51
|
Van Hul M, Cani PD. The gut microbiota in obesity and weight management: microbes as friends or foe? Nat Rev Endocrinol 2023; 19:258-271. [PMID: 36650295 DOI: 10.1038/s41574-022-00794-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/18/2023]
Abstract
Obesity is caused by a long-term difference between energy intake and expenditure - an imbalance that is seemingly easily restored by increasing exercise and reducing caloric consumption. However, as simple as this solution appears, for many people, losing excess weight is difficult to achieve and even more difficult to maintain. The reason for this difficulty is that energy intake and expenditure, and by extension body weight, are regulated through complex hormonal, neural and metabolic mechanisms that are under the influence of many environmental factors and internal responses. Adding to this complexity, the microorganisms (microbes) that comprise the gut microbiota exert direct effects on the digestion, absorption and metabolism of food. Furthermore, the gut microbiota exerts a miscellany of protective, structural and metabolic effects both on the intestinal milieu and peripheral tissues, thus affecting body weight by modulating metabolism, appetite, bile acid metabolism, and the hormonal and immune systems. In this Review, we outline historical and recent advances in understanding how the gut microbiota is involved in regulating body weight homeostasis. We also discuss the opportunities, limitations and challenges of using gut microbiota-related approaches as a means to achieve and maintain a healthy body weight.
Collapse
Affiliation(s)
- Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
52
|
Nishimoto Y, Fujisawa K, Ukawa Y, Kudoh M, Funahashi K, Kishimoto Y, Fukuda S. Effect of urolithin A on the improvement of vascular endothelial function depends on the gut microbiota. Front Nutr 2023; 9:1077534. [PMID: 36687672 PMCID: PMC9854132 DOI: 10.3389/fnut.2022.1077534] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Background Urolithin A (UA) is a metabolite produced by gut microbiota from ingested ellagic acid. Although the effect of ellagic acid intake on vascular endothelial function (VEF) improvement has been reported, the effect of UA intake on VEF improvement remains obscure. In addition, UA has been reported to improve the intestinal barrier function, and UA may have improved VEF by gut microbiome alteration. Objective In this study, we conducted a clinical trial to explore and analyze the effects of UA intake on vascular endothelial function (VEF) and characteristics of the intestinal environment, such as gut microbiome profiling and organic acid composition. Methods A placebo-controlled, randomized, double-blinded, parallel group trial was conducted on participants who could metabolize small amounts of UA from ellagic acid (non-UA producers) and had relatively poor VEF. VEF was assessed using the flow-mediated vasodilatation (FMD) score. Participants were administered placebo, UA 10 mg/day, or UA 50 mg/day for 12 weeks. FMD was measured and fecal samples were collected at 0, 4, 8, and 12 weeks of treatment. Gut microbiome analysis and organic acid level measurements were performed to evaluate the effects of UA intake on the intestinal environment. This clinical trial is publicly registered at the UMIN-CTR, trial number: UMIN000042014. Results The gut microbiota of the UA 50 mg/day group showed a significant increase in alpha diversity (Faith's phylogenetic diversity). Four and nine microbial genera were significantly altered in the UA 10 mg/day and UA 50 mg/day groups, respectively (p < 0.05, not corrected). Participants whose FMD scores improved with UA intake had poor baseline FMD values as well as a low Bacillota/Bacteroidota ratio. Conclusion Urolithin A intake alters the gut microbiota and improves their alpha diversity. In addition, the effect of UA on VEF correlated with the individual gut microbiota. Our results have practical implications for a new approach to providing healthcare that focuses on intestinal environment-based diet therapy.
Collapse
Affiliation(s)
| | - Kota Fujisawa
- Metagen Inc., Tsuruoka, Japan,Department of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Yuichi Ukawa
- Healthcare SBU, DAICEL Corporation, Tokyo, Japan
| | | | | | - Yoshimi Kishimoto
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Setsunan University, Hirakata, Japan
| | - Shinji Fukuda
- Metagen Inc., Tsuruoka, Japan,Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan,Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan,Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan,Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan,*Correspondence: Shinji Fukuda,
| |
Collapse
|
53
|
Wang W, Dang G, Khan I, Ye X, Liu L, Zhong R, Chen L, Ma T, Zhang H. Bacterial Community Characteristics Shaped by Artificial Environmental PM2.5 Control in Intensive Broiler Houses. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:723. [PMID: 36613044 PMCID: PMC9819255 DOI: 10.3390/ijerph20010723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Multilayer cage-houses for broiler rearing have been widely used in intensive Chinese farming in the last decade. This study investigated the characteristics and influencing factors of bacterial communities in the PM2.5 of broiler cage-houses. The PM2.5 samples and environmental variables were collected inside and outside of three parallel broiler houses at the early, middle, and late rearing stages; broiler manure was also gathered simultaneously. The bacterial 16S rRNA sequencing results indicated that indoor bacterial communities were different from the outdoor atmosphere and manure. Furthermore, the variations in airborne bacterial composition and structure were highly influenced by the environmental control variables at different growth stages. The db-RDA results showed that temperature and wind speed, which were artificially modified according to managing the needs for broiler growth, were the main factors affecting the diversity of dominant taxa. Indoor airborne and manurial samples shared numerous common genera, which contained high abundances of manure-origin bacteria. Additionally, the airborne bacterial community tended to stabilize in the middle and late stages, but the population of potentially pathogenic bacteria grew gradually. Overall, this study enhances the understanding of airborne bacteria variations and highlighted the potential role of environmental control measures in intensive farming.
Collapse
Affiliation(s)
- Wenxing Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guoqi Dang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Imran Khan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaobin Ye
- Bureau of Agriculture and Rural Affairs of Luanping County, Chengde 068250, China
| | - Lei Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Teng Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
54
|
Zhao H, Zong Y, Li W, Wang Y, Zhao W, Meng X, Yang F, Kong J, Zhao X, Wang J. Damp-heat constitution influences gut microbiota and urine metabolism of Chinese infants. Heliyon 2022; 9:e12424. [PMID: 36755610 PMCID: PMC9900481 DOI: 10.1016/j.heliyon.2022.e12424] [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: 07/28/2022] [Revised: 11/15/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Background As an increasingly popular complementary and alternative approach for early detection and treatment of disease, traditional Chinese medicine constitution (TCMC) divides human beings into those with balanced constitution (BC) and unbalanced constitution, where damp-heat constitution (DHC) is one of the most unbalanced constitutions. Many studies have been carried out on the microscopic mechanism of constitution classification; however, most of these studies were conducted in adults and rarely in infants. Many diseases are closely related to intestinal microbiota, and metabolites produced by the interaction between microbiota and the body may impact constitution classification. Herein, we investigated the overall constitution distribution in Chinese infants, and analyzed the profiles of gut microbiota and urine metabolites of DHC to further promote the understanding of infants constitution classification. Methods General information was collected and TCMC was evaluated by Constitutional Medicine Questionnaires. 1315 questionnaires were received in a cross-sectional study to investigate the constitution composition in Chinese infants. A total of 56 infants, including 30 DHC and 26 BC, were randomly selected to analyze gut microbiota by 16S rRNA sequencing and urine metabolites by UPLC-Q-TOF/MS method. Results BC was the most common constitution in Chinese infants, DHC was the second common constitution. The gut microbiota and urine metabolites in the DHC group showed different composition compared to the BC group. Four differential genera and twenty differential metabolites were identified. In addition, the combined marker composed of four metabolites may have the high potential to discriminate DHC from BC with an AUC of 0.765. Conclusions The study revealed the systematic differences in the gut microbiota and urine metabolites between infants with DHC and BC. Moreover, the differential microbiota and metabolites may offer objective evidences for constitution classification.
Collapse
Affiliation(s)
- Haihong Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yuhan Zong
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenle Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yaqi Wang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Weibo Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xianghe Meng
- Neurology Department, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Fan Yang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingwei Kong
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd., Beijing, 100015, China
| | - Xiaoshan Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China,School of Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ji Wang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Disease, Beijing University of Chinese Medicine, Beijing, 100029, China,Corresponding author.
| |
Collapse
|
55
|
Singh V, Lee G, Son H, Amani S, Baunthiyal M, Shin JH. Anti-diabetic prospects of dietary bio-actives of millets and the significance of the gut microbiota: A case of finger millet. Front Nutr 2022; 9:1056445. [PMID: 36618686 PMCID: PMC9815516 DOI: 10.3389/fnut.2022.1056445] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Finger millet (Eleusine coracana) is a staple food in several parts of the world because of its high nutritional value. In addition to its high nutrient content, finger millet contains numerous bioactive compounds, including polyphenol (10.2 mg/g TAE), flavonoid (5.54 mg/g CE), phytic acid (0.48%), and dietary fiber (15-20%). Polyphenols are known for their anti-oxidant and anti-diabetic role. Phytic acid, previously considered an anti-nutritive substance, is now regarded as a nutraceutical as it reduces carbohydrate digestibility and thus controls post-prandial glucose levels and obesity. Thus, finger millet is an attractive diet for patients with diabetes. Recent findings have revealed that the anti-oxidant activity and bio-accessibility of finger millet polyphenols increased significantly (P < 0.05) in the colon, confirming the role of the gut microbiota. The prebiotic content of finger millet was also utilized by the gut microbiota, such as Faecalibacterium, Eubacterium, and Roseburia, to generate colonic short-chain fatty acids (SCFAs), and probiotic Bifidobacterium and Lactobacillus, which are known to be anti-diabetic in nature. Notably, finger millet-induced mucus-degrading Akkermansia muciniphila can also help in alleviate diabetes by releasing propionate and Amuc_1100 protein. Various millet bio-actives effectively controlled pathogenic gut microbiota, such as Shigella and Clostridium histolyticum, to lower gut inflammation and, thus, the risk of diabetes in the host. In the current review, we have meticulously examined the role of gut microbiota in the bio-accessibility of millet compounds and their impact on diabetes.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - HyunWoo Son
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sliti Amani
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Mamta Baunthiyal
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering and Technology, Ghurdauri, India,*Correspondence: Mamta Baunthiyal,
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea,Jae-Ho Shin,
| |
Collapse
|
56
|
Wortelboer K, Koopen AM, Herrema H, de Vos WM, Nieuwdorp M, Kemper EM. From fecal microbiota transplantation toward next-generation beneficial microbes: The case of Anaerobutyricum soehngenii. Front Med (Lausanne) 2022; 9:1077275. [PMID: 36544495 PMCID: PMC9760881 DOI: 10.3389/fmed.2022.1077275] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The commensal gut microbiota is important for human health and well-being whereas deviations of the gut microbiota have been associated with a multitude of diseases. Restoration of a balanced and diverse microbiota by fecal microbiota transplantation (FMT) has emerged as a potential treatment strategy and promising tool to study causality of the microbiota in disease pathogenesis. However, FMT comes with logistical challenges and potential safety risks, such as the transfer of pathogenic microorganisms, undesired phenotypes or an increased risk of developing disease later in life. Therefore, a more controlled, personalized mixture of cultured beneficial microbes might prove a better alternative. Most of these beneficial microbes will be endogenous commensals to the host without a long history of safe and beneficial use and are therefore commonly referred to as next-generation probiotics (NGP) or live biotherapeutic products (LBP). Following a previous FMT study within our group, the commensal butyrate producer Anaerobutyricum spp. (previously named Eubacterium hallii) was found to be associated with improved insulin-sensitivity in subjects with the metabolic syndrome. After the preclinical testing with Anaerobutyricum soehngenii in mice models was completed, the strain was produced under controlled conditions and several clinical studies evaluating its safety and efficacy in humans were performed. Here, we describe and reflect on the development of A. soehngenii for clinical use, providing practical guidance for the development and testing of NGPs and reflecting on the current regulatory framework.
