1
|
Schoonakker MP, van Peet PG, van den Burg EL, Numans ME, Ducarmon QR, Pijl H, Wiese M. Impact of dietary carbohydrate, fat or protein restriction on the human gut microbiome: a systematic review. Nutr Res Rev 2025; 38:238-255. [PMID: 38602133 DOI: 10.1017/s0954422424000131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Restriction of dietary carbohydrates, fat and/or protein is often used to reduce body weight and/or treat (metabolic) diseases. Since diet is a key modulator of the human gut microbiome, which plays an important role in health and disease, this review aims to provide an overview of current knowledge of the effects of macronutrient-restricted diets on gut microbial composition and metabolites. A structured search strategy was performed in several databases. After screening for inclusion and exclusion criteria, thirty-six articles could be included. Data are included in the results only when supported by at least three independent studies to enhance the reliability of our conclusions. Low-carbohydrate (<30 energy%) diets tended to induce a decrease in the relative abundance of several health-promoting bacteria, including Bifidobacterium, as well as a reduction in short-chain fatty acid (SCFA) levels in faeces. In contrast, low-fat diets (<30 energy%) increased alpha diversity, faecal SCFA levels and abundance of some beneficial bacteria, including Faecalibacterium prausnitzii. There were insufficient data to draw conclusions concerning the effects of low-protein (<10 energy%) diets on gut microbiota. Although the data of included studies unveil possible benefits of low-fat and potential drawbacks of low-carbohydrate diets for human gut microbiota, the diversity in study designs made it difficult to draw firm conclusions. Using a more uniform methodology in design, sample processing and sharing raw sequence data could foster our understanding of the effects of macronutrient restriction on gut microbiota composition and metabolic dynamics relevant to health. This systematic review was registered at https://www.crd.york.ac.uk/prospero as CRD42020156929.
Collapse
Affiliation(s)
- Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Department of Internal Medicine, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Maria Wiese
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
2
|
Gonzalez-Gil AM, Barnouin Y, Celli A, Viola V, Villarreal MD, Nava MLD, Sciuk A, Qualls C, Armamento-Villareal R, Villareal DT. Metabolic Effects of Testosterone Added to Intensive Lifestyle Intervention in Older Men With Obesity and Hypogonadism. J Clin Endocrinol Metab 2025; 110:e814-e826. [PMID: 38606934 PMCID: PMC11470114 DOI: 10.1210/clinem/dgae249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/26/2024] [Accepted: 04/09/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Whether testosterone replacement therapy (TRT) conveys additional cardiometabolic benefit to an intensive lifestyle therapy (LT) in older men with obesity and hypogonadism remains unclear. OBJECTIVE To determine whether TRT augments the effect of LT on metabolic outcomes in older men with obesity and hypogonadism. DESIGN Secondary analysis of a randomized, double-blind, placebo-controlled trial. SETTING Veterans Affairs Medical Center. PARTICIPANTS Eighty-three older (age ≥ 65 years) men with obesity (body mass index ≥ 30 kg/m2) and persistently low Am testosterone (< 10.4 nmol/L) associated with frailty. INTERVENTIONS LT (weight management and exercise training) plus either testosterone (LT + TRT) or placebo (LT + Pbo) for 6 months. OUTCOME MEASURES The primary outcome was change in glycated hemoglobin (HbA1c). Secondary outcomes included changes in other glucometabolic and lipid profile components, liver enzymes, inflammatory markers, and adipokines; subcutaneous, visceral, intramuscular, and hepatic fat; blood pressure; and metabolic syndrome score. RESULTS HbA1c decreased similarly in LT + TRT and LT + Pbo groups (-0.5 ± 0.1 vs -0.6 ± 0.1%, respectively; P = 0.35). While TRT showed no synergistic effect with LT on ameliorating secondary outcomes, it eliminated the augmentative effect of LT on high-density lipoprotein cholesterol concentration (5.4 ± 1.0 mg/dL in the LT + Pbo group vs 0.2 ± 1.1 mg/dL in the LT + TRT group, P = .01) and adiponectin levels (-408 ± 489 ng/mL in LT + TRT group vs 1832 ± 468 ng/mL in LT + Pbo group, P = .02). CONCLUSION In older men with obesity and hypogonadism, adding TRT for 6 months to LT does not result in further improved cardiometabolic profiles and could potentially blunt some of the metabolic benefits induced by LT.
