metabolism, but how the microbiota play a role in the progression of obesity to T2DM has not been elucidated. In this study, we transplanted healthy or obese-T2DM intestinal microbiota to ZDF and LZ rats, and used 16S rRNA and targeted metabonomics to evaluate the directional effect of the microbiota on the susceptibility of obese rats to T2DM. The glycolipid metabolism phenotype could be changed bidirectionally in obese rats instead of in lean ones. One possible mechanism is that the microbiota and metabolites alter the structure of the intestinal tract, and improve insulin and leptin resistance through JAK2 / IRS / Akt pathway. It is worth noting that 7 genera, such as Lactobacillus, Clostridium and Roche, can regulate 15 metabolites, such as 3-indolpropionic acid, acetic acid and docosahexaenoic acid, and have a significant improvement on glycolipid metabolism phenotype. Attention to intestinal homeostasis may be the key to controlling obesity and preventing T2DM.
Collapse
Affiliation(s)
- Lijing Zhang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wen Zhou
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Libin Zhan
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shenglin Hou
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chunyan Zhao
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingting Bi
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoguang Lu
- Department of Emergency Medicine, Zhongshan Hospital, Dalian University, Dalian 116001, China
| |
Collapse
|
18
|
van Schewick CM, Nöltner C, Abel S, Burns SO, Workman S, Symes A, Guzman D, Proietti M, Bulashevska A, Moreira F, Soetedjo V, Lowe DM, Grimbacher B. Altered Microbiota, Impaired Quality of Life, Malabsorption, Infection, and Inflammation in CVID Patients With Diarrhoea. Front Immunol 2020; 11:1654. [PMID: 32849570 PMCID: PMC7412961 DOI: 10.3389/fimmu.2020.01654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Diarrhoea is the commonest gastrointestinal symptom in patients with common variable immunodeficiency (CVID). Objective: The aim of this study was to describe the prevalence and clinical presentation of chronic and recurrent diarrhoea in the Royal-Free-Hospital (RFH) London CVID cohort, including symptoms, infections, level of inflammation, and microbial diversity. Methods: A cross-sectional study of adult CVID patients (139 out of 172 diagnosed with CVID completed the screening questionnaire). Those with diarrhoea ≥6 days/month had stool and blood samples analysed and completed the short Inflammatory Bowel Disease Questionnaire (sIBDQ). BMI, spleen-size, lymphocytes and gut-microbial diversity were compared. Due to logistical and clinical restraints, not all patients could be analysed on all measures. Results: 46/139 (33.1%) patients had current significant diarrhoea. In patients with past or present diarrhoea, BMI was lower (median 23.7 vs. 26, p = 0.005), malabsorption more common (57.97 vs. 35.71%, p = 0.011). CD4+ lymphocytes were higher in patients with diarrhoea (p = 0.028; n = 138), but CD4+ naïve lymphocytes were significantly higher in non-diarrhoea patients (p = 0.009, N = 28). Nine patients had confirmed or probable current gastrointestinal infections. Calprotectin was >60 μg/g in 13/29 with significant diarrhoea including 9 without infection. SIBDQ revealed a low median score of 4.74. Microbial alpha diversity was significantly lower in CVID patients compared to healthy household controls. There was no significant difference in alpha diversity in relation to antibiotic intake during the 6 weeks prior to providing samples. Conclusion: Patients with CVID and significant diarrhoea had infections, raised calprotectin, malabsorption, a lower BMI, an impaired quality of life (comparable to active IBD), and they differed from non-diarrhoea patients in their lymphocyte phenotyping. Furthermore, microbial diversity was altered. These findings strongly imply that there may be an inflammatory nature and a systemic predisposition to diarrhoea in CVID, which necessitates further investigation.
Collapse
Affiliation(s)
- Cornelia M van Schewick
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom.,Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Institute for Immunodeficiency, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Christina Nöltner
- Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Institute for Immunodeficiency, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Svenja Abel
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom.,Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Institute for Immunodeficiency, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - Sarita Workman
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - Andrew Symes
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - David Guzman
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - Michele Proietti
- Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Institute for Immunodeficiency, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Alla Bulashevska
- Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Institute for Immunodeficiency, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Fernando Moreira
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - Veronika Soetedjo
- Freiburg Center for Data Analysis and Modeling (FDM), IMBI/ZKS, Freiburg, Germany
| | - David M Lowe
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - Bodo Grimbacher
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom.,Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Institute for Immunodeficiency, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,DZIF - German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany.,CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Freiburg, Germany
| |
Collapse
|
19
|
Yue YY, Fan XY, Zhang Q, Lu YP, Wu S, Wang S, Yu M, Cui CW, Sun ZR. Bibliometric analysis of subject trends and knowledge structures of gut microbiota. World J Clin Cases 2020; 8:2817-2832. [PMID: 32742991 PMCID: PMC7360702 DOI: 10.12998/wjcc.v8.i13.2817] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/05/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gut microbiota is an emerging field of research, with related research having breakthrough development in the past 15 years. Bibliometric analysis can be applied to analyze the evolutionary trends and emerging hotspots in this field.
AIM To study the subject trends and knowledge structures of gut microbiota related research fields from 2004 to 2018.
METHODS The literature data on gut microbiota were identified and downloaded from the PubMed database. Through biclustering analysis, strategic diagrams, and social network analysis diagrams, the main trend and knowledge structure of research fields concerning gut microbiota were analyzed to obtain and compare the research hotspots in each period.
RESULTS According to the strategic coordinates and social relationship network map, Clostridium Infections/microbiology, Clostridium Infections/therapy, RNA, Ribosomal, 16S/genetics, Microbiota/genetics, Microbiota/immunology, Dysbiosis/immunology, Infla-mmation/immunology, Fecal Microbiota Transplantation/methods, Fecal Microbiota Transplantation can be used as an emerging research hotspot in the past 5 years (2014-2018).
CONCLUSION Some subjects were not yet fully studied according to the strategic coordinates; and the emerging hotspots in the social network map can be considered as directions of future research.