Collapse
Affiliation(s)
- Koen Wortelboer
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
- Department of Pharmacy, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Annefleur M. Koopen
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
| | - Willem M. de Vos
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Diabetes Center, Department of Internal Medicine, Amsterdam UMC, Location VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - E. Marleen Kemper
- Department of Pharmacy, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
57
|
Freitas RGBON, Vasques ACJ, Fernandes GR, Ribeiro FB, Solar I, Barbosa MG, Almeida-Pititto B, Geloneze B, Ferreira SRG. Gestational weight gain and visceral adiposity in adult offspring: Is there a link with the fecal abundance of Acidaminococcus genus? Eur J Clin Nutr 2022; 76:1705-1712. [PMID: 35906333 DOI: 10.1038/s41430-022-01182-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 11/08/2022]
Abstract
Intrauterine environment can influence the offspring's body adiposity whose distribution affect the cardiometabolic risk. Underlying mechanisms may involve the gut microbiome. We investigated associations of gestational weight gain with the adult offspring's gut microbiota, body adiposity and related parameters in participants of the Nutritionists' Health Study. METHODS This cross-sectional analysis included 114 women who had early life and clinical data, body composition, and biological samples collected. The structure of fecal microbiota was analyzed targeting the V4 region of the 16 S rRNA gene. Beta diversity was calculated by PCoA and PERMANOVA used to test the impact of categorical variables into the diversity. Bacterial clusters were identified based on the Jensen-Shannon divergence matrix and Calinski-Harabasz index. Correlations were tested by Spearman coefficient. RESULTS Median age was 28 (IQR 24-31) years and BMI 24.5 (IQR 21.4-28.0) kg/m2. Fifty-eight participants were assigned to a profile driven by Prevotella and 56 to another driven by Blautia. Visceral adipose tissue was correlated to abundance of Acidaminococcus genus considering the entire sample (r = 0.37; p < 0.001) and the profiles (Blautia: r = 0.35, p = 0.009, and Prevotella: r = 0.38, p = 0.006). In Blautia-driven profile, the same genus was also correlated to maternal gestational weight gain (r = 0.38, p = 0.006). CONCLUSIONS Association of Acidaminococcus with gestational weight gain could reinforce the relevance with mothers' nutritional status for gut colonization at the beginning of life. Whether Acidaminococcus abundance could be a marker for central distribution of adiposity in young women requires further investigation.
Collapse
Affiliation(s)
- R G B O N Freitas
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences - University of Campinas, São Paulo, Brazil
| | - A C J Vasques
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences - University of Campinas, São Paulo, Brazil
- School of Applied Sciences - University of Campinas, São Paulo, Brazil
| | - G R Fernandes
- Oswaldo Cruz Foundation, Belo Horizonte, São Paulo, Brazil
| | - F B Ribeiro
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences - University of Campinas, São Paulo, Brazil
| | - I Solar
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences - University of Campinas, São Paulo, Brazil
- School of Applied Sciences - University of Campinas, São Paulo, Brazil
| | - M G Barbosa
- School of Applied Sciences - University of Campinas, São Paulo, Brazil
| | - B Almeida-Pititto
- Department of Preventive Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - B Geloneze
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences - University of Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, São Paulo, Brazil
| | - S R G Ferreira
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
58
|
Zhang H, Duan Y, Cai F, Cao D, Wang L, Qiao Z, Hong Q, Li N, Zheng Y, Su M, Liu Z, Zhu B. Next-Generation Probiotics: Microflora Intervention to Human Diseases. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5633403. [PMID: 36440358 PMCID: PMC9683952 DOI: 10.1155/2022/5633403] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 06/06/2022] [Indexed: 11/02/2023]
Abstract
With the development of human genome sequencing and techniques such as intestinal microbial culture and fecal microbial transplantation, newly discovered microorganisms have been isolated, cultured, and researched. Consequently, many beneficial probiotics have emerged as next-generation probiotics (NGPs). Currently, "safety," "individualized treatment," and "internal interaction within the flora" are requirements of a potential NGPs. Furthermore, in the complex ecosystem of humans and microbes, it is challenging to identify the relationship between specific strains, specific flora, and hosts to warrant a therapeutic intervention in case of a disease. Thus, this review focuses on the progress made in NGPs and human health research by elucidating the limitations of traditional probiotics; summarizing the functions and strengths of Akkermansia muciniphila, Faecalibacterium prausnitzii, Bacteroides fragilis, Eubacterium hallii, and Roseburia spp. as NGPs; and determining the role of their intervention in treatment of certain diseases. Finally, we aim to provide a reference for developing new probiotics in the future.
Collapse
Affiliation(s)
- Huanchang Zhang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Yunfeng Duan
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Feng Cai
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Demin Cao
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lei Wang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Zhenyi Qiao
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Qing Hong
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Nan Li
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Yuanrong Zheng
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Miya Su
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Baoli Zhu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
59
|
Akkermansia muciniphila: paradigm for next-generation beneficial microorganisms. Nat Rev Gastroenterol Hepatol 2022; 19:625-637. [PMID: 35641786 DOI: 10.1038/s41575-022-00631-9] [Citation(s) in RCA: 504] [Impact Index Per Article: 168.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2022] [Indexed: 12/15/2022]
Abstract
Ever since Akkermansia muciniphila was discovered and characterized two decades ago, numerous studies have shown that the lack or decreased abundance of this commensal bacterium was linked with multiple diseases (such as obesity, diabetes, liver steatosis, inflammation and response to cancer immunotherapies). Although primarily based on simple associations, there are nowadays an increasing number of studies moving from correlations to causality. The causal evidence derived from a variety of animal models performed in different laboratories and recently was also recapitulated in a human proof-of-concept trial. In this Review, we cover the history of the discovery of A. muciniphila and summarize the numerous findings and main mechanisms of action by which this intestinal symbiont improves health. A comparison of this microorganism with other next-generation beneficial microorganisms that are being developed is also made.
Collapse
|
60
|
Dietary fat quality impacts metabolic impairments of type 2 diabetes risk differently in male and female CD-1 ® mice. Br J Nutr 2022; 128:1013-1028. [PMID: 34605388 DOI: 10.1017/s0007114521004001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Metabolic impairments associated with type 2 diabetes, including insulin resistance and loss of glycaemic control, disproportionately impact the elderly. Lifestyle interventions, such as manipulation of dietary fat quality (i.e. fatty acid (FA) composition), have been shown to favourably modulate metabolic health. Yet, whether or not chronic consumption of beneficial FAs can protect against metabolic derangements and disease risk during ageing is not well defined. We sought to evaluate whether long-term dietary supplementation of fish-, dairy- or echium-derived FAs to the average FA profile in a U.S. American diet may offset metabolic impairments in males and females during ageing. One-month-old CD-1® mice were fed isoenergetic, high-fat (40 %) diets with the fat content composed of either 100 % control fat blend (CO) or 70 % CO with 30 % fish oil, dairy fat or echium oil for 13 months. Every 3 months, parameters of glucose homoeostasis were evaluated via glucose and insulin tolerance tests. Glucose tolerance improved in males consuming a diet supplemented with fish oil or echium oil as ageing progressed, but not in females. Yet, females were more metabolically protected than males regardless of age. Additionally, Spearman correlations were performed between indices of glucose homoeostasis and previously reported measurements of diet-derived FA content in tissues and colonic bacterial composition, which also revealed sex-specific associations. This study provides evidence that long-term dietary fat quality influences risk factors of metabolic diseases during ageing in a sex-dependent manner; thus, sex is a critical factor to be considered in future dietary strategies to mitigate type 2 diabetes risk.
Collapse
|
61
|
Wu L, Gao Y, Su Y, Li J, Ren WC, Wang QH, Kuang HX. Probiotics with anti-type 2 diabetes mellitus properties: targets of polysaccharides from traditional Chinese medicine. Chin J Nat Med 2022; 20:641-655. [PMID: 36162950 DOI: 10.1016/s1875-5364(22)60210-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Indexed: 12/12/2022]
Abstract
Traditional Chinese medicine polysaccharides is a biologically active ingredient that is not easy to be digested. It is fermented by intestinal microflora to promote qualitative and selective changes in the composition of the intestinal microbiome, which often result in beneficial effects on the health of the host. People call it "prebiotics". In this review, we systematically summarized the anti-diabetic effect of traditional Chinese medicine polysaccharides. These polysaccharides regulate the metabolism of sugar and lipids by inter-influence with the intestinal microflora, and maintain human health, while improving type 2 diabetes-like symptoms such as high blood glucose, and abnormal glucose and lipid metabolism.
Collapse
Affiliation(s)
- Lun Wu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yue Gao
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Key Laboratory of Medicinal Materials, Chinese Academy of Sciences, Harbin 150040, China
| | - Yang Su
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Key Laboratory of Medicinal Materials, Chinese Academy of Sciences, Harbin 150040, China; Faculty of Microbiology and Immunogenetics, University of California, Los Angeles, CA 90095, USA.
| | - Jing Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Key Laboratory of Medicinal Materials, Chinese Academy of Sciences, Harbin 150040, China
| | - Wen-Chen Ren
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Key Laboratory of Medicinal Materials, Chinese Academy of Sciences, Harbin 150040, China
| | - Qiu-Hong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Hai-Xue Kuang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Key Laboratory of Medicinal Materials, Chinese Academy of Sciences, Harbin 150040, China.
| |
Collapse
|
62
|
Ecker M, Haller D. Bedeutung des Mikrobioms für Adipositas und Glukosestoffwechsel. GYNAKOLOGISCHE ENDOKRINOLOGIE 2022. [DOI: 10.1007/s10304-022-00467-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
63
|
Bai J, Wan Z, Zhang Y, Wang T, Xue Y, Peng Q. Composition and diversity of gut microbiota in diabetic retinopathy. Front Microbiol 2022; 13:926926. [PMID: 36081798 PMCID: PMC9445585 DOI: 10.3389/fmicb.2022.926926] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveDiabetic retinopathy (DR) is one of the most common complications of type 2 diabetes mellitus. The current study investigates the composition, structure, and function of gut microbiota in DR patients and explores the correlation between gut microbiota and clinical characteristics of DR.MethodsA total of 50 stool samples were collected from 50 participants, including 25 DR patients and 25 healthy controls (HCs). 16S ribosomal RNA gene sequencing was used to analyze the gut microbial composition in these two groups. DNA was extracted from the fecal samples using the MiSeq platform.ResultsThe microbial structure and composition of DR patients were different from that of HCs. The microbial richness of gut microbiota in DR was higher than that of normal individuals. The alterations of microbiome of DR patients were associated with disrupted Firmicutes, Bacteroidetes, Synergistota, and Desulfobacterota phyla. In addition, increased levels of Bacteroides, Megamonas, Ruminococcus_torques_group, Lachnoclostridium, and Alistipes, and decreased levels of Blautia, Eubacterium_ hallii_group, Collinsella, Dorea, Romboutsia, Anaerostipes, and Fusicatenibacter genera were observed in the DR groups. Additionally, a stochastic forest model was developed to identify a set of biomarkers with seven bacterial genera that can differentiate patients with DR from those HC. The microbial communities exhibited varied functions in these two groups because of the alterations of the above-mentioned bacterial genera.ConclusionThe altered composition and function of gut microbiota in DR patients indicated that gut microbiome could be used as non-invasive biomarkers, improve clinical diagnostic methods, and identify putative therapeutic targets for DR.
Collapse
|
64
|
Burakova I, Smirnova Y, Gryaznova M, Syromyatnikov M, Chizhkov P, Popov E, Popov V. The Effect of Short-Term Consumption of Lactic Acid Bacteria on the Gut Microbiota in Obese People. Nutrients 2022; 14:3384. [PMID: 36014890 PMCID: PMC9415828 DOI: 10.3390/nu14163384] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity is a problem of modern health care that causes the occurrence of many concomitant diseases: arterial hypertension, diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular diseases. New strategies for the treatment and prevention of obesity are being developed that are based on using probiotics for modulation of the gut microbiota. Our study aimed to evaluate the bacterial composition of the gut of obese patients before and after two weeks of lactic acid bacteria (Lactobacillus acidophilus, Lactiplantibacillus plantarum, Limosilactobacillus fermentum, and Lactobacillus delbrueckii) intake. The results obtained showed an increase in the number of members of the phylum Actinobacteriota in the group taking nutritional supplements, while the number of phylum Bacteroidota decreased in comparison with the control group. There has also been an increase in potentially beneficial groups: Bifidobacterium, Blautia, Eubacterium, Anaerostipes, Lactococcus, Lachnospiraceae ND3007, Streptococcus, Escherichia-Shigella, and Lachnoclostridium. Along with this, a decrease in the genera was demonstrated: Faecalibacterium, Pseudobutyrivibrio, Subdoligranulum, Faecalibacterium, Clostridium sensu stricto 1 and 2, Catenibacterium, Megasphaera, Phascolarctobacterium, and the Oscillospiraceae NK4A214 group, which contribute to the development of various metabolic disorders. Modulation of the gut microbiota by lactic acid bacteria may be one of the ways to treat obesity.