Collapse
Affiliation(s)
- Adrian M. Gonzalez-Gil
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Yoann Barnouin
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Alessandra Celli
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Viola Viola
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Marcos D. Villarreal
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Maria Liza Duremdes Nava
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Adam Sciuk
- Section of Radiology, Michael E DeBakey VA Medical Center, Houston TX, USA
| | - Clifford Qualls
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, USA
| | - Reina Armamento-Villareal
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| | - Dennis T. Villareal
- Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston TX, USA
- Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine. Houston, TX, USA
| |
Collapse
|
3
|
Caprara G, Pallavi R, Sanyal S, Pelicci PG. Dietary Restrictions and Cancer Prevention: State of the Art. Nutrients 2025; 17:503. [PMID: 39940361 PMCID: PMC11820753 DOI: 10.3390/nu17030503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Worldwide, almost 10 million cancer deaths occurred in 2022, a number that is expected to rise to 16.3 million by 2040. Primary prevention has long been acknowledged as a crucial approach to reducing cancer incidence. In fact, between 30 and 50 percent of all tumors are known to be preventable by eating a healthy diet, staying active, avoiding alcohol, smoking, and being overweight. Accordingly, many international organizations have created tumor prevention guidelines, which underlie the importance of following a diet that emphasizes eating plant-based foods while minimizing the consumption of red/processed meat, sugars, processed foods, and alcohol. However, further research is needed to define the relationship between the effect of specific diets or nutritional components on cancer prevention. Interestingly, reductions in food intake and dietetic restrictions can extend the lifespan of yeast, nematodes, flies, and rodents. Despite controversial results in humans, those approaches have the potential to ameliorate health via direct and indirect effects on specific signaling pathways involved in cancer onset. Here, we describe the latest knowledge on the cancer-preventive potential of dietary restrictions and the biochemical processes involved. Molecular, preclinical, and clinical studies evaluating the effects of different fasting strategies will also be reviewed.
Collapse
Affiliation(s)
- Greta Caprara
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Shalini Sanyal
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| |
Collapse
|
4
|
Su W, Chen H, Hu D, Ye B, Zhang W, Zhang G, Si X, Zhou X. The Causal Role of Esophageal Cancer and Gut Microbiota: A Bidirectional Mendelian Randomization Study. J Evid Based Integr Med 2025; 30:2515690X251324793. [PMID: 40012260 PMCID: PMC11866380 DOI: 10.1177/2515690x251324793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 10/10/2024] [Accepted: 02/15/2025] [Indexed: 02/28/2025] Open
Abstract
AIMS Gut microbiota are reported to be associated with the incidence and prognosis of Esophageal cancer (EC) but their genetic association is unclear. We carried out a bidirectional MR analysis to assess the causal relationship between EC and gut microbiota from fecal samples. METHODS The microbiome genome-wide association studies (GWAS) data of 18,340 individuals provided by MiBioGen consortium and the EC GWAS data (740 esophageal cancers cases and 372 016 controls) provided by UK Biobank were respectively utilized as exposure and/or outcome data. Reliable single nucleotide polymorphisms (SNPs) were obtained after rigorous screening. A bidirectional Mendelian randomization (MR) analysis was conducted using the inverse-variance weighted (IVW) method. The sensitivity analyses including the MR-Egger method, weighted median, weighed mode and leave-one-out method were performed to examine the stability, heterogeneity and pleiotropy of the results. RESULTS Forward MR analysis revealed the increase in abundance of the microbial trait by each standard deviation was associated with a higher risk of EC (Coprobacter (OR = 1.001,95%CI = 1.000-1.002, P = .0281, FDR = 0.0424); Ruminococcus1(OR = 1.001,95%CI = 1.000-1.002, P = .0318, FDR = 0.0424); Senegalimassilia (OR = 1.002,95%CI = 1.000-1.003, P = .0062, FDR = 0.0372); Veillonella (OR = 1.001,95%CI = 1.000-1.002, P = .0182, FDR = 0.0372)) or a lower risk of EC (Eubacterium oxidoreducens (OR = 0.999, 95%CI = 0.998-1.000, P = .0379, FDR = 00 433); Lachnospira (OR = 0.998,95%CI = 0.996-1.000, P = .0186, FDR = 0.0372); Romboutsia (OR = 