Collapse
Affiliation(s)
- Yuan-Yi Yue
- Department of Gastroenterology Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Xin-Yue Fan
- Student Affairs Department, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Qiang Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province
| | - Yi-Ping Lu
- BioBank, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Si Wu
- BioBank, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Shuang Wang
- BioBank, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Miao Yu
- BioBank, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Chang-Wan Cui
- BioBank, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Zheng-Rong Sun
- BioBank, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
20
|
Subramaniam R, Aliakbarian H, Bhutta HY, Harris DA, Tavakkoli A, Sheu EG. Sleeve Gastrectomy and Roux-en-Y Gastric Bypass Attenuate Pro-inflammatory Small Intestinal Cytokine Signatures. Obes Surg 2020; 29:3824-3832. [PMID: 31363962 DOI: 10.1007/s11695-019-04059-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Bariatric surgery rapidly induces improvements in type 2 diabetes (T2D) in concert with reduction in systemic markers of inflammation. The impact of bariatric surgery on local intestinal immunity is not known. We hypothesize that sleeve gastrectomy (SG) and gastric bypass (RYGB) surgeries resolve obesity-induced intestinal inflammation, thereby promoting T2D resolution. METHODS SG and RYGB, or control surgery was performed in SD rats (n = 4-6/group). Key cytokines involved in insulin resistance (TNF-α, IFN-γ), inflammasome activation (IL-1β, IL-18), inflammation resolution (IL-10, IL-33), and Th17 cell responses (IL-17, IL-23) were measured by qPCR in mucosal scrapings of jejunum at 4 weeks post-surgery. Intestinal cytokine expressions were correlated with weight change, systemic and portal glucose, and insulin levels in response to an enteral glucose load. RESULTS SG downregulated IL-17 and IL-23 in both proximal and distal jejunum, and IFN-γ was reduced only in distal jejunum (p < 0.05). Jejunal IL-17 and IL-23 expression correlated positively with weight changes after SG (0.93 and 0.98, respectively; p < 0.05). Changes in IFN-γ correlated strongly with insulin levels in portal and systemic circulation (0.99 and 0.95, respectively, p < 0.05). As with SG, IFN-γ, IL-17, and IL-23 were significantly reduced by RYGB. RYGB also reduced TNF-α and IL-18 and increased IL-33 levels (p < 0.05). CONCLUSIONS RYGB and SG reduce expression of pro-inflammatory cytokines IL-17, IL-23, and IFN-γ in the jejunum. RYGB showed attenuation of additional pro-inflammatory cytokines and enhanced expression of IL-33. Post-surgical changes in intestinal IL-17, IL-23, and IFN-γ correlate strongly with changes in weight and glucose-triggered insulin responses.
Collapse
Affiliation(s)
- Renuka Subramaniam
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hassan Aliakbarian
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hina Y Bhutta
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - David A Harris
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ali Tavakkoli
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Eric G Sheu
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
21
|
Oligosaccharides from Morinda officinalis Slow the Progress of Aging Mice by Regulating the Key Microbiota-Metabolite Pairs. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9306834. [PMID: 31929824 PMCID: PMC6942866 DOI: 10.1155/2019/9306834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/26/2019] [Accepted: 11/27/2019] [Indexed: 11/17/2022]
Abstract
The gut microbiota is considered an important factor in the progression of Alzheimer's disease (AD). Active research on the association between the metabolome and the gut microbiome is ongoing and can provide a large amount of beneficial information about the interactions between the microbiome and the metabolome. Previous studies have shown that the oligosaccharides from Morinda officinalis (OMO) can delay the progress of AD in model animals by regulating the diversity of the gut microbiome and metabolic components, and the correlation between the gut microbiome and metabolic components still needs to be further verified. This study applied a new two-level strategy to investigate and ensure the accuracy and consistency of the results. This strategy can be used to determine the association between the gut microbiome and serum metabolome in APP/PS1 transgenic mice and C57BL/6J male mice. The “4C0d-2 spp.-Cholesterol,” “CW040 spp.-L-valine,” “CW040 spp.-L-acetylcarnitine,” “RF39 spp.-L-valine,” “TM7-3 spp.-L-valine,” and “TM7-3 spp.-L-acetylcarnitine” associations among specific “microbiota-metabolite” pairs were further identified based on univariate and multivariate correlation analyses and functional analyses. The key relevant pairs were verified by an independent oligosaccharide intervention study, and the gut microbiome and serum metabolome of the OMO intervention group were similar to those of the normal group. The results indicate that OMO can significantly suppress Alzheimer's disease by regulating the key microbiota-metabolite pairs. Therefore, this two-level strategy is effective in identifying the principal correlations in large datasets obtained from combinations of multiomic studies and further enhancing our understanding of the correlation between the brain and gut in patients with AD.
Collapse
|
22
|
Sakwinska O, Bosco N. Host Microbe Interactions in the Lactating Mammary Gland. Front Microbiol 2019; 10:1863. [PMID: 31456777 PMCID: PMC6701204 DOI: 10.3389/fmicb.2019.01863] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
The bacteria present in human milk constitute the human milk microbiome (hMM). Both the older culture-based work and the more recent studies using molecular detection of bacterial DNA have reached similar conclusions: the hMM mostly consists of commensal staphylococci such as Staphylococcus epidermidis, and streptococci. The prevalence of other bacterial groups such lactobacilli varies widely, while the abundance and prevalence of bifidobacteria is generally low. Recently, the hMM became accepted as a part of a physiologically normal state with suggested potential health benefits. Most research on the hMM has focused on its composition and potential effect on the breastfed infant. A major role as a microbiome inoculum for the infant gut has been proposed, but remains to be clearly demonstrated. Herein, we also discuss the emerging connection between the hMM and mammary gland physiology and lactation. Similarities between the mammary gland and mucosal interfaces are considerable, and in particular mucosal-like immune attributes of mammary gland. The potential role of hMM-host interactions in the mammary gland in maternal health is explored with a primary focus on lactational mastitis.
Collapse
Affiliation(s)
- Olga Sakwinska
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Nestlé Research Singapore Hub, Singapore, Singapore
| |
Collapse
|
23
|
Chen H, Zeng F, Li S, Liu Y, Gong S, Lv X, Zhang J, Liu B. Spirulina active substance mediated gut microbes improve lipid metabolism in high-fat diet fed rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
24
|
Zhai Q, Xiao Y, Li P, Tian F, Zhao J, Zhang H, Chen W. Varied doses and chemical forms of selenium supplementation differentially affect mouse intestinal physiology. Food Funct 2019; 10:5398-5412. [DOI: 10.1039/c9fo00278b] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Varied doses and chemical forms of selenium supplementation differentially affect mouse intestinal physiology and perturbed the fecal metabolic profiles of and jejunal protein expression in mice.