Collapse
Affiliation(s)
- Inna Burakova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
| | - Yuliya Smirnova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Mariya Gryaznova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Mikhail Syromyatnikov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Pavel Chizhkov
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Evgeny Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
| | - Vasily Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| |
Collapse
|
65
|
Xiao Y, Su D, Hu X, Yang G, Shan Y. Neohesperidin Dihydrochalcone Ameliorates High-Fat Diet-Induced Glycolipid Metabolism Disorder in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9421-9431. [PMID: 35862634 DOI: 10.1021/acs.jafc.2c03574] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
High-fat diet (HFD) is closely related to the formation of metabolic diseases. Studies have confirmed that neohesperidin dihydrochalcone (NHDC) possesses the biological activity of preventing glycolipid metabolism disorder. To explore the mechanism of its preventive activity against glucolipid metabolism disorder, HFD-treated rats were orally administered with NHDC for 12 weeks continuously. The results showed that, compared with the HFD group, the intervention of 40-80 mg/kg body weight of NHDC effectively downregulated the level of fasting blood glucose. Western blot analysis revealed that the treatment of NHDC alleviated the inhibitory effect of HFD on the expression of hepatic GLUT-4 and IRS-1. Further studies confirmed that NHDC reduced the degree of HFD-stimulated inflammation of ileum through the TLR4/MyD88/NF-κB signaling pathway. Moreover, ileum intestinal flora analysis showed that intragastric administration of NHDC reversed the change of Proteobacteria abundance and the Firmicutes/Bacteroidetes (F/B) ratio caused by HFD. At the generic level, NHDC promoted the relative abundance of Coprococcus, Bifidobacterium, Clostridium, Oscillospira, and [Eubacterium], while reducing the relative abundance of Defluviitalea and Prevotella. Taken together, these findings suggest that NHDC possesses the biological activity of improving HFD-induced glycolipid metabolism disorder.
Collapse
Affiliation(s)
- Yecheng Xiao
- Longping Branch Graduate School, Hunan University, Changsha, Hunan 410125, China
- Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan 410125, China
| | - Donglin Su
- Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan 410125, China
| | - Xing Hu
- Lianyuan Kanglu Biotech Co., Ltd., Lianyuan, Hunan 417100, China
| | - Guliang Yang
- National Engineering Laboratory for Rice and By-Products Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Yang Shan
- Longping Branch Graduate School, Hunan University, Changsha, Hunan 410125, China
- Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan 410125, China
| |
Collapse
|
66
|
Koopen A, Witjes J, Wortelboer K, Majait S, Prodan A, Levin E, Herrema H, Winkelmeijer M, Aalvink S, Bergman JJGHM, Havik S, Hartmann B, Levels H, Bergh PO, van Son J, Balvers M, Bastos DM, Stroes E, Groen AK, Henricsson M, Kemper EM, Holst J, Strauch CM, Hazen SL, Bäckhed F, De Vos WM, Nieuwdorp M, Rampanelli E. Duodenal Anaerobutyricum soehngenii infusion stimulates GLP-1 production, ameliorates glycaemic control and beneficially shapes the duodenal transcriptome in metabolic syndrome subjects: a randomised double-blind placebo-controlled cross-over study. Gut 2022; 71:1577-1587. [PMID: 34697034 PMCID: PMC9279853 DOI: 10.1136/gutjnl-2020-323297] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/09/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Although gut dysbiosis is increasingly recognised as a pathophysiological component of metabolic syndrome (MetS), the role and mode of action of specific gut microbes in metabolic health remain elusive. Previously, we identified the commensal butyrogenic Anaerobutyricum soehngenii to be associated with improved insulin sensitivity in subjects with MetS. In this proof-of-concept study, we investigated the potential therapeutic effects of A. soehngenii L2-7 on systemic metabolic responses and duodenal transcriptome profiles in individuals with MetS. DESIGN In this randomised double-blind placebo-controlled cross-over study, 12 male subjects with MetS received duodenal infusions of A. soehngenii/ placebo and underwent duodenal biopsies, mixed meal tests (6 hours postinfusion) and 24-hour continuous glucose monitoring. RESULTS A. soehngenii treatment provoked a markedly increased postprandial excursion of the insulinotropic hormone glucagon-like peptide 1 (GLP-1) and an elevation of plasma secondary bile acids, which were positively associated with GLP-1 levels. Moreover, A. soehngenii treatment robustly shaped the duodenal expression of 73 genes, with the highest fold induction in the expression of regenerating islet-protein 1B (REG1B)-encoding gene. Strikingly, duodenal REG1B expression positively correlated with GLP-1 levels and negatively correlated with peripheral glucose variability, which was significantly diminished in the 24 hours following A. soehngenii intake. Mechanistically, Reg1B expression is induced upon sensing butyrate or bacterial peptidoglycan. Importantly, A. soehngenii duodenal administration was safe and well tolerated. CONCLUSIONS A single dose of A. soehngenii improves peripheral glycaemic control within 24 hours; it specifically stimulates intestinal GLP-1 production and REG1B expression. Further studies are needed to delineate the specific pathways involved in REG1B induction and function in insulin sensitivity. TRIAL REGISTRATION NUMBER NTR-NL6630.
Collapse
Affiliation(s)
- Annefleur Koopen
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Julia Witjes
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Koen Wortelboer
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Soumia Majait
- Clinical Pharmacy, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Andrei Prodan
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Evgeni Levin
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Hilde Herrema
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Steven Aalvink
- Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Stephan Havik
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Bolette Hartmann
- Biomedical Sciences, University of Copenhagen Novo Nordisk Foundation Center for Basic Metabolic Research, Kobenhavn, Denmark
| | - Han Levels
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Per-Olof Bergh
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Goteborg, Sweden
| | - Jamie van Son
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Manon Balvers
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | | | - Erik Stroes
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Albert K Groen
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Marcus Henricsson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Goteborg, Sweden
| | | | - Jens Holst
- Biomedical Sciences, University of Copenhagen Novo Nordisk Foundation Center for Basic Metabolic Research, Kobenhavn, Denmark
| | - Christopher M Strauch
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stanley L Hazen
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fredrik Bäckhed
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Goteborg, Sweden
| | - Willem M De Vos
- Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Elena Rampanelli
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| |
Collapse
|
67
|
Lactobacillus plantarum ZY08 relieves chronic alcohol-induced hepatic steatosis and liver injury in mice via restoring intestinal flora homeostasis. Food Res Int 2022; 157:111259. [DOI: 10.1016/j.foodres.2022.111259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022]
|
68
|
Amin H, Šantl-Temkiv T, Cramer C, Vestergaard DV, Holst GJ, Elholm G, Finster K, Bertelsen RJ, Schlünssen V, Sigsgaard T, Marshall IPG. Cow Farmers’ Homes Host More Diverse Airborne Bacterial Communities Than Pig Farmers’ Homes and Suburban Homes. Front Microbiol 2022; 13:883991. [PMID: 35847077 PMCID: PMC9278274 DOI: 10.3389/fmicb.2022.883991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/05/2022] [Indexed: 01/04/2023] Open
Abstract
Living on a farm has been linked to a lower risk of immunoregulatory disorders, such as asthma, allergy, and inflammatory bowel disease. It is hypothesized that a decrease in the diversity and composition of indoor microbial communities is a sensible explanation for the upsurge in immunoregulatory diseases, with airborne bacteria contributing to this protective effect. However, the composition of this potentially beneficial microbial community in various farm and suburban indoor environments is still to be characterized. We collected settled airborne dust from stables and the associated farmers’ homes and from suburban homes using electrostatic dust collectors (EDCs) over a period of 14 days. Then, quantitative PCR (qPCR) was used to assess bacterial abundance. The V3–V4 region of the bacterial 16S rRNA gene was amplified and sequenced using Ilumina MiSeq in order to assess microbial diversity. The Divisive Amplicon Denoising Algorithm (DADA2) algorithm was used for the inference of amplicon sequence variants from amplicon data. Airborne bacteria were significantly more abundant in farmers’ indoor environments than in suburban homes (p < 0.001). Cow farmers’ homes had significantly higher bacterial diversity than pig farmers’ and suburban homes (p < 0.001). Bacterial taxa, such as Firmicutes, Prevotellaceae, Lachnospiraceae, and Lactobacillus were significantly more abundant in farmers’ homes than suburban homes, and the same was true for beneficial intestinal bacterial species, such as Lactobacillus amylovorus, Eubacterium hallii, and Faecalibacterium prausnitzii. Furthermore, we found a higher similarity between bacterial communities in individual farmers’ homes and their associated cow stables than for pig stables. Our findings contribute with important knowledge on bacterial composition, abundance, and diversity in different environments, which is highly valuable in the discussion on how microbial exposure may contribute to the development of immune-mediated diseases in both children and adults.
Collapse
Affiliation(s)
- Hesham Amin
- Department of Clinical Science, University of Bergen, Bergen, Norway
- *Correspondence: Hesham Amin,
| | - Tina Šantl-Temkiv
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
| | - Christine Cramer
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
- Department of Occupational Medicine, Danish Ramazzini Center, Aarhus University Hospital, Aarhus, Denmark
| | - Ditte V. Vestergaard
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Gitte J. Holst
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Grethe Elholm
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Kai Finster
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
| | | | - Vivi Schlünssen
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
- The National Research Center for the Working Environment, Copenhagen, Denmark
| | - Torben Sigsgaard
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Ian P. G. Marshall
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
69
|
Huang C, Yi P, Zhu M, Zhou W, Zhang B, Yi X, Long H, Zhang G, Wu H, Tsokos GC, Zhao M, Lu Q. Safety and efficacy of fecal microbiota transplantation for treatment of systemic lupus erythematosus: An EXPLORER trial. J Autoimmun 2022; 130:102844. [PMID: 35690527 DOI: 10.1016/j.jaut.2022.102844] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022]
Abstract
Gut microbiota dysbiosis is involved in the development of systemic lupus erythematosus (SLE). The safety and efficacy of fecal microbiota transplantation (FMT) for the treatment of SLE patients has not been explored. In this 12-week, single-arm pilot clinical trial of oral encapsulated fecal microbiome from healthy donors to patients with active SLE, we aimed to evaluate the safety and efficacy of FMT in patients with SLE (ChiCTR2000036352). 20 SLE patients with SLEDAI ≥6 were recruited. FMT was administered once a week for three consecutive weeks along with standard treatment and the patients were followed for 12 weeks. Safety was evaluated throughout the trial. The primary endpoint was the SLE Responder Index-4 (SRI-4) at week 12. Microbiome composition, levels of short chain fatty acids (SCFAs) in the gut and of cytokines in the sera were measured along with lymphocyte phenotyping. No serious adverse events were observed after FMT. At week 12, the SRI-4 response rate was 42.12%, and significant reductions in the SLEDAI-2K scores and the level of serum anti-dsDNA antibody were observed compared to baseline. Significant enrichment of SCFAs-producing bacterial taxa and reduction of inflammation-related bacterial taxa were observed, along with increased production of SCFAs in the gut and reduced levels of IL-6 and CD4+ memory/naïve ratio in the peripheral blood. Furthermore, SRI-4 responding patients displayed specific microbiota signatures both before and after FMT. The first clinical trial of FMT in active SLE patients provide supportive evidence that FMT might be a feasible, safe, and potentially effective therapy in SLE patients by modifying the gut microbiome and its metabolic profile.
Collapse
Affiliation(s)
- Cancan Huang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Ping Yi
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Ming Zhu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Wenhui Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Bo Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Xiaoqing Yi
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Guiying Zhang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China.
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
70
|
Pianko MJ, Golob JL. Host-microbe interactions and outcomes in multiple myeloma and hematopoietic stem cell transplantation. Cancer Metastasis Rev 2022; 41:367-382. [PMID: 35488106 PMCID: PMC9378527 DOI: 10.1007/s10555-022-10033-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/16/2022] [Indexed: 12/12/2022]
Abstract
Microbiota are essential to normal immune development and there is growing recognition of its importance to human health and disease and deepening understanding of the complexity of host-microbe interactions in the human gut and other tissues. Commensal microbes not only can influence host immunity locally through impacts of bioactive microbial metabolites and direct interactions with epithelial cells and innate immune receptors but also can exert systemic immunomodulatory effects via impacts on host immune cells capable of trafficking beyond the gut. Emerging data suggest microbiota influence the development of multiple myeloma (MM), a malignancy of the immune system derived from immunoglobulin-producing bone marrow plasma cells, through the promotion of inflammation. Superior treatment outcomes for MM correlate with a higher abundance of commensal microbiota capable of influencing inflammatory responses through the production of butyrate. In patients with hematologic malignancies, higher levels of diversity of the gut microbiota correlate with superior outcomes after hematopoietic stem cell transplantation. Correlative data support the impact of commensal microbiota on survival, risk of infection, disease relapse, and graft-versus-host disease (GVHD) after transplant. In this review, we will discuss the current understanding of the role of host-microbe interactions and the inflammatory tumor microenvironment of multiple myeloma, discuss data describing the key role of microbiota in hematopoietic stem cell transplantation for treatment of hematologic malignancies, and highlight several possible concepts for interventions directed at the gut microbiota to influence treatment outcomes.