0.999,95%CI = 0.998-1.000, P = .0482, FDR = 0.0482); Turicibacter (OR = 0.999,95%CI = 0.998-1.000, P = .0133, FDR = 0.0372)). Reverse MR analysis showed that genetic liability to EC was also causally linked toincreased susceptibility of changes in the gut microbiome (genera Eggerthella (Beta = 37.63,95%CI = 4.76-70.50, P = .0248, FDR = 0.0331); Coprococcus 2 (Beta = 23.90,95%CI = 1.65-46.15, P = .0353, FDR = 0.0353); Christensenellaceae R.7 (Beta = 22.75,95%CI = 4.22-41.28, P = .0161, FDR = 0.0322); Intestinimonas (Beta = -33.24,95%CI = -54.90-11.58, P = .0026, FDR = 0.0104)). CONCLUSIONS Our findings supported a bidirectionally causal relationship between gut microbiota and EC, implying the potential role of gut microbiota in preventing the occurrence and development of EC.
Collapse
Affiliation(s)
- Wei Su
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Han Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Die Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bixing Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weifeng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinmin Si
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoying Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Hamamah S, Iatcu OC, Covasa M. Dietary Influences on Gut Microbiota and Their Role in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Nutrients 2024; 17:143. [PMID: 39796579 PMCID: PMC11722922 DOI: 10.3390/nu17010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major contributor to liver-related morbidity, cardiovascular disease, and metabolic complications. Lifestyle interventions, including diet and exercise, are first line in treating MASLD. Dietary approaches such as the low-glycemic-index Mediterranean diet, the ketogenic diet, intermittent fasting, and high fiber diets have demonstrated potential in addressing the metabolic dysfunction underlying this condition. The development and progression of MASLD are closely associated with taxonomic shifts in gut microbial communities, a relationship well-documented in the literature. Given the importance of diet as a primary treatment for MASLD, it is important to understand how gut microbiota and their metabolic byproducts mediate favorable outcomes induced by healthy dietary patterns. Conversely, microbiota changes conferred by unhealthy dietary patterns such as the Western diet may induce dysbiosis and influence steatotic liver disease through promoting hepatic inflammation, up-regulating lipogenesis, dysregulating bile acid metabolism, increasing insulin resistance, and causing oxidative damage in hepatocytes. Although emerging evidence has identified links between diet, microbiota, and development of MASLD, significant gaps remain in understanding specific microbial roles, metabolite pathways, host interactions, and causal relationships. Therefore, this review aims to provide mechanistic insights into the role of microbiota-mediated processes through the analysis of both healthy and unhealthy dietary patterns and their contribution to MASLD pathophysiology. By better elucidating the interplay between dietary nutrients, microbiota-mediated processes, and the onset and progression of steatotic liver disease, this work aims to identify new opportunities for targeted dietary interventions to treat MASLD efficiently.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA;
| | - Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| |
Collapse
|
6
|
Luo J, Liang S, Jin F. Gut microbiota and healthy longevity. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2590-2602. [PMID: 39110402 DOI: 10.1007/s11427-023-2595-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/01/2024] [Indexed: 12/18/2024]
Abstract
Recent progress on the underlying biological mechanisms of healthy longevity has propelled the field from elucidating genetic modification of healthy longevity hallmarks to defining mechanisms of gut microbiota influencing it. Importantly, the role of gut microbiota in the healthy longevity of the host may provide unprecedented opportunities to decipher the plasticity of lifespan on a natural evolutionary scale and shed light on using microbiota-targeted strategies to promote healthy aging and combat age-related diseases. This review investigates how gut microbiota affects healthy longevity, focusing on the mechanisms through which gut microbiota modulates it. Specifically, we focused on the ability of gut microbiota to enhance the intestinal barrier integrity, provide protection from inflammaging, ameliorate nutrientsensing pathways, optimize mitochondrial function, and improve defense against age-related diseases, thus participating in enhancing longevity and healthspan.