Collapse
Affiliation(s)
- Qixiao Zhai
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Yue Xiao
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Peng Li
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- National Engineering Research Center for Functional Food
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Wei Chen
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| |
Collapse
|
25
|
Oral consumption of cinnamon enhances the expression of immunity and lipid absorption genes in the small intestinal epithelium and alters the gut microbiota in normal mice. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.08.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
26
|
Chen T, You Y, Xie G, Zheng X, Zhao A, Liu J, Zhao Q, Wang S, Huang F, Rajani C, Wang C, Chen S, Ni Y, Yu H, Deng Y, Wang X, Jia W. Strategy for an Association Study of the Intestinal Microbiome and Brain Metabolome Across the Lifespan of Rats. Anal Chem 2018; 90:2475-2483. [PMID: 29353471 DOI: 10.1021/acs.analchem.7b02859] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is increased appreciation for the diverse roles of the microbiome-gut-brain axis on mammalian growth and health throughout the lifespan. Numerous studies have demonstrated that the gut microbiome and their metabolites are extensively involved in the communication between brain and gut. Association study of brain metabolome and gut microbiome is an active field offering large amounts of information on the interaction of microbiome, brain and gut but data size and complicated hierarchical relationships were found to be major obstacles to the formation of significant, reproducible conclusions. This study addressed a two-level strategy of brain metabolome and gut microbiome association analysis of male Wistar rats in the process of growth, employing several analytical platforms and various bioinformatics methods. Trajectory analysis showed that the age-related brain metabolome and gut microbiome had similarity in overall alteration patterns. Four high taxonomical level correlated pairs of "metabolite type-bacterial phylum", including "lipids-Spirochaetes", "free fatty acids (FFAs)-Firmicutes", "bile acids (BAs)-Firmicutes", and "Neurotransmitters-Bacteroidetes", were screened out based on unit- and multivariant correlation analysis and function analysis. Four groups of specific "metabolite-bacterium" association pairs from within the above high level key pairs were further identified. The key correlation pairs were validated by an independent animal study. This two-level strategy is effective in identifying principal correlations in big data sets obtained from the systematic multiomics study, furthering our understanding on the lifelong connection between brain and gut.
Collapse
Affiliation(s)
- Tianlu Chen
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Yijun You
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Guoxiang Xie
- University of Hawaii Cancer Center , Honolulu, Hawaii 96813, United States
| | - Xiaojiao Zheng
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Aihua Zhao
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Jiajian Liu
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Qing Zhao
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Shouli Wang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Fengjie Huang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China
| | - Cynthia Rajani
- University of Hawaii Cancer Center , Honolulu, Hawaii 96813, United States
| | - Congcong Wang
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Shaoqiu Chen
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Yan Ni
- University of Hawaii Cancer Center , Honolulu, Hawaii 96813, United States
| | - Herbert Yu
- University of Hawaii Cancer Center , Honolulu, Hawaii 96813, United States
| | - Youping Deng
- Biostatistics and Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa , Honolulu, Hawaii 96813, United States
| | - Xiaoyan Wang
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Wei Jia
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233, China.,University of Hawaii Cancer Center , Honolulu, Hawaii 96813, United States
| |
Collapse
|
27
|
Pan YY, Zeng F, Guo WL, Li TT, Jia RB, Huang ZR, Lv XC, Zhang J, Liu B. Effect of Grifola frondosa 95% ethanol extract on lipid metabolism and gut microbiota composition in high-fat diet-fed rats. Food Funct 2018; 9:6268-6278. [DOI: 10.1039/c8fo01116h] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This study aimed to investigate the effects of 95% ethanol extract of G. frondosa (GF95) on lipid metabolism and gut microbiota composition in high-fat diet (HFD) fed rats.
Collapse
Affiliation(s)
- Yu-Yang Pan
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
| | - Feng Zeng
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
| | - Wei-Ling Guo
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
| | - Tian-Tian Li
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
| | - Rui-Bo Jia
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
| | - Zi-Rui Huang
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
| | - Xu-Cong Lv
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
- National Engineering Research Center of JUNCAO Technology
| | - Jiachao Zhang
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
- College of Food Science of Technology
| | - Bin Liu
- College of Food Science
- Fujian Agriculture and Forestry University
- Fuzhou
- China
- National Engineering Research Center of JUNCAO Technology
| |
Collapse
|
28
|
Rodrigues RR, Greer RL, Dong X, DSouza KN, Gurung M, Wu JY, Morgun A, Shulzhenko N. Antibiotic-Induced Alterations in Gut Microbiota Are Associated with Changes in Glucose Metabolism in Healthy Mice. Front Microbiol 2017; 8:2306. [PMID: 29213261 PMCID: PMC5702803 DOI: 10.3389/fmicb.2017.02306] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome plays an important role in health and disease. Antibiotics are known to alter gut microbiota, yet their effects on glucose tolerance in lean, normoglycemic mice have not been widely investigated. In this study, we aimed to explore mechanisms by which treatment of lean mice with antibiotics (ampicillin, metronidazole, neomycin, vancomycin, or their cocktail) influences the microbiome and glucose metabolism. Specifically, we sought to: (i) study the effects on body weight, fasting glucose, glucose tolerance, and fasting insulin, (ii) examine the changes in expression of key genes of the bile acid and glucose metabolic pathways in the liver and ileum, (iii) identify the shifts in the cecal microbiota, and (iv) infer interactions between gene expression, microbiome, and the metabolic parameters. Treatment with individual or a cocktail of antibiotics reduced fasting glucose but did not affect body weight. Glucose tolerance changed upon treatment with cocktail, ampicillin, or vancomycin as indicated by reduced area under the curve of the glucose tolerance test. Antibiotic treatment changed gene expression in the ileum and liver, and shifted the alpha and beta diversities of gut microbiota. Network analyses revealed associations between Akkermansia muciniphila with fasting glucose and liver farsenoid X receptor (Fxr) in the top ranked host-microbial interactions, suggesting possible mechanisms by which this bacterium can mediate systemic changes in glucose metabolism. We observed Bacteroides uniformis to be positively and negatively correlated with hepatic Fxr and Glucose 6-phosphatase, respectively. Overall, our transkingdom network approach is a useful hypothesis generating strategy that offers insights into mechanisms by which antibiotics can regulate glucose tolerance in non-obese healthy animals. Experimental validation of our predicted microbe-phenotype interactions can help identify mechanisms by which antibiotics affect host phenotypes and gut microbiota.