Collapse
Affiliation(s)
- Matthew J Pianko
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.
| | - Jonathan L Golob
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology & Immunology, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
71
|
Chakaroun R, Massier L, Musat N, Kovacs P. New Paradigms for Familiar Diseases: Lessons Learned on Circulatory Bacterial Signatures in Cardiometabolic Diseases. Exp Clin Endocrinol Diabetes 2022; 130:313-326. [PMID: 35320847 DOI: 10.1055/a-1756-4509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Despite the strongly accumulating evidence for microbial signatures in metabolic tissues, including the blood, suggesting a novel paradigm for metabolic disease development, the notion of a core blood bacterial signature in health and disease remains a contentious concept. Recent studies clearly demonstrate that under a strict contamination-free environment, methods such as 16 S rRNA gene sequencing, fluorescence in-situ hybridization, transmission electron microscopy, and several more, allied with advanced bioinformatics tools, allow unambiguous detection and quantification of bacteria and bacterial DNA in human tissues. Bacterial load and compositional changes in the blood have been reported for numerous disease states, suggesting that bacteria and their components may partially induce systemic inflammation in cardiometabolic disease. This concept has been so far primarily based on measurements of surrogate parameters. It is now highly desirable to translate the current knowledge into diagnostic, prognostic, and therapeutic approaches.This review addresses the potential clinical relevance of a blood bacterial signature pertinent to cardiometabolic diseases and outcomes and new avenues for translational approaches. It discusses pitfalls related to research in low bacterial biomass while proposing mitigation strategies for future research and application approaches.
Collapse
Affiliation(s)
- Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.,Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, Sweden
| | - Lucas Massier
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.,Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Niculina Musat
- Department of Isotope Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.,Deutsches Zentrum für Diabetesforschung eV, Neuherberg, Germany
| |
Collapse
|
72
|
Wang Y, Wu G, Zhao L, Wang W. Nutritional Modulation of Gut Microbiota Alleviates Severe Gastrointestinal Symptoms in a Patient with Post-Acute COVID-19 Syndrome. mBio 2022; 13:e0380121. [PMID: 35254129 PMCID: PMC9040862 DOI: 10.1128/mbio.03801-21] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023] Open
Abstract
With the increase in total coronavirus disease 2019 (COVID-19) infection cases, post-acute COVID-19 syndrome, defined as experiencing ongoing health problems 4 or more weeks after the first severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has become a new arising public health concern. As part of post-acute COVID-19 syndrome, gastrointestinal symptoms might be associated with dysbiosis of the gut microbiota, which has the potential to become a target for intervention. In this study, a patient with post-acute COVID-19 syndrome with long-lasting severe gastrointestinal symptoms was provided 2-month expanded access to a high-fiber formula with investigational new drug (IND) status developed to alleviate COVID-19-related symptoms by modulating the gut microbiota. Symptoms including severe "loss of appetite," palpitation, and anxiety were significantly alleviated by the end of the intervention. The medication dosage for controlling nausea decreased during the intervention. The serum lipid profile, insulin level, and leptin level were improved compared to the baseline values. Significant structural changes of the patient's gut microbiota and reduced microbial fermentation activity in the small intestine were found during the intervention. Eighteen amplicon sequence variants (ASVs) of the V4 region of the 16S rRNA gene significantly responded to this nutritional intervention. Six out of the 18 ASVs were also found to be negatively correlated with symptom severity/medication dosage. Five of the six ASVs (ASV0AKS_Oscillibacter, ASV009F_Anaerofustis, ASV02YT_Blautia, ASV07LA_Blautia, and ASV0AM6_Eubacterium hallii) were potential short-chain fatty acid (SCFA)-producing bacteria, which might be associated with the alleviation of symptoms. Our study indicates the feasibility of alleviating gastrointestinal symptoms in patients with post-acute COVID-19 syndrome by way of nutritional modulation of their gut microbiota. IMPORTANCE It has become evident that the care of patients with COVID-19 does not end at the time of negative SARS-CoV-2 detection, as the number of patients with post-acute COVID-19 syndrome increases with an ever-increasing total infected patient population. This case report shows the possibility of alleviating the gastrointestinal symptoms of post-acute COVID-19 syndrome via microbiota-targeted nutritional intervention. As a promising strategy, it might not only improve the quality of life of patients but also reduce the burden to the public health system when the end of the COVID-19 pandemic is not in sight.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Guojun Wu
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Liping Zhao
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Weizheng Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
73
|
Li J, Li Y, Ivey KL, Wang DD, Wilkinson JE, Franke A, Lee KH, Chan AT, Huttenhower C, Hu FB, Rimm EB, Sun Q. Interplay between diet and gut microbiome, and circulating concentrations of trimethylamine N-oxide: findings from a longitudinal cohort of US men. Gut 2022; 71:724-733. [PMID: 33926968 PMCID: PMC8553812 DOI: 10.1136/gutjnl-2020-322473] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Gut-produced trimethylamine N-oxide (TMAO) is postulated as a possible link between red meat intake and poor cardiometabolic health. We investigated whether gut microbiome could modify associations of dietary precursors with TMAO concentrations and cardiometabolic risk markers among free-living individuals. DESIGN We collected up to two pairs of faecal samples (n=925) and two blood samples (n=473), 6 months apart, from 307 healthy men in the Men's Lifestyle Validation Study. Diet was assessed repeatedly using food-frequency questionnaires and diet records. We profiled faecal metagenome and metatranscriptome using shotgun sequencing and identified microbial taxonomic and functional features. RESULTS TMAO concentrations were associated with the overall microbial compositions (permutational analysis of variance (PERMANOVA) test p=0.001). Multivariable taxa-wide association analysis identified 10 bacterial species whose abundance was significantly associated with plasma TMAO concentrations (false discovery rate <0.05). Higher habitual intake of red meat and choline was significantly associated with higher TMAO concentrations among participants who were microbial TMAO-producers (p<0.05), as characterised based on four abundant TMAO-predicting species, but not among other participants (for red meat, P-interaction=0.003; for choline, P-interaction=0.03). Among abundant TMAO-predicting species, Alistipes shahii significantly strengthened the positive association between red meat intake and HbA1c levels (P-interaction=0.01). Secondary analyses revealed that some functional features, including choline trimethylamine-lyase activating enzymes, were associated with TMAO concentrations. CONCLUSION We identified microbial taxa that were associated with TMAO concentrations and modified the associations of red meat intake with TMAO concentrations and cardiometabolic risk markers. Our data underscore the interplay between diet and gut microbiome in producing potentially bioactive metabolites that may modulate cardiometabolic health.
Collapse
Affiliation(s)
- Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Kerry L. Ivey
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Microbiome and Host Health Program, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Dong D. Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jeremy E. Wilkinson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Adrian Franke
- Analytical Biochemistry Shared Resource, University of Hawaii Cancer Center, Honolulu, HI
| | - Kyu Ha Lee
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Andrew T. Chan
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Eric B. Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Qi Sun
- Nutrition, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA .,Epidemiology, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
74
|
Yang F, Zhu W, Edirisuriya P, Ai Q, Nie K, Ji X, Zhou K. Characterization of metabolites and biomarkers for the probiotic effects of Clostridium cochlearium on high-fat diet-induced obese C57BL/6 mice. Eur J Nutr 2022; 61:2217-2229. [DOI: 10.1007/s00394-022-02840-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/11/2022] [Indexed: 12/17/2022]
|
75
|
Le Roy T, Moens de Hase E, Van Hul M, Paquot A, Pelicaen R, Régnier M, Depommier C, Druart C, Everard A, Maiter D, Delzenne NM, Bindels LB, de Barsy M, Loumaye A, Hermans MP, Thissen JP, Vieira-Silva S, Falony G, Raes J, Muccioli GG, Cani PD. Dysosmobacter welbionis is a newly isolated human commensal bacterium preventing diet-induced obesity and metabolic disorders in mice. Gut 2022; 71:534-543. [PMID: 34108237 PMCID: PMC8862106 DOI: 10.1136/gutjnl-2020-323778] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/20/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To investigate the abundance and the prevalence of Dysosmobacter welbionis J115T, a novel butyrate-producing bacterium isolated from the human gut both in the general population and in subjects with metabolic syndrome. To study the impact of this bacterium on host metabolism using diet-induced obese and diabetic mice. DESIGN We analysed the presence and abundance of the bacterium in 11 984 subjects using four human cohorts (ie, Human Microbiome Project, American Gut Project, Flemish Gut Flora Project and Microbes4U). Then, we tested the effects of daily oral gavages with live D. welbionis J115T on metabolism and several hallmarks of obesity, diabetes, inflammation and lipid metabolism in obese/diabetic mice. RESULTS This newly identified bacterium was detected in 62.7%-69.8% of the healthy population. Strikingly, in obese humans with a metabolic syndrome, the abundance of Dysosmobacter genus correlates negatively with body mass index, fasting glucose and glycated haemoglobin. In mice, supplementation with live D. welbionis J115T, but not with the pasteurised bacteria, partially counteracted diet-induced obesity development, fat mass gain, insulin resistance and white adipose tissue hypertrophy and inflammation. In addition, live D. welbionis J115T administration protected the mice from brown adipose tissue inflammation in association with increased mitochondria number and non-shivering thermogenesis. These effects occurred with minor impact on the mouse intestinal microbiota composition. CONCLUSIONS These results suggest that D. welbionis J115T directly and beneficially influences host metabolism and is a strong candidate for the development of next-generation beneficial bacteria targeting obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Tiphaine Le Roy
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Emilie Moens de Hase
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Adrien Paquot
- Louvain Drug Research Institute (LDRI), Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Rudy Pelicaen
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marion Régnier
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Clara Depommier
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Céline Druart
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Dominique Maiter
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marie de Barsy
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Audrey Loumaye
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Michel P Hermans
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium,Center for Microbiology, VIB, Leuven, Belgium
| | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium,Center for Microbiology, VIB, Leuven, Belgium
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium,Center for Microbiology, VIB, Leuven, Belgium
| | - Giulio G Muccioli
- Louvain Drug Research Institute (LDRI), Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
76
|
Corona-Cervantes K, Parra-Carriedo A, Hernández-Quiroz F, Martínez-Castro N, Vélez-Ixta JM, Guajardo-López D, García-Mena J, Hernández-Guerrero C. Physical and Dietary Intervention with Opuntia ficus-indica (Nopal) in Women with Obesity Improves Health Condition through Gut Microbiota Adjustment. Nutrients 2022; 14:1008. [PMID: 35267983 PMCID: PMC8912383 DOI: 10.3390/nu14051008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity is a multifactorial disease resulting in excessive accumulation of fat. Worldwide, obesity is an important public health problem, affecting a large proportion of the world population. The tender cactus Opuntia ficus-indica, commonly known in Mexico as "nopal", is widely distributed in this country, Latin America, South Africa, and the Mediterranean area. Nopal cladodes are commonly marketed in different forms as fresh, frozen, or pre-cooked, and used as fresh green vegetable. The aim of this study was to evaluate the capability of nopal to improve the health condition of participants affected by obesity, in a physical and dietary intervention, through gut microbiota modification. These results were contrasted with the effect of nopal in the gut microbiota of normal weight participants. We describe the association among biochemical, anthropometric markers, and the gut microbiota diversity found in fecal samples of the obese and normal weight groups. The results presented in this work suggest that caloric restriction, addition of nopal to the diet and physical activity, promote changes in the gut microbiota in obese women, improving the host metabolism, as suggested by the correlation between some bacterial species with biochemical and anthropometrical parameters.
Collapse
Affiliation(s)
- Karina Corona-Cervantes
- Departamento de Genética y Biología Molecular, Cinvestav, Avenida IPN 2508, Ciudad de Mexico 07360, Mexico; (K.C.-C.); (F.H.-Q.); (J.M.V.-I.); (J.G.-M.)
| | - Alicia Parra-Carriedo
- Departamento de Salud, Universidad Iberoamericana, Ciudad de México, Paseo de la Reforma 880, Ciudad de Mexico 01219, Mexico; (A.P.-C.); (N.M.-C.); (D.G.-L.)
| | - Fernando Hernández-Quiroz
- Departamento de Genética y Biología Molecular, Cinvestav, Avenida IPN 2508, Ciudad de Mexico 07360, Mexico; (K.C.-C.); (F.H.-Q.); (J.M.V.-I.); (J.G.-M.)
| | - Noemí Martínez-Castro
- Departamento de Salud, Universidad Iberoamericana, Ciudad de México, Paseo de la Reforma 880, Ciudad de Mexico 01219, Mexico; (A.P.-C.); (N.M.-C.); (D.G.-L.)
| | - Juan Manuel Vélez-Ixta
- Departamento de Genética y Biología Molecular, Cinvestav, Avenida IPN 2508, Ciudad de Mexico 07360, Mexico; (K.C.-C.); (F.H.-Q.); (J.M.V.-I.); (J.G.-M.)
| | - Diana Guajardo-López
- Departamento de Salud, Universidad Iberoamericana, Ciudad de México, Paseo de la Reforma 880, Ciudad de Mexico 01219, Mexico; (A.P.-C.); (N.M.-C.); (D.G.-L.)