Collapse
Affiliation(s)
- Jia Luo
- College of Psychology, Sichuan Normal University, Chengdu, 610066, China
| | - Shan Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
7
|
Chanda W, Jiang H, Liu SJ. The Ambiguous Correlation of Blautia with Obesity: A Systematic Review. Microorganisms 2024; 12:1768. [PMID: 39338443 PMCID: PMC11433710 DOI: 10.3390/microorganisms12091768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 09/30/2024] Open
Abstract
Obesity is a complex and multifactorial disease with global epidemic proportions, posing significant health and economic challenges. Whilst diet and lifestyle are well-established contributors to the pathogenesis, the gut microbiota's role in obesity development is increasingly recognized. Blautia, as one of the major intestinal bacteria of the Firmicutes phylum, is reported with both potential probiotic properties and causal factors for obesity in different studies, making its role controversial. To summarize the current understanding of the Blautia-obesity correlation and to evaluate the evidence from animal and clinical studies, we used "Blautia" AND "obesity" as keywords searching through PubMed and SpringerLink databases for research articles. After removing duplicates and inadequate articles using the exclusion criteria, we observed different results between studies supporting and opposing the beneficial role of Blautia in obesity at the genus level. Additionally, several studies showed probiotic effectiveness at the species level for Blautia coccoides, B. wexlerae, B. hansenii, B. producta, and B. luti. Therefore, the current evidence does not demonstrate Blautia's direct involvement as a pathogenic microbe in obesity development or progression, which informs future research and therapeutic strategies targeting the gut Blautia in obesity management.
Collapse
Affiliation(s)
- Warren Chanda
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- Pathology and Microbiology Department, School of Medicine and Health Sciences, Mulungushi University, Livingstone P.O. Box 60009, Zambia
| | - He Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- State Key Laboratory of Microbial Resources, and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Fazzone B, Anderson EM, Rozowsky JM, Yu X, O’Malley KA, Robinson S, Scali ST, Cai G, Berceli SA. Short-Term Dietary Restriction Potentiates an Anti-Inflammatory Circulating Mucosal-Associated Invariant T-Cell Response. Nutrients 2024; 16:1245. [PMID: 38674935 PMCID: PMC11053749 DOI: 10.3390/nu16081245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Short-term protein-calorie dietary restriction (StDR) is a promising preoperative strategy for modulating postoperative inflammation. We have previously shown marked gut microbial activity during StDR, but relationships between StDR, the gut microbiome, and systemic immunity remain poorly understood. Mucosal-associated invariant T-cells (MAITs) are enriched on mucosal surfaces and in circulation, bridge innate and adaptive immunity, are sensitive to gut microbial changes, and may mediate systemic responses to StDR. Herein, we characterized the MAIT transcriptomic response to StDR using single-cell RNA sequencing of human PBMCs and evaluated gut microbial species-level changes through sequencing of stool samples. Healthy volunteers underwent 4 days of DR during which blood and stool samples were collected before, during, and after DR. MAITs composed 2.4% of PBMCs. More MAIT genes were differentially downregulated during DR, particularly genes associated with MAIT activation (CD69), regulation of pro-inflammatory signaling (IL1, IL6, IL10, TNFα), and T-cell co-stimulation (CD40/CD40L, CD28), whereas genes associated with anti-inflammatory IL10 signaling were upregulated. Stool analysis showed a decreased abundance of multiple MAIT-stimulating Bacteroides species during DR. The analyses suggest that StDR potentiates an anti-inflammatory MAIT immunophenotype through modulation of TCR-dependent signaling, potentially secondary to gut microbial species-level changes.