Collapse
Affiliation(s)
- Richard R. Rodrigues
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Renee L. Greer
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, United States
| | - Xiaoxi Dong
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Karen N. DSouza
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Manoj Gurung
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, United States
| | - Jia Y. Wu
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, United States
| | - Andrey Morgun
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Natalia Shulzhenko
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
29
|
Mooney D, Edgar D, Einarsson G, Downey D, Elborn S, Tunney M. Chronic lung disease in common variable immune deficiency (CVID): A pathophysiological role for microbial and non-B cell immune factors. Crit Rev Microbiol 2017; 43:508-519. [PMID: 28068853 DOI: 10.1080/1040841x.2016.1268568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
One of the most common and most severe forms of primary antibody deficiency encountered in the clinical setting is a heterogeneous group of syndromes termed common variable immune deficiency (CVID). This disorder is characterized by reduced immunoglobulin production and increased susceptibility to infection, particularly of the respiratory tract. Infection and subsequent immunological/inflammatory processes may contribute to the development of pulmonary complications such as bronchiectasis and interstitial lung disease. Immunoglobulin replacement and/or antibiotic therapy, to prevent infection, are routinely prescribed treatments. However, chronic lung disease, the major cause of morbidity and mortality in this patient cohort, may still progress. This clinical progression suggests that pathogens recalcitrant to currently prescribed treatments and other immunological defects may be contributing to the development of pulmonary disease. This review describes the potential role of microbiological and non-B cell immunological factors, including T-cells, neutrophils, complement, toll like receptors, and antimicrobial peptides, in the pathogenicity of chronic lung disease in patients with CVID.
Collapse
Affiliation(s)
- Denver Mooney
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- b Centre for Experimental Medicine, School of Medicine , Dentistry and Biomedical Sciences. Queen's University Belfast , Belfast , United Kingdom
| | - David Edgar
- c T he Royal Hospitals, Belfast Health and Social Care Trust , Regional Immunology Service , Belfast , United Kingdom
| | - Gisli Einarsson
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- b Centre for Experimental Medicine, School of Medicine , Dentistry and Biomedical Sciences. Queen's University Belfast , Belfast , United Kingdom
| | - Damian Downey
- d Belfast City Hospital, Belfast Health and Social Care Trust , Regional Respiratory Centre , Belfast , United Kingdom
| | - Stuart Elborn
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- b Centre for Experimental Medicine, School of Medicine , Dentistry and Biomedical Sciences. Queen's University Belfast , Belfast , United Kingdom
| | - Michael Tunney
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- e School of Pharmacy , Queen's University Belfast , Belfast , United Kingdom
| |
Collapse
|
30
|
Lewis MC, Merrifield CA, Berger B, Cloarec O, Duncker S, Mercenier A, Nicholson JK, Holmes E, Bailey M. Early intervention with Bifidobacterium lactis NCC2818 modulates the host-microbe interface independent of the sustained changes induced by the neonatal environment. Sci Rep 2017; 7:5310. [PMID: 28706260 PMCID: PMC5509696 DOI: 10.1038/s41598-017-05689-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/31/2017] [Indexed: 12/26/2022] Open
Abstract
Inflammatory and metabolic diseases can originate during early-life and have been correlated with shifts in intestinal microbial ecology. Here we demonstrate that minor environmental fluctuations during the early neonatal period had sustained effects on the developing porcine microbiota and host-microbe interface. These inter-replicate effects appear to originate during the first day of life, and are likely to reflect very early microbiota acquisition from the environment. We statistically link early systemic inflammation with later local increases in inflammatory cytokine (IL-17) production, which could have important enteric health implications. Immunity, intestinal barrier function, host metabolism and host-microbiota co-metabolism were further modified by Bifidobacterium lactis NCC2818 supplementation, although composition of the in situ microbiota remained unchanged. Finally, our robust model identified novel, strong correlations between urinary metabolites (eg malonate, phenylacetylglycine, alanine) and mucosal immunoglobulin (IgM) and cytokine (IL-10, IL-4) production, thus providing the possibility of the development of urinary ‘dipstick’ tests to assess non-accessible mucosal immune development and identify early precursors (biomarkers) of disease. These results have important implications for infants exposed to neonatal factors including caesarean delivery, antibiotic therapy and delayed discharge from hospital environments, which may predispose to the development of inflammatory and metabolic diseases in later life.
Collapse
Affiliation(s)
- Marie C Lewis
- Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading, RG6 6AP, UK.
| | - Claire A Merrifield
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, SW7 2AZ, London, UK
| | - Bernard Berger
- Nestlé Research Centre, Vers-chez-les-Blanc, 1000, Lausanne 26, Switzerland
| | | | - Swantje Duncker
- Nestlé Research Centre, Vers-chez-les-Blanc, 1000, Lausanne 26, Switzerland
| | - Annick Mercenier
- Nestlé Research Centre, Vers-chez-les-Blanc, 1000, Lausanne 26, Switzerland
| | - Jeremy K Nicholson
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, SW7 2AZ, London, UK
| | - Elaine Holmes
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, SW7 2AZ, London, UK
| | - Mick Bailey
- Infection and Immunity, Department of Clinical Veterinary Science, University of Bristol, Langford House, Langford, North Somerset, BS40 5DU, UK
| |
Collapse
|
31
|
Vyshenska D, Lam KC, Shulzhenko N, Morgun A. Interplay between viruses and bacterial microbiota in cancer development. Semin Immunol 2017; 32:14-24. [PMID: 28602713 DOI: 10.1016/j.smim.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/03/2017] [Accepted: 05/30/2017] [Indexed: 12/29/2022]
Abstract
During the last few decades we have become accustomed to the idea that viruses can cause tumors. It is much less considered and discussed, however, that most people infected with oncoviruses will never develop cancer. Therefore, the genetic and environmental factors that tip the scales from clearance of viral infection to development of cancer are currently an area of active investigation. Microbiota has recently emerged as a potentially critical factor that would affect this balance by increasing or decreasing the ability of viral infection to promote carcinogenesis. In this review, we provide a model of microbiome contribution to the development of oncogenic viral infections and viral associated cancers, give examples of this process in human tumors, and describe the challenges that prevent progress in the field as well as their potential solutions.
Collapse
Affiliation(s)
- Dariia Vyshenska
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA
| | - Khiem C Lam
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Oregon State University, 208 Dryden Hall, Corvallis, OR 97331, USA.
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA.
| |
Collapse
|
32
|
Abstract
The three common themes that underlie the induction and perpetuation of autoimmunity are genetic predisposition, environmental factors, and immune regulation. Environmental factors have gained much attention for their role in triggering autoimmunity, with increasing evidence of their influence as demonstrated by epidemiological studies, laboratory research, and animal studies. Environmental factors known to trigger and perpetuate autoimmunity include infections, gut microbiota, as well as physical and environmental agents. To address these issues, we will review major potential mechanisms that underlie autoimmunity including molecular mimicry, epitope spreading, bystander activation, polyclonal activation of B and T cells, infections, and autoinflammatory activation of innate immunity. The association of the gut microbiota on autoimmunity will be particularly highlighted by their interaction with pharmaceutical agents that may lead to organ-specific autoimmunity. Nonetheless, and we will emphasize this point, the precise mechanism of environmental influence on disease pathogenesis remains elusive.