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Avenida IPN 2508, Ciudad de Mexico 07360, Mexico; (K.C.-C.); (F.H.-Q.); (J.M.V.-I.); (J.G.-M.)
| | - César Hernández-Guerrero
- Departamento de Salud, Universidad Iberoamericana, Ciudad de México, Paseo de la Reforma 880, Ciudad de Mexico 01219, Mexico; (A.P.-C.); (N.M.-C.); (D.G.-L.)
| |
Collapse
|
77
|
Jing Y, Yuan Y, Monson M, Wang P, Mu F, Zhang Q, Na W, Zhang K, Wang Y, Leng L, Li Y, Luan P, Wang N, Guo R, Lamont SJ, Li H, Yuan H. Multi-Omics Association Reveals the Effects of Intestinal Microbiome-Host Interactions on Fat Deposition in Broilers. Front Microbiol 2022; 12:815538. [PMID: 35250914 PMCID: PMC8892104 DOI: 10.3389/fmicb.2021.815538] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Growing evidence indicates that gut microbiota factors cannot be viewed as independent in the occurrence of obesity. Because the gut microbiome is highly dimensional and complex, studies on interactions between gut microbiome and host in obesity are still rare. To explore the relationship of gut microbiome-host interactions with obesity, we performed multi-omics associations of gut metagenome, intestinal transcriptome, and host obesity phenotypes in divergently selected obese-lean broiler lines. Metagenomic shotgun sequencing generated a total of 450 gigabases of clean data from 80 intestinal segment contents of 20 broilers (10 of each line). The microbiome comparison showed that microbial diversity and composition in the duodenum, jejunum, ileum, and ceca were altered variously between the lean- and fat-line broilers. We identified two jejunal microbes (Escherichia coli and Candidatus Acetothermia bacterium) and four cecal microbes (Alistipes sp. CHKCI003, Ruminococcaceae bacterium CPB6, Clostridiales bacterium, and Anaeromassilibacillus sp. An200), which were significantly different between the two lines (FDR < 0.05). When comparing functional metagenome, the fat-line broilers had an intensive microbial metabolism in the duodenum and jejunum but degenerative microbial activities in the ileum and ceca. mRNA-sequencing identified a total of 1,667 differentially expressed genes (DEG) in the four intestinal compartments between the two lines (| log2FC| > 1.5 and FDR < 0.05). Multi-omics associations showed that the 14 microbial species with abundances that were significantly related with abdominal fat relevant traits (AFRT) also have significant correlations with 155 AFRT-correlated DEG (p < 0.05). These DEG were mainly involved in lipid metabolism, immune system, transport and catabolism, and cell growth-related pathways. The present study constructed a gut microbial gene catalog of the obese-lean broiler lines. Intestinal transcriptome and metagenome comparison between the two lines identified candidate DEG and differential microbes for obesity, respectively. Multi-omics associations suggest that abdominal fat deposition may be influenced by the interactions of specific gut microbiota abundance and the expression of host genes in the intestinal compartments in which the microbes reside. Our study explored the interactions between gut microbiome and host intestinal gene expression in lean and obese broilers, which may expand knowledge on the relationships between obesity and gut microbiome.
Collapse
Affiliation(s)
- Yang Jing
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yuqi Yuan
- Novogene Bioinformatics Institute, Beijing, China
| | - Melissa Monson
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Peng Wang
- Novogene Bioinformatics Institute, Beijing, China
| | - Fang Mu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Qi Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Wei Na
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ke Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yuxiang Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Li Leng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yumao Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Peng Luan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ning Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Rongjun Guo
- Novogene Bioinformatics Institute, Beijing, China
| | - Susan J. Lamont
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Hui Yuan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
78
|
Voland L, Le Roy T, Debédat J, Clément K. Gut microbiota and vitamin status in persons with obesity: A key interplay. Obes Rev 2022; 23:e13377. [PMID: 34767276 DOI: 10.1111/obr.13377] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/28/2022]
Abstract
There are numerous factors involved in obesity progression and maintenance including systemic low-grade inflammation, adipose tissue dysfunction, or gut microbiota dysbiosis. Recently, a growing interest has arisen for vitamins' role in obesity and related disorders, both at the host and gut bacterial level. Indeed, vitamins are provided mostly by food, but some, from the B and K groups in particular, can be synthesized by the gut bacterial ecosystem and absorbed in the colon. Knowing that vitamin deficiency can alter many important cellular functions and lead to serious health issues, it is important to carefully monitor the vitamin status of patients with obesity and potentially already existing comorbidities as well as to examine the dysbiotic gut microbiota and thus potentially altered bacterial metabolism of vitamins. In this review, we examined both murine and human studies, to assess the prevalence of sub-optimal levels of several vitamins in obesity and metabolic alterations. This review also examines the relationship between vitamins and the gut microbiota in terms of vitamin production and the modulation of the gut bacterial ecosystem in conditions of vitamin shortage or supplementation. Furthermore, some strategies to improve vitamin status of patients with severe obesity are proposed within this review.
Collapse
Affiliation(s)
- Lise Voland
- Inserm, Sorbonne University, Nutrition and obesities: systemic approaches (NutriOmics), Paris, France
| | - Tiphaine Le Roy
- Inserm, Sorbonne University, Nutrition and obesities: systemic approaches (NutriOmics), Paris, France
| | - Jean Debédat
- Inserm, Sorbonne University, Nutrition and obesities: systemic approaches (NutriOmics), Paris, France
| | - Karine Clément
- Inserm, Sorbonne University, Nutrition and obesities: systemic approaches (NutriOmics), Paris, France.,Public hospital of Paris, Nutrition department, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
79
|
De Filippis F, Esposito A, Ercolini D. Outlook on next-generation probiotics from the human gut. Cell Mol Life Sci 2022; 79:76. [PMID: 35043293 PMCID: PMC11073307 DOI: 10.1007/s00018-021-04080-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022]
Abstract
Probiotics currently available on the market generally belong to a narrow range of microbial species. However, recent studies about the importance of the gut microbial commensals on human health highlighted that the gut microbiome is an unexplored reservoir of potentially beneficial microbes. For this reason, academic and industrial research is focused on identifying and testing novel microbial strains of gut origin for the development of next-generation probiotics. Although several of these are promising for the prevention and treatment of many chronic diseases, studies on human subjects are still scarce and approval from regulatory agencies is, therefore, rare. In addition, some issues need to be overcome before implementing their wide application on the market, such as the best methods for cultivation and storage of these oxygen-sensitive taxa. This review summarizes the most recent evidence related to NGPs and provides an outlook to the main issues that still limit their wide employment.
Collapse
Affiliation(s)
- Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Alessia Esposito
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy.
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
80
|
Zhang Q, Hu WM, Deng YL, Wan JJ, Wang YJ, Jin P. Dysbiosis of gut microbiota and decreased propionic acid associated with metabolic abnormality in Cushing's syndrome. Front Endocrinol (Lausanne) 2022; 13:1095438. [PMID: 36755580 PMCID: PMC9901362 DOI: 10.3389/fendo.2022.1095438] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Chronic hypercortisolism leads to a phenotype resembling metabolic syndrome. We aimed to investigate the association between gut microbiota and metabolic abnormalities in endogenous hypercortisolism (Cushing's syndrome). METHODS A total of 23 patients with Cushing's syndrome (18 female and 5 men, aged 47.24 ± 12.99 years) and 30 age-, sex-and BMI-matched healthy controls (18 female and 12 men, aged 45.03 ± 6.69 years) were consecutively recruited. Differences in gut microbiota and plasma short-chain fatty acid (SCFAs) concentrations between the Cushing's syndrome patients and controls were analyzed by 16S rRNA sequencing and gas chromatography-mass spectrometry (GC-MS). RESULTS Compared to the controls, the Simpson and Pielou indices of α diversity were dramatically decreased in Cushing's syndrome (P < 0.05). The gut microbiota community structure differed significantly between Cushing's syndrome patients and controls. Compared to controls, the bacterial communities of the Cushing's syndrome patients were enriched in Proteobacteria and Escherichia-Shigella, and depleted in Firmicutes, including Agathobacter, Blautia, Anaerostipes, Eubacterium_eligens_group, and Lachnospira. Spearman analysis demonstrated that HbA1c, SBP, DBP, and cortisol levels were significantly positively correlated with Proteobacteria and Escherichia-Shigella, whereas negatively correlated with Agathobacter, Blautia, Anaerostipes, Eubacterium_hallii_group, and Lachnospira, etc. Cushing's syndrome patients also had a lower propionic acid concentration (0.151±0.054 vs. 0.205±0.032 µg/mL, P=0.039) than controls. Furthermore, the level of propionic acid was negatively correlated with systolic pressure and cortisol levels (P<0.05). CONCLUSION Gut microbiota dysbiosis and decreased propionic acid levels were observed in patients with Cushing's, suggesting that the gut microbiota may be a potential therapeutic intervention target to improve hypercortisolism-related metabolic abnormalities.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Wen-mu Hu
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Yu-ling Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jin-jing Wan
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Yu-jun Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Ping Jin
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- *Correspondence: Ping Jin,
| |
Collapse
|
81
|
Zhang X, Liu L, Luo J, Peng X. Anti-aging potency correlates with metabolites from in vitro fermentation of edible fungal polysaccharides using human fecal intestinal microflora. Food Funct 2022; 13:11592-11603. [DOI: 10.1039/d2fo01951e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aging is a natural process in which the structural integrity of an organism declines over time.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jianming Luo
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| |
Collapse
|
82
|
Wang D, Doestzada M, Chen L, Andreu-Sánchez S, van den Munckhof ICL, Augustijn HE, Koehorst M, Ruiz-Moreno AJ, Bloks VW, Riksen NP, Rutten JHW, Joosten LAB, Netea MG, Wijmenga C, Zhernakova A, Kuipers F, Fu J. Characterization of gut microbial structural variations as determinants of human bile acid metabolism. Cell Host Microbe 2021; 29:1802-1814.e5. [PMID: 34847370 DOI: 10.1016/j.chom.2021.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/06/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022]
Abstract
Bile acids (BAs) facilitate intestinal fat absorption and act as important signaling molecules in host-gut microbiota crosstalk. BA-metabolizing pathways in the microbial community have been identified, but it remains largely unknown how the highly variable genomes of gut bacteria interact with host BA metabolism. We characterized 8,282 structural variants (SVs) of 55 bacterial species in the gut microbiomes of 1,437 individuals from two cohorts and performed a systematic association study with 39 plasma BA parameters. Both variations in SV-based continuous genetic makeup and discrete clusters showed correlations with BA metabolism. Metagenome-wide association analysis identified 809 replicable associations between bacterial SVs and BAs and SV regulators that mediate the effects of lifestyle factors on BA metabolism. This is the largest microbial genetic association analysis to demonstrate the impact of bacterial SVs on human BA composition, and it highlights the potential of targeting gut microbiota to regulate BA metabolism through lifestyle intervention.