Collapse
Affiliation(s)
- Brian Fazzone
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
| | - Erik M. Anderson
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
| | - Jared M. Rozowsky
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
| | - Xuanxuan Yu
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA;
| | - Kerri A. O’Malley
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL 32608, USA
| | - Scott Robinson
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL 32608, USA
| | - Salvatore T. Scali
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL 32608, USA
| | - Guoshuai Cai
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA;
| | - Scott A. Berceli
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL 32611, USA; (B.F.); (E.M.A.); (K.A.O.); (S.R.); (S.T.S.)
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL 32608, USA
| |
Collapse
|
9
|
Mei X, Li Y, Zhang X, Zhai X, Yang Y, Li Z, Li L. Maternal Phlorizin Intake Protects Offspring from Maternal Obesity-Induced Metabolic Disorders in Mice via Targeting Gut Microbiota to Activate the SCFA-GPR43 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4703-4725. [PMID: 38349207 DOI: 10.1021/acs.jafc.3c06370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Maternal obesity increases the risk of obesity and metabolic disorders (MDs) in offspring, which can be mediated by the gut microbiota. Phlorizin (PHZ) can improve gut dysbiosis and positively affect host health; however, its transgenerational metabolic benefits remain largely unclear. This study aimed to investigate the potential of maternal PHZ intake in attenuating the adverse impacts of a maternal high-fat diet on obesity-related MDs in dams and offspring. The results showed that maternal PHZ reduced HFD-induced body weight gain and fat accumulation and improved glucose intolerance and abnormal lipid profiles in both dams and offspring. PHZ improved gut dysbiosis by promoting expansion of SCFA-producing bacteria, Akkermansia and Blautia, while inhibiting LPS-producing and pro-inflammatory bacteria, resulting in significantly increased fecal SCFAs, especially butyric acid, and reduced serum lipopolysaccharide levels and intestinal inflammation. PHZ also promoted intestinal GLP-1/2 secretion and intestinal development and enhanced gut barrier function by activating G protein-coupled receptor 43 (GPR43) in the offspring. Antibiotic-treated mice receiving FMT from PHZ-regulated offspring could attenuate MDs induced by receiving FMT from HFD offspring through the gut microbiota to activate the GPR43 pathway. It can be regarded as a promising functional food ingredient for preventing intergenerational transmission of MDs and breaking the obesity cycle.