Collapse
Affiliation(s)
- Annarosa Floreani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Patrick S C Leung
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy
| | - M Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
33
|
Khedkar CD, Kalyankar SD, Patel AR, Khedkar CC. Effect of Synbiotic-Assisted Modulation of Gastrointestinal Microbiota on Human Health. NUTRIENTS IN DAIRY AND THEIR IMPLICATIONS ON HEALTH AND DISEASE 2017:223-236. [DOI: 10.1016/b978-0-12-809762-5.00018-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
34
|
Mattner J. Impact of Microbes on the Pathogenesis of Primary Biliary Cirrhosis (PBC) and Primary Sclerosing Cholangitis (PSC). Int J Mol Sci 2016; 17:1864. [PMID: 27834858 PMCID: PMC5133864 DOI: 10.3390/ijms17111864] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 02/07/2023] Open
Abstract
Primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC) represent the major clinical entities of chronic cholestatic liver diseases. Both disorders are characterized by portal inflammation and slowly progress to obliterative fibrosis and eventually liver cirrhosis. Although immune-pathogenic mechanisms have been implicated in the pathogenesis of PBC and PSC, neither disorder is considered to be a classical autoimmune disease, as PSC and PBC patients do not respond to immune-suppressants. Furthermore, the decreased bile flow resulting from the immune-mediated tissue assault and the subsequent accumulation of toxic bile products in PBC and PSC not only perpetuates biliary epithelial damage, but also alters the composition of the intestinal and biliary microbiota and its mutual interactions with the host. Consistent with the close association of PSC and inflammatory bowel disease (IBD), the polyclonal hyper IgM response in PBC and (auto-)antibodies which cross-react to microbial antigens in both diseases, an expansion of individual microbes leads to shifts in the composition of the intestinal or biliary microbiota and a subsequent altered integrity of epithelial layers, promoting microbial translocation. These changes have been implicated in the pathogenesis of both devastating disorders. Thus, we will discuss here these recent findings in the context of novel and alternative therapeutic options.
Collapse
MESH Headings
- Anti-Bacterial Agents/therapeutic use
- Antibodies, Bacterial/biosynthesis
- Bacterial Translocation
- Bile/drug effects
- Bile/microbiology
- Cholangiopancreatography, Endoscopic Retrograde
- Cholangitis, Sclerosing/diagnostic imaging
- Cholangitis, Sclerosing/drug therapy
- Cholangitis, Sclerosing/immunology
- Cholangitis, Sclerosing/microbiology
- Gastrointestinal Microbiome/drug effects
- Host-Pathogen Interactions
- Humans
- Immunoglobulin M/biosynthesis
- Liver Cirrhosis, Biliary/diagnostic imaging
- Liver Cirrhosis, Biliary/drug therapy
- Liver Cirrhosis, Biliary/immunology
- Liver Cirrhosis, Biliary/microbiology
Collapse
Affiliation(s)
- Jochen Mattner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Wasserturmstr. 3/5, D-91054 Erlangen, Germany.
| |
Collapse
|
35
|
Greer R, Dong X, Morgun A, Shulzhenko N. Investigating a holobiont: Microbiota perturbations and transkingdom networks. Gut Microbes 2016; 7:126-35. [PMID: 26979110 PMCID: PMC4856449 DOI: 10.1080/19490976.2015.1128625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The scientific community has recently come to appreciate that, rather than existing as independent organisms, multicellular hosts and their microbiota comprise a complex evolving superorganism or metaorganism, termed a holobiont. This point of view leads to a re-evaluation of our understanding of different physiological processes and diseases. In this paper we focus on experimental and computational approaches which, when combined in one study, allowed us to dissect mechanisms (traditionally named host-microbiota interactions) regulating holobiont physiology. Specifically, we discuss several approaches for microbiota perturbation, such as use of antibiotics and germ-free animals, including advantages and potential caveats of their usage. We briefly review computational approaches to characterize the microbiota and, more importantly, methods to infer specific components of microbiota (such as microbes or their genes) affecting host functions. One such approach called transkingdom network analysis has been recently developed and applied in our study. (1) Finally, we also discuss common methods used to validate the computational predictions of host-microbiota interactions using in vitro and in vivo experimental systems.
Collapse
Affiliation(s)
- Renee Greer
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Xiaoxi Dong
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | | |
Collapse
|
36
|
Vitetta L, Hall S, Coulson S. Metabolic Interactions in the Gastrointestinal Tract (GIT): Host, Commensal, Probiotics, and Bacteriophage Influences. Microorganisms 2015; 3:913-32. [PMID: 27682125 PMCID: PMC5023274 DOI: 10.3390/microorganisms3040913] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 11/25/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022] Open
Abstract
Life on this planet has been intricately associated with bacterial activity at all levels of evolution and bacteria represent the earliest form of autonomous existence. Plants such as those from the Leguminosae family that form root nodules while harboring nitrogen-fixing soil bacteria are a primordial example of symbiotic existence. Similarly, cooperative activities between bacteria and animals can also be observed in multiple domains, including the most inhospitable geographical regions of the planet such as Antarctica and the Lower Geyser Basin of Yellowstone National Park. In humans bacteria are often classified as either beneficial or pathogenic and in this regard we posit that this artificial nomenclature is overly simplistic and as such almost misinterprets the complex activities and inter-relationships that bacteria have with the environment as well as the human host and the plethora of biochemical activities that continue to be identified. We further suggest that in humans there are neither pathogenic nor beneficial bacteria, just bacteria embraced by those that tolerate the host and those that do not. The densest and most complex association exists in the human gastrointestinal tract, followed by the oral cavity, respiratory tract, and skin, where bacteria—pre- and post-birth—instruct the human cell in the fundamental language of molecular biology that normally leads to immunological tolerance over a lifetime. The overall effect of this complex output is the elaboration of a beneficial milieu, an environment that is of equal or greater importance than the bacterium in maintaining homeostasis.
Collapse
Affiliation(s)
- Luis Vitetta
- Medlab Clinical Ltd., Sydney 2015 Australia.
- Sydney Medical School, University of Sydney, Sydney 2006, Australia.
| | - Sean Hall
- Medlab Clinical Ltd., Sydney 2015 Australia.
| | - Samantha Coulson
- Medlab Clinical Ltd., Sydney 2015 Australia.