Collapse
Affiliation(s)
- Daoming Wang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Marwah Doestzada
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Lianmin Chen
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Inge C L van den Munckhof
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Hannah E Augustijn
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Martijn Koehorst
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen 9713AV, the Netherlands
| | - Angel J Ruiz-Moreno
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Vincent W Bloks
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands; Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn 53113, Germany; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova 200349, Romania
| | - Cisca Wijmenga
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands
| | - Folkert Kuipers
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen 9713AV, the Netherlands
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen 9713AV, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen 9713AV, the Netherlands.
| |
Collapse
|
83
|
Kumari M, Singh P, Nataraj BH, Kokkiligadda A, Naithani H, Azmal Ali S, Behare PV, Nagpal R. Fostering next-generation probiotics in human gut by targeted dietary modulation: An emerging perspective. Food Res Int 2021; 150:110716. [PMID: 34865747 DOI: 10.1016/j.foodres.2021.110716] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/07/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022]
Abstract
Emerging evidence and an in-depth understanding of the microbiome have helped in identifying beneficial commensals and their therapeutic potentials. Specific commensal taxa/ strains of the human gut microbiome have been positively associated with human health and recently termed as next-generation probiotics (NGPs). Of these, Akkermansia muciniphila, Ruminococcus bromii, Faecalibacterium prausnitzii, Anaerobutyricum hallii, and Roseburia intestinalis are the five most relevant gut-derived NGPs that have demonstrated therapeutic potential in managing metabolic diseases. Specific and natural dietary interventions can modulate the abundance and activity of these beneficial bacteria in the gut. Hence, the understanding of targeted stimulation of specific NGP by specific probiotic-targeted diets (PTD) is indispensable for the rational application of their combination. The supplementation of NGP with its specific PTD will help the strain(s) to compete with harmful microbes and acquire its niche. This combination would enhance the effectiveness of NGPs to be used as "live biotherapeutic products" or food nutraceuticals. Under the current milieu, we review various PTDs that influence the abundance of specific potential NGPs, and contemplates potential interactions between diet, microbes, and their effects on host health. Taking into account the study mentioned, we propose that combining NGPs will provide an alternate solution for developing the new diet in conjunction with PTD.
Collapse
Affiliation(s)
- Manorama Kumari
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Parul Singh
- Proteomics and Cell Biology Lab, Animal Biotechnology Center, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Basavaprabhu H Nataraj
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Anusha Kokkiligadda
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Harshita Naithani
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Syed Azmal Ali
- Proteomics and Cell Biology Lab, Animal Biotechnology Center, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Pradip V Behare
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India.
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
84
|
Li L, Zhang Y, Speakman JR, Hu S, Song Y, Qin S. The gut microbiota and its products: Establishing causal relationships with obesity related outcomes. Obes Rev 2021; 22:e13341. [PMID: 34490704 DOI: 10.1111/obr.13341] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
Gut microorganisms not only participate in the metabolism of carbohydrate, lipids, protein, and polypeptides in the intestine but also directly affect the metabolic phenotypes of the host. Although many studies have described the apparent effects of gut microbiota on human health, the development of metagenomics and culturomics in the past decade has generated a large amount of evidence suggesting a causal relationship between gut microbiota and obesity. The interaction between the gut microbiota and host is realized by microbial metabolites with multiple biological functions. We concentrated here on several representative beneficial species connected with obesity as well as the mechanisms, with particular emphasis on microbiota-dependent metabolites. Finally, we consider the potential clinical significance of these relationships to fuel the conception and realization of novel therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Lili Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Yubing Zhang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.,College of Life Sciences, Yantai University, Yantai, China
| | - John Roger Speakman
- Shenzhen Key Laboratory for Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shanliang Hu
- Department of Radiotherapy, Yantai Yuhuangding Hospital, Yantai, China
| | - Yipeng Song
- Department of Radiotherapy, Yantai Yuhuangding Hospital, Yantai, China
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|
85
|
Sun S, Sun L, Wang K, Qiao S, Zhao X, Hu X, Chen W, Zhang S, Li H, Dai H, Liu H. The gut commensal fungus, Candida parapsilosis, promotes high fat-diet induced obesity in mice. Commun Biol 2021; 4:1220. [PMID: 34697386 PMCID: PMC8546080 DOI: 10.1038/s42003-021-02753-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/05/2021] [Indexed: 01/07/2023] Open
Abstract
Gut fungi is known to play many important roles in human health regulations. Herein, we investigate the anti-obesity efficacy of the antifungal antibiotics (amphotericin B, fluconazole and 5-fluorocytosine) in the high fat diet-fed (HFD) mice. Supplementation of amphotericin B or fluconazole in water can effectively inhibit obesity and its related disorders, whereas 5-fluorocytosine exhibit little effects. The gut fungus Candida parapsilosis is identified as a key commensal fungus related to the diet-induced obesity by the culture-dependent method and the inoculation assay with C. parapsilosis in the fungi-free mice. In addition, the increase of free fatty acids in the gut due to the production of fungal lipases from C. parapsilosis is confirmed as one mechanism by which C. parapsilosis promotes obesity. The current study demonstrates the gut C. parapsilosis as a causal fungus for the development of diet-induced obesity in mice and highlights the therapeutic strategy targeting the gut fungi. Shanshan Sun, Li Sun, Kai Wang, et al. report that the gut commensal Candida parapsilosis is a causative fungus for the development of high fat-diet induced obesity in mice. Their results suggest that fungi could represent possible targets for combating obesity.
Collapse
Affiliation(s)
- Shanshan Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China.,The Second Hospital of Anhui Medical University, Hefei, China
| | - Li Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinyue Zhao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xiaomin Hu
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Wei Chen
- Department of Clinical Nutrition, Dept. of Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Hantian Li
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
86
|
Aguirre AM, Yalcinkaya N, Wu Q, Swennes A, Tessier ME, Roberts P, Miyajima F, Savidge T, Sorg JA. Bile acid-independent protection against Clostridioides difficile infection. PLoS Pathog 2021; 17:e1010015. [PMID: 34665847 PMCID: PMC8555850 DOI: 10.1371/journal.ppat.1010015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/29/2021] [Accepted: 10/07/2021] [Indexed: 12/21/2022] Open
Abstract
Clostridioides difficile infections occur upon ecological / metabolic disruptions to the normal colonic microbiota, commonly due to broad-spectrum antibiotic use. Metabolism of bile acids through a 7α-dehydroxylation pathway found in select members of the healthy microbiota is regarded to be the protective mechanism by which C. difficile is excluded. These 7α-dehydroxylated secondary bile acids are highly toxic to C. difficile vegetative growth, and antibiotic treatment abolishes the bacteria that perform this metabolism. However, the data that supports the hypothesis that secondary bile acids protect against C. difficile infection is supported only by in vitro data and correlative studies. Here we show that bacteria that 7α-dehydroxylate primary bile acids protect against C. difficile infection in a bile acid-independent manner. We monoassociated germ-free, wildtype or Cyp8b1-/- (cholic acid-deficient) mutant mice and infected them with C. difficile spores. We show that 7α-dehydroxylation (i.e., secondary bile acid generation) is dispensable for protection against C. difficile infection and provide evidence that Stickland metabolism by these organisms consumes nutrients essential for C. difficile growth. Our findings indicate secondary bile acid production by the microbiome is a useful biomarker for a C. difficile-resistant environment but the microbiome protects against C. difficile infection in bile acid-independent mechanisms. Secondary bile acid production by the colonic microbiome strongly correlates with an environment that is resistant to C. difficile invasion. However, it remained unclear if these bile acids provided in vivo protection. Here, we show that members of the microbiome that generate secondary bile acids (e.g., C. scindens) protect against C. difficile disease independently of secondary bile acid generation. These results are important because efforts to restore colonization resistance (e.g., FMT or precision bacterial therapy) focus on restoring secondary bile acid generation. Instead, restoring the organisms that produce 5-aminovalerate or consume proline / glycine are more important.
Collapse
Affiliation(s)
- Andrea Martinez Aguirre
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Nazli Yalcinkaya
- Baylor College of Medicine & Texas Children’s Hospital, Houston, Texas, United States of America
| | - Qinglong Wu
- Baylor College of Medicine & Texas Children’s Hospital, Houston, Texas, United States of America
| | - Alton Swennes
- Baylor College of Medicine & Texas Children’s Hospital, Houston, Texas, United States of America
| | - Mary Elizabeth Tessier
- Baylor College of Medicine & Texas Children’s Hospital, Houston, Texas, United States of America
| | - Paul Roberts
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Fabio Miyajima
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
- Oswaldo Cruz Foundation, Ceara branch, Fortaleza, Brazil
| | - Tor Savidge
- Baylor College of Medicine & Texas Children’s Hospital, Houston, Texas, United States of America
| | - Joseph A. Sorg
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
87
|
Arora T, Tremaroli V. Therapeutic Potential of Butyrate for Treatment of Type 2 Diabetes. Front Endocrinol (Lausanne) 2021; 12:761834. [PMID: 34737725 PMCID: PMC8560891 DOI: 10.3389/fendo.2021.761834] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/23/2021] [Indexed: 12/18/2022] Open
Abstract
Metagenomics studies have shown that type 2 diabetes (T2D) is associated with an altered gut microbiota. Whereas different microbiota patterns have been observed in independent human cohorts, reduction of butyrate-producing bacteria has consistently been found in individuals with T2D, as well as in those with prediabetes. Butyrate is produced in the large intestine by microbial fermentations, particularly of dietary fiber, and serves as primary fuel for colonocytes. It also acts as histone deacetylase inhibitor and ligand to G-protein coupled receptors, affecting cellular signaling in target cells, such as enteroendocrine cells. Therefore, butyrate has become an attractive drug target for T2D, and treatment strategies have been devised to increase its intestinal levels, for example by supplementation of butyrate-producing bacteria and dietary fiber, or through fecal microbiota transplant (FMT). In this review, we provide an overview of current literature indicating that these strategies have yielded encouraging results and short-term benefits in humans, but long-term improvements of glycemic control have not been reported so far. Further studies are required to find effective approaches to restore butyrate-producing bacteria and butyrate levels in the human gut, and to investigate their impact on glucose regulation in T2D.
Collapse
Affiliation(s)
- Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
88
|
He F, Zhang T, Xue K, Fang Z, Jiang G, Huang S, Li K, Gu Z, Shi H, Zhang Z, Zhu H, Lin L, Li J, Xiao F, Shan H, Yan R, Li X, Yan Z. Fecal multi-omics analysis reveals diverse molecular alterations of gut ecosystem in COVID-19 patients. Anal Chim Acta 2021; 1180:338881. [PMID: 34538334 PMCID: PMC8310733 DOI: 10.1016/j.aca.2021.338881] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022]
Abstract
Gut ecosystem has profound effects on host physiology and health. Gastrointestinal (GI) symptoms were frequently observed in patients with COVID-19. Compared with other organs, gut antiviral response can result in more complicated immune responses because of the interactions between the gut microbiota and host immunity. However, there are still large knowledge gaps in the impact of COVID-19 on gut molecular profiles and commensal microbiome, hindering our comprehensive understanding of the pathogenesis of SARS-CoV-2 and the treatment of COVID-19. We performed longitudinal stool multi-omics profiling to systemically investigate the molecular phenomics alterations of gut ecosystem in COVID-19. Gut proteomes of COVID-19 were characterized by disturbed immune, proteolysis and redox homeostasis. The expression and glycosylation of proteins involved in neutrophil degranulation and migration were suppressed, while those of proteases were upregulated. The variable domains of Ig heavy chains were downregulated and the overall glycosylation of IgA heavy chain constant regions, IgGFc-binding protein, and J chain were suppressed with glycan-specific variations. There was a reduction of beneficial gut bacteria and an enrichment of bacteria derived deleterious metabolites potentially associated with multiple types of diseases (such as ethyl glucuronide). The reduction of Ig heave chain variable domains may contribute to the increase of some Bacteroidetes species. Many bacteria ceramide lipids with a C17-sphingoid based were downregulated in COVID-19. In many cases, the gut phenome did not restore two months after symptom onset. Our study indicates widely disturbed gut molecular profiles which may play a role in the development of symptoms in COVID-19. Our findings also emphasis the need for ongoing investigation of the long-term gut molecular and microbial alterations during COVID-19 recovery process. Considering the gut ecosystem as a potential target could offer a valuable approach in managing the disease.