Collapse
Affiliation(s)
- Xueran Mei
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Yi Li
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney 2052, Australia
| | - Xiaoyu Zhang
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Xiwen Zhai
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney 2052, Australia
| | - Yi Yang
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
| | - Zhengjuan Li
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Liping Li
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| |
Collapse
|
10
|
Stols-Gonçalves D, Mak AL, Madsen MS, van der Vossen EWJ, Bruinstroop E, Henneman P, Mol F, Scheithauer TPM, Smits L, Witjes J, Meijnikman AS, Verheij J, Nieuwdorp M, Holleboom AG, Levin E. Faecal Microbiota transplantation affects liver DNA methylation in Non-alcoholic fatty liver disease: a multi-omics approach. Gut Microbes 2023; 15:2223330. [PMID: 37317027 DOI: 10.1080/19490976.2023.2223330] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023] Open
Abstract
Individuals with nonalcoholic fatty liver disease (NAFLD) have an altered gut microbiota composition. Moreover, hepatic DNA methylation may be altered in the state of NAFLD. Using a fecal microbiota transplantation (FMT) intervention, we aimed to investigate whether a change in gut microbiota composition relates to altered liver DNA methylation in NAFLD. Moreover, we assessed whether plasma metabolite profiles altered by FMT relate to changes in liver DNA methylation. Twenty-one individuals with NAFLD underwent three 8-weekly vegan allogenic donor (n = 10) or autologous (n = 11) FMTs. We obtained hepatic DNA methylation profiles from paired liver biopsies of study participants before and after FMTs. We applied a multi-omics machine learning approach to identify changes in the gut microbiome, peripheral blood metabolome and liver DNA methylome, and analyzed cross-omics correlations. Vegan allogenic donor FMT compared to autologous FMT induced distinct differential changes in I) gut microbiota profiles, including increased abundance of Eubacterium siraeum and potential probiotic Blautia wexlerae; II) plasma metabolites, including altered levels of phenylacetylcarnitine (PAC) and phenylacetylglutamine (PAG) both from gut-derived phenylacetic acid, and of several choline-derived long-chain acylcholines; and III) hepatic DNA methylation profiles, most importantly in Threonyl-TRNA Synthetase 1 (TARS) and Zinc finger protein 57 (ZFP57). Multi-omics analysis showed that Gemmiger formicillis and Firmicutes bacterium_CAG_170 positively correlated with both PAC and PAG. E siraeum negatively correlated with DNA methylation of cg16885113 in ZFP57. Alterations in gut microbiota composition by FMT caused widespread changes in plasma metabolites (e.g. PAC, PAG, and choline-derived metabolites) and liver DNA methylation profiles in individuals with NAFLD. These results indicate that FMTs might induce metaorganismal pathway changes, from the gut bacteria to the liver.
Collapse
Affiliation(s)
- Daniela Stols-Gonçalves
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mette S Madsen
- Gubra, Hørsholm, Denmark
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Eveline Bruinstroop
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Peter Henneman
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Femke Mol
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Torsten P M Scheithauer
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Internal Medicine, Amsterdam University Medical Centre (UMC), Vrije Universiteit (VU) University Medical Centre, Amsterdam, Netherlands
| | - Loek Smits
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Julia Witjes
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Abraham Stijn Meijnikman
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joanne Verheij
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan G Holleboom
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Evgeni Levin
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Horaizon BV, Delft, The Netherlands
| |
Collapse
|
11
|
Zheng R, Xiang X, Shi Y, Qiu A, Luo X, Xie J, Russell R, Zhang D. Chronic jet lag alters gut microbiome and mycobiome and promotes the progression of MAFLD in HFHFD-fed mice. Front Microbiol 2023; 14:1295869. [PMID: 38130943 PMCID: PMC10733492 DOI: 10.3389/fmicb.2023.1295869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common chronic liver disease worldwide. Circadian disruptors, such as chronic jet lag (CJ), may be new risk factors for MAFLD development. However, the roles of CJ on MAFLD are insufficiently understood, with mechanisms remaining elusive. Studies suggest a link between gut microbiome dysbiosis and MAFLD, but most of the studies are mainly focused on gut bacteria, ignoring other components of gut microbes, such as gut fungi (mycobiome), and few studies have addressed the rhythm of the gut fungi. This study explored the effects of CJ on MAFLD and its related microbiotic and mycobiotic mechanisms in mice fed a high fat and high fructose diet (HFHFD). Forty-eight C57BL6J male mice were divided into four groups: mice on a normal diet exposed to a normal circadian cycle (ND-NC), mice on a normal diet subjected to CJ (ND-CJ), mice on a HFHFD exposed to a normal circadian cycle (HFHFD-NC), and mice on a HFHFD subjected to CJ (HFHFD-CJ). After 16 weeks, the composition and rhythm of microbiota and mycobiome in colon contents were compared among groups. The results showed that CJ exacerbated hepatic steatohepatitis in the HFHFD-fed mice. Compared with HFHFD-NC mice, HFHFD-CJ mice had increases in Aspergillus, Blumeria and lower abundances of Akkermansia, Lactococcus, Prevotella, Clostridium, Bifidobacterium, Wickerhamomyces, and Saccharomycopsis genera. The fungi-bacterial interaction network became more complex after HFHFD and/or CJ interventions. The study revealed that CJ altered the composition and structure of the gut bacteria and fungi, disrupted the rhythmic oscillation of the gut microbiota and mycobiome, affected interactions among the gut microbiome, and promoted the progression of MAFLD in HFHFD mice.