- Sydney Medical School, University of Sydney, Sydney 2006, Australia.
| |
Collapse
|
37
|
Morgun A, Dzutsev A, Dong X, Greer RL, Sexton DJ, Ravel J, Schuster M, Hsiao W, Matzinger P, Shulzhenko N. Uncovering effects of antibiotics on the host and microbiota using transkingdom gene networks. Gut 2015; 64:1732-43. [PMID: 25614621 PMCID: PMC5166700 DOI: 10.1136/gutjnl-2014-308820] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Despite widespread use of antibiotics for the treatment of life-threatening infections and for research on the role of commensal microbiota, our understanding of their effects on the host is still very limited. DESIGN Using a popular mouse model of microbiota depletion by a cocktail of antibiotics, we analysed the effects of antibiotics by combining intestinal transcriptome together with metagenomic analysis of the gut microbiota. In order to identify specific microbes and microbial genes that influence the host phenotype in antibiotic-treated mice, we developed and applied analysis of the transkingdom network. RESULTS We found that most antibiotic-induced alterations in the gut can be explained by three factors: depletion of the microbiota; direct effects of antibiotics on host tissues and the effects of remaining antibiotic-resistant microbes. Normal microbiota depletion mostly led to downregulation of different aspects of immunity. The two other factors (antibiotic direct effects on host tissues and antibiotic-resistant microbes) primarily inhibited mitochondrial gene expression and amounts of active mitochondria, increasing epithelial cell death. By reconstructing and analysing the transkingdom network, we discovered that these toxic effects were mediated by virulence/quorum sensing in antibiotic-resistant bacteria, a finding further validated using in vitro experiments. CONCLUSIONS In addition to revealing mechanisms of antibiotic-induced alterations, this study also describes a new bioinformatics approach that predicts microbial components that regulate host functions and establishes a comprehensive resource on what, why and how antibiotics affect the gut in a widely used mouse model of microbiota depletion by antibiotics.
Collapse
Affiliation(s)
- Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, Oregon,
USA,Ghost Lab, National Institute of Allergy and Infectious Diseases,
National Institutes of Health, Bethesda, Maryland, USA
| | - Amiran Dzutsev
- Cancer and Inflammation Program, National Cancer Institute/Leidos
Biomedical Research, Inc., Frederick, Maryland, USA
| | - Xiaoxi Dong
- College of Pharmacy, Oregon State University, Corvallis, Oregon,
USA
| | - Renee L Greer
- College of Veterinary Medicine, Oregon State University, Corvallis,
Oregon, USA
| | - D Joseph Sexton
- Department of Microbiology, Oregon State University, Corvallis,
Oregon, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of
Medicine, Baltimore, Maryland, USA
| | - Martin Schuster
- Department of Microbiology, Oregon State University, Corvallis,
Oregon, USA
| | - William Hsiao
- University of British Columbia, Vancouver, British Columbia,
Canada
| | - Polly Matzinger
- Ghost Lab, National Institute of Allergy and Infectious Diseases,
National Institutes of Health, Bethesda, Maryland, USA
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Oregon State University, Corvallis,
Oregon, USA,Ghost Lab, National Institute of Allergy and Infectious Diseases,
National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
38
|
Liu S, Bennett DC, Tun HM, Kim JE, Cheng KM, Zhang H, Leung FC. The effect of diet and host genotype on ceca microbiota of Japanese quail fed a cholesterol enriched diet. Front Microbiol 2015; 6:1092. [PMID: 26500632 PMCID: PMC4595795 DOI: 10.3389/fmicb.2015.01092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/22/2015] [Indexed: 11/23/2022] Open
Abstract
Two Japanese quail strains, respectively atherosclerosis-susceptible (SUS) and –resistant (RES), have been shown to be good models to study cholesterol metabolism and transportation associated with atherosclerosis. Our objective was to examine possible difference in cecal microbiota between these strains when fed a control diet and a cholesterol enriched diet, to determine how host genotype and diet could affect the cecal microbiome that may play a part in cholesterol metabolism. A factorial study with both strains and two diets (control, cholesterol) was carried out. Cecal content was collected from 12 week old quail that have been on their respective diets for 6 weeks. DNA was extracted from the samples and the variable region 3–5 of the bacterial 16S rRNA gene was amplified. The amplicon libraries were subjected to pyrosequencing. Principal Component Analysis (PCA) of β-diversity showed four distinct microbiota communities that can be assigned to the 4 treatment groups (RES/control, RES/cholesterol, SUS/control, SUS/cholesterol). At the Phylum level, the 4 treatment groups has distinct Firmicutes community characteristics but no significant difference in Bacteroidetes. Eubacterium dolichum was rare in RES/control but became overabundant in RES/cholesterol. An unclassified species of Lactobacillaceae was found in abundance in SUS/control but the same species was rare in RES/cholesterol. On the other hand, two Lactobacillus species were only found in RES/control and an unclassified Lachnospiraceae species was abundant in RES/cholesterol but rare in SUS/control. The abundance of four species of Lachnospiraceae, three species of Ruminococcaceae and one species of Coprobacillaceae was positively correlated with plasma Total Cholesterol, plasma LDL, and LDL/HDL ratio. Our study of cecal microbiota in these quail has demonstrated that selection for susceptibility/resistance to diet induced atherosclerosis has also affected the quail's cecal environment to host distinctly different cecal microbiome.
Collapse
Affiliation(s)
- Shasha Liu
- The State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences Beijing, China ; Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Darin C Bennett
- Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Hein M Tun
- School of Biological Sciences, The University of Hong Kong Hong Kong, Hong Kong
| | - Ji-Eun Kim
- Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Kimberly M Cheng
- Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Hongfu Zhang
- The State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences Beijing, China
| | - Frederick C Leung
- School of Biological Sciences, The University of Hong Kong Hong Kong, Hong Kong
| |
Collapse
|
39
|
Giacomin P, Zakrzewski M, Croese J, Su X, Sotillo J, McCann L, Navarro S, Mitreva M, Krause L, Loukas A, Cantacessi C. Experimental hookworm infection and escalating gluten challenges are associated with increased microbial richness in celiac subjects. Sci Rep 2015; 5:13797. [PMID: 26381211 PMCID: PMC4585380 DOI: 10.1038/srep13797] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/04/2015] [Indexed: 02/08/2023] Open
Abstract
The intestinal microbiota plays a critical role in the development of the immune system. Recent investigations have highlighted the potential of helminth therapy for treating a range of inflammatory disorders, including celiac disease (CeD); however, the mechanisms by which helminths modulate the immune response of the human host and ameliorate CeD pathology are unknown. In this study, we investigated the potential role of alterations in the human gut microbiota in helminth-mediated suppression of an inflammatory disease. We assessed the qualitative and quantitative changes in the microbiota of human volunteers with CeD prior to and following infection with human hookworms, and following challenge with escalating doses of dietary gluten. Experimental hookworm infection of the trial subjects resulted in maintenance of the composition of the intestinal flora, even after a moderate gluten challenge. Notably, we observed a significant increase in microbial species richness over the course of the trial, which could represent a potential mechanism by which hookworms can regulate gluten-induced inflammation and maintain intestinal immune homeostasis.