Collapse
Affiliation(s)
- Feixiang He
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China,Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Ting Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Kewen Xue
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Zhaoxiong Fang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Siwen Huang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Kexue Li
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Zhiqiang Gu
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Honggang Shi
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Zhenyi Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Huijin Zhu
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Lu Lin
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Jialin Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Fei Xiao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China,Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao,Corresponding author
| | - Xiaofeng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China,Corresponding author
| | - Zhixiang Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China,Corresponding author
| |
Collapse
|
89
|
Zhu LB, Zhang YC, Huang HH, Lin J. Prospects for clinical applications of butyrate-producing bacteria. World J Clin Pediatr 2021; 10:84-92. [PMID: 34616650 PMCID: PMC8465514 DOI: 10.5409/wjcp.v10.i5.84] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/13/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
As the major source of energy for colonic mucosal cells and as an important regulator of gene expression, inflammation, differentiation, and apoptosis in host cells, microbiota-derived butyrate can enhance the intestinal mucosal immune barrier, modulate systemic immune response, and prevent infections. Maintaining a certain level of butyrate production in the gut can help balance intestinal microbiota, regulate host immune response, and promote the development and maintenance of the intestinal mucosal barrier. Butyrate-producing bacteria act as probiotics and play important roles in a variety of normal biological functions. Bacteriotherapeutic supplementation by using fecal microbiota transplantation to restore butyrate-producing commensal bacteria in the gut has been very successful in the treatment of recurrent and refractory Clostridium difficile (C. difficile) infection or C. difficile-negative nosocomial diarrhea. Administration of probiotics that include butyrate-producing bacteria may have a role in the treatment of inflammatory bowel diseases and in the prevention of necrotizing enterocolitis and late-onset sepsis in premature infants. Furthermore, modulating gut microbiota with dietary approaches may improve intestinal dysbiosis commonly seen in patients with obesity-associated metabolic disorders. Supplementation with a butyrate-producing bacterial stain might be used to increase energy expenditure, improve insulin sensitivity, and to help control obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Li-Bin Zhu
- Department of Pediatric Surgery, The Second Affiliated Hospital & Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Yu-Chen Zhang
- Department of Pediatric Surgery, The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Han-Hui Huang
- Department of Pediatric Surgery, The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Jing Lin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
90
|
Beneficial effects of a combination of Clostridium cochlearium and Lactobacillus acidophilus on body weight gain, insulin sensitivity, and gut microbiota in high-fat diet-induced obese mice. Nutrition 2021; 93:111439. [PMID: 34507264 DOI: 10.1016/j.nut.2021.111439] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/10/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Species Lactobacillus acidophilus and butyrate producer Clostridium cochlearium have been shown to have potential antiobesity effects. The aim of this study was to show that the combination of C. cochlearium and L. acidophilus (CC-LA) has beneficial effects on body weight control and glucose homeostasis in high-fat diet-induced obese (DIO) mice. METHODS In this study, thirty-six 6-wk-old male C57BL/6 mice were randomly assigned to three groups of 12 mice each. The experimental group (CC-LA) was administered with CC-LA mixture and fed ad libitum with a high-fat diet. High-fat diet (HF) control and low-fat diet (LF) control groups were treated with the same dose of sterile water as the CC-LA group. RESULTS After 17 wk of dietary intervention, the CC-LA group showed 17% less body weight gain than the HF group did (P < 0.01). The CC-LA group also showed significantly reduced incremental area under the curve of oral glucose tolerance test and homeostatic model assessment for insulin resistance compared with the HF group. The results from 16S rRNA sequencing analysis of gut microbiota showed that the CC-LA administration led to overall increased α-diversity indices, and a significant microbial separation from the HF group. The ratio of Firmicutes to Bacteroidetes (F/B) was reduced from 3.30 in the HF group to 1.94 in the CC-LA group. The relative abundances of certain obesity-related taxa were also decreased by CC-LA administration. CONCLUSION The present study provided evidence that the CC-LA combination reduced obesity and improved glucose metabolism in high-fat diet-treated DIO mice, potentially mediated by the modulation of gut microbiota.
Collapse
|
91
|
Zhao T, Zhan L, Zhou W, Chen W, Luo J, Zhang L, Weng Z, Zhao C, Liu S. The Effects of Erchen Decoction on Gut Microbiota and Lipid Metabolism Disorders in Zucker Diabetic Fatty Rats. Front Pharmacol 2021; 12:647529. [PMID: 34366839 PMCID: PMC8339961 DOI: 10.3389/fphar.2021.647529] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a chronic metabolic disease caused by genetic and environmental factors that has become a serious global health problem. There is evidence that gut microbiota is closely related to the occurrence and development of obesity. Erchen Decoction (ECD), a traditional Chinese medicine, has been widely used for clinical treatment and basic research of obesity and related metabolic diseases in recent years. It can significantly improve insulin resistance (IR) and lipid metabolism disorders. However, there is no microbiological study on its metabolic regulation. In this study, we investigated the effects of ECD on obesity, especially lipid metabolism and the composition and function of gut microbiota in Zucker diabetic fatty (ZDF) rats, and explored the correlation between the biomarkers of gut microbiota and metabolite and host phenotype. The results showed that ECD could reduce body weight, improve IR and lipid metabolism, and reduce the concentration of free fatty acids (FFA) released from white adipose tissue (WAT) due to excessive lipolysis by interfering with the insulin receptor substrate 1 (IRS1)/protein kinase B (AKT)/protein kinase A (PKA)/hormone-sensitive triglyceride lipase (HSL) signaling pathway in ZDF rats. Additionally, ECD gradually adjusted the overall structure of changed gut microbiota, reversed the relative abundance of six genera, and changed the function of gut microbiota by reducing the content of propionic acid, a metabolite of gut microbiota, in ZDF rats. A potentially close relationship between biomarkers, especially Prevotella, Blautia, and Holdemania, propionic acid and host phenotypes were demonstrated through correlation analysis. The results suggested that the beneficial effects of ECD on obesity, especially lipid metabolism disorders, are related to the regulation of gut microbiota in ZDF rats. This provides a basis for further research on the mechanism and clinical application of ECD to improve obesity via gut microbiota.
Collapse
Affiliation(s)
- Tian Zhao
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Libin Zhan
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen Zhou
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wanxin Chen
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jintong Luo
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lijing Zhang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zebin Weng
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Zhao
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shenlin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
92
|
Anselmi G, Gagliardi L, Egidi G, Leone S, Gasbarrini A, Miggiano GAD, Galiuto L. Gut Microbiota and Cardiovascular Diseases: A Critical Review. Cardiol Rev 2021; 29:195-204. [PMID: 32639240 DOI: 10.1097/crd.0000000000000327] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human intestine contains the largest and most diverse ecosystem of microbes. The main function of the intestinal bacterial flora is to limit the growth of potentially pathogenic microorganisms. However, the intestinal microbiota is increasingly emerging as a risk factor for the development of cardiovascular disease (CVD). The gut microbiota-derived metabolites, such as short-chain fatty acids, trimethylamine-N-oxide, bile acids, and polyphenols play a pivotal role in maintaining healthy cardiovascular function, and when dysregulated, can potentially lead to CVD. In particular, changes in the composition and diversity of gut microbiota, known as dysbiosis, have been associated with atherosclerosis, hypertension, and heart failure. Nonetheless, the underlying mechanisms remain yet to be fully understood. Therefore, the microbiota and its metabolites have become a new therapeutic target for the prevention and treatment of CVD. In addition to a varied and balanced diet, the use of prebiotic and probiotic treatments or selective trimethylamine-N-oxide inhibitors could play a pivotal role in the prevention of CVD, especially in patients with a high metabolic risk.
Collapse
Affiliation(s)
- Gaia Anselmi
- From the UOC di Nutrizione Clinica, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucilla Gagliardi
- From the UOC di Nutrizione Clinica, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gabriele Egidi
- From the UOC di Nutrizione Clinica, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sabrina Leone
- From the UOC di Nutrizione Clinica, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- UOC di Medicina Interna e Gastroenterologia, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giacinto Abele Donato Miggiano
- From the UOC di Nutrizione Clinica, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Leonarda Galiuto
- From the UOC di Nutrizione Clinica, Area Medicina Interna, Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
93
|
Seegers JFML, Gül IS, Hofkens S, Brosel S, Schreib G, Brenke J, Donath C, de Vos WM. Toxicological safety evaluation of live Anaerobutyricum soehngenii strain CH106. J Appl Toxicol 2021; 42:244-257. [PMID: 34184753 PMCID: PMC9292162 DOI: 10.1002/jat.4207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
The gut commensal Anaerobutyricum soehngenii is an anaerobe that can produce both propionate and butyrate, metabolites that have been shown to have a positive effect on gut and overall health. Murine and human dose finding studies have shown that oral intake of A. soehngenii has a positive influence on peripheral insulin resistance, thereby reducing the risk of type 2 diabetes. A recent human intervention provided support for the mode of action of A. soehngenii as it affected gene expression in the duodenum, stimulated the secretion of GLP‐1 and improved insulin sensitivity. For these reasons A. soehngenii has been proposed as a food ingredient. Before introducing this bacterium to the food chain, however, it must be established that oral intake of live A. soehngenii bacteria does not pose any health risk. As part of the safety analysis of A. soehngenii strain CH106, we performed genotoxicity assays to determine its mutagenic potential (bacterial reverse mutation and in vitro mammalian cell micronucleus tests) and a 90‐day subchronic toxicity study in rats to determine overall toxicity potential. The results of both genotoxicity studies were negative, showing no genotoxic effects. For the 90‐day subchronic toxicity study, no adverse events were registered that could be attributed to the feeding with A. soehngenii strain CH106. Even at the highest dose, which exceeds the expected daily human intake more than 100‐fold, no adverse events were observed. These result support the conclusion that the use of A. soehngenii strain CH106 as a food ingredient is safe. Based on its health beneficial effects, the commensal bacterium Anaerobutyricum soehngenii strain CH106 is proposed as a food ingredient. As part of a safety assessment, toxic potential was tested through genotoxicity assays and a 90‐day subchronic toxicity study in rats. No genotoxic effects could be detected, while the rat study did not reveal any A. soehngenii‐related adverse events. The observations support the conclusion that the use of A. soehngenii strain CH106 as a food ingredient is safe.
Collapse
Affiliation(s)
| | | | | | - Sonja Brosel
- BSL BIOSERVICE Scientific Laboratories Münich, Planegg, Germany
| | - Gudrun Schreib
- Eurofins Biopharm Product Testing Münich, Planegg, Germany
| | - Jara Brenke
- Eurofins Biopharm Product Testing Münich, Planegg, Germany
| | - Claudia Donath
- Eurofins Biopharm Product Testing Münich, Planegg, Germany
| | - Willem M de Vos
- Caelus Pharmaceuticals BV, Zegveld, The Netherlands.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands.,Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
94
|
The Microbiota and the Gut-Brain Axis in Controlling Food Intake and Energy Homeostasis. Int J Mol Sci 2021; 22:ijms22115830. [PMID: 34072450 PMCID: PMC8198395 DOI: 10.3390/ijms22115830] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity currently represents a major societal and health challenge worldwide. Its prevalence has reached epidemic proportions and trends continue to rise, reflecting the need for more effective preventive measures. Hypothalamic circuits that control energy homeostasis in response to food intake are interesting targets for body-weight management, for example, through interventions that reinforce the gut-to-brain nutrient signalling, whose malfunction contributes to obesity. Gut microbiota-diet interactions might interfere in nutrient sensing and signalling from the gut to the brain, where the information is processed to control energy homeostasis. This gut microbiota-brain crosstalk is mediated by metabolites, mainly short chain fatty acids, secondary bile acids or amino acids-derived metabolites and subcellular bacterial components. These activate gut-endocrine and/or neural-mediated pathways or pass to systemic circulation and then reach the brain. Feeding time and dietary composition are the main drivers of the gut microbiota structure and function. Therefore, aberrant feeding patterns or unhealthy diets might alter gut microbiota-diet interactions and modify nutrient availability and/or microbial ligands transmitting information from the gut to the brain in response to food intake, thus impairing energy homeostasis. Herein, we update the scientific evidence supporting that gut microbiota is a source of novel dietary and non-dietary biological products that may beneficially regulate gut-to-brain communication and, thus, improve metabolic health. Additionally, we evaluate how the feeding time and dietary composition modulate the gut microbiota and, thereby, the intraluminal availability of these biological products with potential effects on energy homeostasis. The review also identifies knowledge gaps and the advances required to clinically apply microbiome-based strategies to improve the gut-brain axis function and, thus, combat obesity.
Collapse
|
95
|
Liu X, Cheng YW, Shao L, Sun SH, Wu J, Song QH, Zou HS, Ling ZX. Gut microbiota dysbiosis in Chinese children with type 1 diabetes mellitus: An observational study. World J Gastroenterol 2021; 27:2394-2414. [PMID: 34040330 PMCID: PMC8130045 DOI: 10.3748/wjg.v27.i19.2394] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/17/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gut microbiota dysbiosis is reportedly actively involved in autoimmune diseases such as type 1 diabetes mellitus (T1DM). However, the alterations in the gut microbiota and their correlation with fasting blood glucose (FBG) in Chinese children with T1DM remain unclear. AIM To investigate alterations in the gut microbiota in Chinese children with T1DM and their associations with clinical indicators. METHODS Samples from 51 children with T1DM and 47 age-matched and gender-matched healthy controls were obtained, to explore the structural and functional alterations in the fecal microbiota. The V3-V4 regions of the 16S rRNA gene were sequenced on a MiSeq instrument, and the association with FBG were analyzed. RESULTS We found that the bacterial diversity was significantly increased in the T1DM-associated fecal microbiota, and changes in the microbial composition were observed at different taxonomic levels. The T1DM-reduced differential taxa, such as Bacteroides vulgatus ATCC8482, Bacteroides ovatus, Bacteroides xylanisolvens, and Flavonifractor plautii, were negatively correlated with FBG, while the T1DM-enriched taxa, such as Blautia, Eubacterium hallii group, Anaerostipes hadrus, and Dorea longicatena, were positively correlated with FBG. Bacteroides vulgatus ATCC8482, Bacteroides ovatus, the Eubacterium hallii group, and Anaerostipes hadrus, either alone or in combination, could be used as noninvasive diagnostic biomarkers to discriminate children with T1DM from healthy controls. In addition, the functional changes in the T1DM-associated fecal microbiota also suggest that these fecal microbes were associated with altered functions and metabolic activities, such as glycan biosynthesis and metabolism and lipid metabolism, which might play vital roles in the pathogenesis and development of T1DM. CONCLUSION Our present comprehensive investigation of the T1DM-associated fecal microbiota provides novel insights into the pathogenesis of the disease and sheds light on the diagnosis and treatment of T1DM.