Collapse
Affiliation(s)
- Ruoyi Zheng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, China
| | - Xingwei Xiang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Anqi Qiu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Luo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junyan Xie
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ryan Russell
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Gabbia D, De Martin S. Targeting the Adipose Tissue-Liver-Gut Microbiota Crosstalk to Cure MASLD. BIOLOGY 2023; 12:1471. [PMID: 38132297 PMCID: PMC10741127 DOI: 10.3390/biology12121471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
The gut microbiota is a complex system, playing a peculiar role in regulating innate and systemic immunity. Increasing evidence links dysfunctional gut microbiota to metabolic dysfunction-associated steatotic liver disease (MASLD) due to the activation of multiple pathways in the gut and in the liver, including those mediated by Toll-like receptors (TLRs), that sustain hepatic inflammation. Thus, many efforts have been made to unravel the role of microbiota-associated dysfunction in MASLD, with the final aim of finding novel strategies to improve liver steatosis and function. Moreover, recent evidence underlines the role of adipose tissue in sustaining hepatic inflammation during MASLD development. In this review, we focus on the recently discovered strategies proposed to improve the alteration of gut microbiota observed in MASLD patients, with a particular insight into those known to modulate gut microbiota-associated dysfunction and to affect the complex crosstalk between the gut, the adipose tissue, and the liver.
Collapse
Affiliation(s)
- Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 351131 Padova, Italy;
| | | |
Collapse
|
13
|
Aparicio A, Sun Z, Gold DR, Litonjua AA, Weiss ST, Lee-Sarwar K, Liu YY. Genotype-microbiome-metabolome associations in early childhood, and their link to BMI and childhood obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.13.23298467. [PMID: 38014043 PMCID: PMC10680902 DOI: 10.1101/2023.11.13.23298467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The influence of genotype on defining the human gut microbiome has been extensively studied, but definite conclusions have not yet been found. To fill this knowledge gap, we leverage data from children enrolled in the Vitamin D Antenatal Asthma Reduction Trial (VDAART) from 6 months to 8 years old. We focus on a pool of 12 genes previously found to be associated with the gut microbiome in independent studies, establishing a Bonferroni corrected significance level of p-value < 2.29 × 10 -6 . We identified significant associations between SNPs in the FHIT gene (known to be associated with obesity and type 2 diabetes) and obesity-related microbiome features, and the children's BMI through their childhood. Based on these associations, we defined a set of SNPs of interest and a set of taxa of interest. Taking a multi-omics approach, we integrated plasma metabolome data into our analysis and found simultaneous associations among children's BMI, the SNPs of interest, and the taxa of interest, involving amino acids, lipids, nucleotides, and xenobiotics. Using our association results, we constructed a quadripartite graph where each disjoint node set represents SNPs in the FHIT gene, microbial taxa, plasma metabolites, or BMI measurements. Network analysis led to the discovery of patterns that identify several genetic variants, microbial taxa and metabolites as new potential markers for obesity, type 2 diabetes, or insulin resistance risk.