Collapse
Affiliation(s)
- Paul Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Martha Zakrzewski
- Bioinformatics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - John Croese
- Prince Charles Hospital, Brisbane, QLD, Australia
| | - Xiaopei Su
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Leisa McCann
- Prince Charles Hospital, Brisbane, QLD, Australia
| | - Severine Navarro
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Makedonka Mitreva
- The Genome Institute, and.,Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lutz Krause
- Bioinformatics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Cinzia Cantacessi
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
West CE, Jenmalm MC, Prescott SL. The gut microbiota and its role in the development of allergic disease: a wider perspective. Clin Exp Allergy 2015; 45:43-53. [PMID: 24773202 DOI: 10.1111/cea.12332] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The gut microbiota are critical in the homoeostasis of multiple interconnected host metabolic and immune networks. If early microbial colonization is delayed, the gut-associated lymphoid tissues (GALT) fail to develop, leading to persistent immune dysregulation in mice. Microbial colonization has also been proposed as a major driver for the normal age-related maturation of both Th1 and T regulatory (Treg) pathways that appear important in suppressing early propensity for Th2 allergic responses. There is emerging evidence that resident symbionts induce tolerogenic gut-associated Treg cells and dendritic cells that ensure the preferential growth of symbionts; keeping pathogenic strains in check and constraining proinflammatory Th1, Th2, and Th17 clones. Some effects of symbionts are mediated by short-chain fatty acids, which play a critical role in mucosal integrity and local and systemic metabolic function and stimulate the regulatory immune responses. The homoeostatic IL-10/TGF-β dominated tolerogenic response within the GALT also signals the production of secretory IgA, which have a regulating role in mucosal integrity. Contrary to the 'sterile womb' paradigm, recent studies suggest that maternal microbial transfer to the offspring begins during pregnancy, providing a pioneer microbiome. It is likely that appropriate microbial stimulation both pre- and postnatally is required for optimal Th1 and Treg development to avoid the pathophysiological processes leading to allergy. Disturbed gut colonization patterns have been associated with allergic disease, but whether microbial variation is the cause or effect of these diseases is still under investigation. We are far from understanding what constitutes a 'healthy gut microbiome' that promotes tolerance. This remains a major limitation and might explain some of the inconsistency in human intervention studies with prebiotics and probiotics. Multidisciplinary integrative approaches with researchers working in networks, using harmonized outcomes and methodologies, are needed to advance our understanding in this field.
Collapse
Affiliation(s)
- C E West
- International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden; Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
41
|
Larsen PE, Dai Y. Metabolome of human gut microbiome is predictive of host dysbiosis. Gigascience 2015; 4:42. [PMID: 26380076 PMCID: PMC4570295 DOI: 10.1186/s13742-015-0084-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/28/2015] [Indexed: 01/01/2023] Open
Abstract
Background Humans live in constant and vital symbiosis with a closely linked bacterial ecosystem called the microbiome, which influences many aspects of human health. When this microbial ecosystem becomes disrupted, the health of the human host can suffer; a condition called dysbiosis. However, the community compositions of human microbiomes also vary dramatically from individual to individual, and over time, making it difficult to uncover the underlying mechanisms linking the microbiome to human health. We propose that a microbiome’s interaction with its human host is not necessarily dependent upon the presence or absence of particular bacterial species, but instead is dependent on its community metabolome; an emergent property of the microbiome. Results Using data from a previously published, longitudinal study of microbiome populations of the human gut, we extrapolated information about microbiome community enzyme profiles and metabolome models. Using machine learning techniques, we demonstrated that the aggregate predicted community enzyme function profiles and modeled metabolomes of a microbiome are more predictive of dysbiosis than either observed microbiome community composition or predicted enzyme function profiles. Conclusions Specific enzyme functions and metabolites predictive of dysbiosis provide insights into the molecular mechanisms of microbiome–host interactions. The ability to use machine learning to predict dysbiosis from microbiome community interaction data provides a potentially powerful tool for understanding the links between the human microbiome and human health, pointing to potential microbiome-based diagnostics and therapeutic interventions. Electronic supplementary material The online version of this article (doi:10.1186/s13742-015-0084-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter E Larsen
- Bioengineering Department, University of Illinois at Chicago, 851 South Morgan, SEO218, Chicago, IL 60607 USA ; Argonne National Laboratory, Biosciences Division, 9700 South Cass Ave, Argonne, IL 60439 USA
| | - Yang Dai
- Bioengineering Department, University of Illinois at Chicago, 851 South Morgan, SEO218, Chicago, IL 60607 USA
| |
Collapse
|
42
|
Rogers GB. The human microbiome: opportunities and challenges for clinical care. Intern Med J 2015; 45:889-98. [DOI: 10.1111/imj.12650] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/19/2014] [Indexed: 12/11/2022]
Affiliation(s)
- G. B. Rogers
- Microbiome Research; South Australian Health and Medical Research Institute Infection and Immunity Theme; School of Medicine; Flinders University; Adelaide South Australia Australia
| |
Collapse
|
43
|
Hu C, Wong FS, Wen L. Type 1 diabetes and gut microbiota: Friend or foe? Pharmacol Res 2015; 98:9-15. [PMID: 25747961 PMCID: PMC4469505 DOI: 10.1016/j.phrs.2015.02.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/25/2015] [Accepted: 02/25/2015] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes is a T cell-mediated autoimmune disease. Environmental factors play an important role in the initiation of the disease in genetically predisposed individuals. With the improved control of infectious disease, the incidence of autoimmune diseases, particularly type 1 diabetes, has dramatically increased in developed countries. Increasing evidence suggests that gut microbiota are involved in the pathogenesis of type 1 diabetes. Here we focus on recent advances in this field and provide a rationale for novel therapeutic strategies targeting gut microbiota for the prevention of type 1 diabetes.