Collapse
Affiliation(s)
- Xia Liu
- Department of Intensive Care Unit, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Yi-Wen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Li Shao
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310000, Zhejiang Province, China
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310000, Zhejiang Province, China
| | - Shu-Hong Sun
- Department of Laboratory Medicine, Linyi People’s Hospital, Linyi 276000, Shandong Province, China
| | - Jian Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Qing-Hai Song
- Department of Geriatrics, Lishui Second People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Hong-Sheng Zou
- Department of Intensive Care Unit, People’s Hospital of Rongcheng, Rongcheng 264300, Shandong Province, China
| | - Zong-Xin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
96
|
Kong XJ, Liu J, Liu K, Koh M, Sherman H, Liu S, Tian R, Sukijthamapan P, Wang J, Fong M, Xu L, Clairmont C, Jeong MS, Li A, Lopes M, Hagan V, Dutton T, Chan ST(P, Lee H, Kendall A, Kwong K, Song Y. Probiotic and Oxytocin Combination Therapy in Patients with Autism Spectrum Disorder: A Randomized, Double-Blinded, Placebo-Controlled Pilot Trial. Nutrients 2021; 13:1552. [PMID: 34062986 PMCID: PMC8147925 DOI: 10.3390/nu13051552] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is a rapidly growing neurodevelopmental disorder. Both probiotics and oxytocin were reported to have therapeutic potential; however, the combination therapy has not yet been studied. We conducted a randomized, double-blinded, placebo-controlled, 2-stage pilot trial in 35 individuals with ASD aged 3-20 years (median = 10.30 years). Subjects were randomly assigned to receive daily Lactobacillus plantarum PS128 probiotic (6 × 1010 CFUs) or a placebo for 28 weeks; starting on week 16, both groups received oxytocin. The primary outcomes measure socio-behavioral severity using the Social Responsiveness Scale (SRS) and Aberrant Behavior Checklist (ABC). The secondary outcomes include measures of the Clinical Global Impression (CGI) scale, fecal microbiome, blood serum inflammatory markers, and oxytocin. All outcomes were compared between the two groups at baseline, 16 weeks, and 28 weeks into treatment. We observed improvements in ABC and SRS scores and significant improvements in CGI-improvement between those receiving probiotics and oxytocin combination therapy compared to those receiving placebo (p < 0.05). A significant number of favorable gut microbiome network hubs were also identified after combination therapy (p < 0.05). The favorable social cognition response of the combination regimen is highly correlated with the abundance of the Eubacterium hallii group. Our findings suggest synergic effects between probiotics PS128 and oxytocin in ASD patients, although further investigation is warranted.
Collapse
Affiliation(s)
- Xue-Jun Kong
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Jun Liu
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
- Harvard Medical School, Boston, MA 02115, USA; (P.S.); (L.X.); (H.L.)
| | - Kevin Liu
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Madelyn Koh
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Hannah Sherman
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Siyu Liu
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Ruiyi Tian
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | | | - Jiuju Wang
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Michelle Fong
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Lei Xu
- Harvard Medical School, Boston, MA 02115, USA; (P.S.); (L.X.); (H.L.)
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Cullen Clairmont
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Min-Seo Jeong
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Alice Li
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Maria Lopes
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Veronica Hagan
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Tess Dutton
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Suk-Tak (Phoebe) Chan
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Hang Lee
- Harvard Medical School, Boston, MA 02115, USA; (P.S.); (L.X.); (H.L.)
- MGH Biostatistics Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amy Kendall
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Kenneth Kwong
- Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; (J.L.); (K.L.); (M.K.); (H.S.); (S.L.); (R.T.); (J.W.); (M.F.); (C.C.); (M.-S.J.); (A.L.); (M.L.); (V.H.); (T.D.); (S.-T.C.); (A.K.); (K.K.)
| | - Yiqing Song
- Department of Epidemiology, Indiana University, Richard M. Fairbanks School of Public Health, Indianapolis, IN 46202, USA;
| |
Collapse
|
97
|
Singh TP, Natraj BH. Next-generation probiotics: a promising approach towards designing personalized medicine. Crit Rev Microbiol 2021; 47:479-498. [PMID: 33822669 DOI: 10.1080/1040841x.2021.1902940] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Second brain, forgotten organ, individual's identity card, and host's fingerprint are the few collective terms that are often used to describe the gut microbiome because of its variability, accountability, and its role in deciding the host's health. Also, the understanding of this host health-gut microbiota relationship can create an opportunity to control an individual's health by manipulating the gut microbiota composition. Several approaches like administration of probiotic, prebiotics, synbiotics, faecal microbiota transplantation have been tried to mitigate the dysbiosis originated ill effects. But the effects of these approaches are highly generic and non-specific. This creates the necessity to design personalized medicine that focuses on treatment of specific disease considering the individual specific gut microbiome. The health promoting commensals could be the new promising prophylactic and therapeutic agents for designing personalized medicine. These commensals are designated as next-generation probiotics (NGPs) and their unusual characteristics, unknown identity and special growth requirements have presented difficulties for researcher, industrial exploitation, and regulatory agencies. In this perspective, this review discusses the concept of NGPs, NGP candidates as tool for designing personalized medicine, designer probiotics as NGPs, required regulatory framework, and propose a road map to develop the NGP based product.
Collapse
Affiliation(s)
- Tejinder Pal Singh
- Dairy Microbiology Department, College of Dairy Science and Technology, Lala Lajpat Rai University of Veterinary and Animal Science, Hisar, India
| | | |
Collapse
|
98
|
Inhibitory Effects of Breast Milk-Derived Lactobacillus rhamnosus Probio-M9 on Colitis-Associated Carcinogenesis by Restoration of the Gut Microbiota in a Mouse Model. Nutrients 2021; 13:nu13041143. [PMID: 33808480 PMCID: PMC8065529 DOI: 10.3390/nu13041143] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 01/18/2023] Open
Abstract
Chronic inflammation is a risk factor for colorectal cancer, and inflammatory cytokines secreted from inflammatory cells and active oxygen facilitate tumorigenesis. Intestinal bacteria are thought to regulate tumorigenesis. The longer the breastfeeding period, the lower is the risk of inflammatory bowel disease. Here, we investigated preventive effects of the probiotic Lactobacillus rhamnosus M9 (Probio-M9) on colitis-associated tumorigenesis. An inflammatory colorectal tumor model was established using a 6-week-old male C57BL/6NCrSlc mouse, which was intraperitoneally administered with azoxymethane (AOM: 12 mg/kg body weight). On weeks 2 and 4, 2% dextran sulfate sodium (DSS) was administered to mice for 7 days through drinking water. On weeks 8 and 10, Probio-M9 (2 × 109/day) was orally administered for 7 days. Animals were sacrificed at 20 weeks after AOM administration and immunohistochemical staining and Western blotting was performed. The α-diversity of microflora (Shannon index), principal coordinate analysis, and distribution of intestinal bacterium genera and metabolic pathways were compared. The AOM/DSS group showed weight loss, diarrhea, intestinal shortening, increased number of colon tumors, proliferating tumorigenesis, increased inflammation score, fibrosis, increased CD68+, or CD163+ macrophage cells in the subserosal layer of non-tumor areas. Inflammation and tumorigenesis ameliorated after Probio-M9 treatment. Fecal microbial functions were altered by AOM/DSS treatment. Probio-M9 significantly upregulated the fecal microbial diversity and reversed fecal microbial functions. Thus, Probio-M9 could suppress tumor formation in the large intestine by regulating the intestinal environment and ameliorating inflammation, suggesting its therapeutic potential for treatment of inflammation and colitis-associated tumorigenesis.
Collapse
|
99
|
Huang Y, Wang Z, Ma H, Ji S, Chen Z, Cui Z, Chen J, Tang S. Dysbiosis and Implication of the Gut Microbiota in Diabetic Retinopathy. Front Cell Infect Microbiol 2021; 11:646348. [PMID: 33816351 PMCID: PMC8017229 DOI: 10.3389/fcimb.2021.646348] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
The pathogenesis of type 2 diabetes mellitus (T2DM) is commonly associated with altered gut bacteria. However, whether the microbial dysbiosis that exists in human diabetic patients with or without retinopathy is different remains largely unknown. Here, we collected clinical information and fecal samples from 75 participants, including 25 diabetic patients without retinopathy (DM), 25 diabetic patients with retinopathy (DR), and 25 healthy controls (HC). The gut microbial composition in the three groups was analyzed using 16S ribosomal RNA (rRNA) gene sequencing. Microbial structure and composition differed in the three groups. The α and β diversities in both the DM and DR groups were reduced compared with those in the HC group. Blautia was the most abundant genus, especially in the DM group. In addition, increased levels of Bifidobacterium and Lactobacillus and decreased levels of Escherichia-Shigella, Faecalibacterium, Eubacterium_hallii_group and Clostridium genera were observed in the DM and DR groups compared with the HC group. Furthermore, a biomarker set of 25 bacterial families, which could distinguish patients in the DR group from those in the DM and HC groups was identified, with the area under the curve values ranging from 0.69 to 0.85. Of note, Pasteurellaceae, which was increased in DM and decreased in DR compared with HC, generated a high AUC (0.74) as an individual predictive biomarker. Moreover, 14 family biomarkers were associated with fasting blood glucose levels or diabetes, with most of them being negatively correlated. In summary, our study establishes compositional alterations of gut microbiota in DM and DR, suggesting the potential use of gut microbiota as a non-invasive biomarker for clinical and differential diagnosis, as well as identifying potential therapeutic targets of diabetic retinopathy.
Collapse
Affiliation(s)
- Yinhua Huang
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China
| | - Zhijie Wang
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China
| | | | | | | | | | - Jiansu Chen
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
100
|
Relationship between Nutrient Intake and Human Gut Microbiota in Monozygotic Twins. ACTA ACUST UNITED AC 2021; 57:medicina57030275. [PMID: 33809761 PMCID: PMC8002349 DOI: 10.3390/medicina57030275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Background and Objectives: The gut microbiota is associated with human health and dietary nutrition. Various studies have been reported in this regard, but it is difficult to clearly analyze human gut microbiota as individual differences are significant. The causes of these individual differences in intestinal microflora are genetic and/or environmental. In this study, we focused on differences between identical twins in Japan to clarify the effects of nutrients consumed on the entire gut microbiome, while excluding genetic differences. Materials and Methods: We selected healthy Japanese monozygotic twins for the study and confirmed their zygosity by matching 15 short tandem repeat loci. Their fecal samples were subjected to 16S rRNA sequencing and bioinformatics analyses to identify and compare the fluctuations in intestinal bacteria. Results: We identified 12 genera sensitive to environmental factors, and found that Lactobacillus was relatively unaffected by environmental factors. Moreover, we identified protein, fat, and some nutrient intake that can affect 12 genera, which have been identified to be more sensitive to environmental factors. Among the 12 genera, Bacteroides had a positive correlation with retinol equivalent intake (rs = 0.38), Lachnospira had a significantly negative correlation with protein, sodium, iron, vitamin D, vitamin B6, and vitamin B12 intake (rs = −0.38, −0.41, −0.39, −0.63, −0.42, −0.49, respectively), Lachnospiraceae ND3007 group had a positive correlation with fat intake (rs = 0.39), and Lachnospiraceae UCG-008 group had a negative correlation with the saturated fatty acid intake (rs = −0.45). Conclusions: Our study is the first to focus on the relationship between human gut microbiota and nutrient intake using samples from Japanese twins to exclude the effects of genetic factors. These findings will broaden our understanding of the more intuitive relationship between nutrient intake and the gut microbiota and can be a useful basis for finding useful biomarkers that contribute to human health.
Collapse
|