Collapse
Affiliation(s)
- Changyun Hu
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - F Susan Wong
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
44
|
Abstract
The incidence of allergic diseases is increasing, both in developed and developing countries, concomitantly with the rise in living standards and the adoption of a 'western lifestyle'. For two decades, the hygiene hypothesis - which proposes that the lack of early childhood exposure to infectious agents increases susceptibility to allergic diseases in later life - provided the conceptual framework for unravelling the mechanisms that could account for the increased incidence of allergic diseases. In this Review, we discuss recent evidence that highlights the role of diet as a key factor influencing immune homeostasis and the development of allergic diseases through a complex interplay between nutrients, their metabolites and immune cell populations. Although further investigations are still required to understand these complex relationships, recent data have established a possible connection between metabolic homeostasis and allergic diseases.
Collapse
|
45
|
|
46
|
Abstract
OBJECTIVE Despite the use of HAART to control HIV, systemic immune activation and inflammation persists with the consequence of developing serious non-AIDS events. The mechanisms that contribute to persistent systemic immune activation have not been well defined. The intestine is the major source of "sterile" inflammation and plays a critical role in immune function; thus, we sought to determine whether intestinal gene expression was altered in virally controlled HIV-infected individuals. DESIGN AND METHODS Gene expression was compared in biopsy samples collected from HIV-uninfected and HIV-infected individuals from the ileum, right colon (ascending colon), and left colon (sigmoid). Affymetrix gene arrays were performed on tissues and pathway analyses were conducted. Gene expression was correlated with systemic markers of intestinal barrier dysfunction and inflammation and intestinal microbiota composition. RESULTS Genes involved in cellular immune response, cytokine signaling, pathogen-influenced signaling, humoral immune response, apoptosis, intracellular and second messenger signaling, cancer, organismal growth and development, and proliferation and development were upregulated in the intestine of HIV-infected individuals with differences observed in the ileum, right, and left colon. Gene expression in the ileum primarily correlated with systemic markers of inflammation (e.g., IL7R, IL2, and TLR2 with serum TNF) whereas expression in the colon correlated with the microbiota community (e.g., IFNG, IL1B, and CD3G with Bacteroides). CONCLUSION These data demonstrate persistent, proinflammatory changes in the intestinal mucosa of virally suppressed HIV-infected individuals. These changes in intestinal gene expression may be the consequence of or contribute to barrier dysfunction and intestinal dysbiosis observed in HIV.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The complex biochemical composition and physical structure of the milk fat globule (MFG) are presented as a basis for its paradoxical metabolic fate: MFG is a rapid conveyor of energy through its triacylglycerol (TAG) core but contains some low-digestible bioactive complex lipids and proteins, which influence lipid metabolism and contribute to intestinal and systemic health. RECENT FINDINGS MFG structure modulates gastrointestinal lipolysis, postprandial lipemia and even the postprandial fate of ingested fatty acids. Proof-of-concept of the nutritional programming induced by early consumption of an emulsion biomimetic of MFG compared with a typical infant formula was published in an animal model (mice). The metabolic response to a high-fat diet during adulthood was improved following neonatal exposure to the biomimetic emulsion. SUMMARY MFG TAG are tailored with a unique regiodistribution delivering in priority short to medium-chain fatty acids in gastric phase, an important amount of quickly metabolizable oleic acid and protecting palmitic acid in sn-2 position. MFG digestion may not only trigger rapid TAG and chylomicron plasma peaks with fast clearance but also the luminal release of nonhydrolysable bioactive compounds (glycosylated compounds and sphingomyelin), which contribute to intestinal and systemic health by shaping the microbiota and modulating the immune system. These bioactive compounds form self-assembled structures, protect specific micronutrients and lower cholesterol absorption. The health benefits of MFG consumption or of some of its fractions (MFGM) under specific structures are steadily being demonstrated with still much unsolved questions especially for populations with high nutritional needs (e.g. elderly, infants).
Collapse
Affiliation(s)
- Claire Bourlieu
- aINRA, UMR1253, STLO, Rennes bINRA, UMR1397, INSERM U1060, CarMeN laboratory, Villeurbanne, France
| | | |
Collapse
|
48
|
|
49
|
Gut microbiota-generated metabolites in animal health and disease. Nat Chem Biol 2014; 10:416-24. [PMID: 24838170 DOI: 10.1038/nchembio.1535] [Citation(s) in RCA: 501] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 04/22/2014] [Indexed: 12/27/2022]
Abstract
Gut microbiota is found in virtually any metazoan, from invertebrates to vertebrates. It has long been believed that gut microbiota, more specifically, the activity of the microbiome and its metabolic products, directly influence a variety of aspects in metazoan physiology. However, the exact molecular relationship among microbe-derived gut metabolites, host signaling pathways, and host physiology remains to be elucidated. Here we review recent discoveries regarding the molecular links between gut metabolites and host physiology in different invertebrate and vertebrate animal models. We describe the different roles of gut microbiome activity and their metabolites in regulating distinct host physiology and the molecular mechanisms by which gut metabolites cause physiological homeostasis via regulating specific host signaling pathways. Future studies in this direction using different animal models will provide the key concepts to understanding the evolutionarily conserved chemical dialogues between gut microbiota and metazoan cells and also human diseases associated with gut microbiota and metabolites.
Collapse
|
50
|
Kim BJ, Lee SY, Kim HB, Lee E, Hong SJ. Environmental changes, microbiota, and allergic diseases. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2014; 6:389-400. [PMID: 25228995 PMCID: PMC4161679 DOI: 10.4168/aair.2014.6.5.389] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/12/2013] [Indexed: 12/26/2022]
Abstract
During the last few decades, the prevalence of allergic disease has increased dramatically. The development of allergic diseases has been attributed to complex interactions between environmental factors and genetic factors. Of the many possible environmental factors, most research has focused on the most commonly encountered environmental factors, such as air pollution and environmental microbiota in combination with climate change. There is increasing evidence that such environmental factors play a critical role in the regulation of the immune response that is associated with allergic diseases, especially in genetically susceptible individuals. This review deals with not only these environmental factors and genetic factors but also their interactions in the development of allergic diseases. It will also emphasize the need for early interventions that can prevent the development of allergic diseases in susceptible populations and how these interventions can be identified.
Collapse
Affiliation(s)
- Byoung-Ju Kim
- Department of Pediatrics, Inje University Haeundae Paik Hospital, Busan, Korea
| | - So-Yeon Lee
- Department of Pediatrics, Hallym University Sacred Heart Hospital, University of Hallym College of Medicine, Anyang, Korea
| | - Hyo-Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Eun Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, University of Ulsan College of Medicine, Seoul, Korea. ; Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, University of Ulsan College of Medicine, Seoul, Korea. ; Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|