Collapse
|
14
|
Chaiyasut C, Sivamaruthi BS, Lailerd N, Sirilun S, Thangaleela S, Khongtan S, Bharathi M, Kesika P, Saelee M, Choeisoongnern T, Fukngoen P, Peerajan S, Sittiprapaporn P. Influence of Bifidobacterium breve on the Glycaemic Control, Lipid Profile and Microbiome of Type 2 Diabetic Subjects: A Preliminary Randomized Clinical Trial. Pharmaceuticals (Basel) 2023; 16:ph16050695. [PMID: 37242478 DOI: 10.3390/ph16050695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most highly prevalent metabolic disorders worldwide. Uncontrolled T2DM can lead to other health threats such as cardiac arrest, lower-limb amputation, blindness, stroke, impaired kidney function, and microvascular and macrovascular complications. Many studies have demonstrated the association between gut microbiota and diabetes development and probiotic supplementation in improving glycemic properties in T2DM. The study aimed to evaluate the influence of Bifidobacterium breve supplementation on glycemic control, lipid profile, and microbiome of T2DM subjects. Forty participants were randomly divided into two groups, and they received probiotics (50 × 109 CFU/day) or placebo interventions (corn starch; 10 mg/day) for 12 weeks. The changes in the blood-urea nitrogen (BUN), aspartate aminotransferase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP), fasting blood sugar (FBS), glycated hemoglobin (HbA1c), total cholesterol (TC), triglycerides (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), creatinine levels, and other factors such as body-mass index, visceral fat, body fat, and body weight were assessed at baseline and after 12 weeks. B. breve supplementation significantly reduced BUN, creatinine, LDL, TG, and HbA1c levels compared to the placebo group. Significant changes were observed in the microbiome of the probiotic-treated group compared to the placebo group. Firmicutes and proteobacteria were predominant in the placebo and probiotic-treated groups. Genera Streptococcus, Butyricicoccus, and species Eubacterium hallii were significantly reduced in the probiotic-treated group compared to the placebo. Overall results suggested that B. breve supplementation could prevent worsening of representative clinical parameters in T2DM subjects. The current study has limitations, including fewer subjects, a single probiotic strain, and fewer metagenomic samples for microbiome analysis. Therefore, the results of the current study require further validation using more experimental subjects.
Collapse
Affiliation(s)
- Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Bhagavathi Sundaram Sivamaruthi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Narissara Lailerd
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasithorn Sirilun
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Subramanian Thangaleela
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Suchanat Khongtan
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Muruganantham Bharathi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Periyanaina Kesika
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Manee Saelee
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| | - Thiwanya Choeisoongnern
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| | - Pranom Fukngoen
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Phakkharawat Sittiprapaporn
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| |
Collapse
|
15
|
Neto J, Jantsch J, Rodrigues F, Squizani S, Eller S, Oliveira TF, Silveira AK, Moreira JCF, Giovenardi M, Porawski M, Guedes RP. Impact of cafeteria diet and n3 supplementation on the intestinal microbiota, fatty acids levels, neuroinflammatory markers and social memory in male rats. Physiol Behav 2023; 260:114068. [PMID: 36567032 DOI: 10.1016/j.physbeh.2022.114068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/08/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To assess the effects of omega-3 (n3) supplementation on intestinal microbiota, fatty acids profile, neuroinflammation, and social memory of cafeteria diet (CAF)-fed rats. METHODS Male Wistar rats were fed with CAF for 20 weeks. Omega-3 (500 mg/kg/day) was supplemented between the 16th and 20th week. Colon morphology, intestinal microbiota composition, short-chain fatty acids (SCFA) and lipopolysaccharide (LPS) in the plasma, fatty acids profile, TLR-4 and claudin-5 expressions in the brain, and social memory were investigated. RESULTS CAF reduced colon length, crypts' depth, and microbiota diversity, while n3 increased the Firmicutes/Bacteroidetes ratio. CAF increased SCFA plasma levels, but n3 reduced butyrate and isobutyrate in obese rats. LPS was increased in CAF-fed rats, and n3 decreased its levels. In the cerebral cortex, n3 increased caprylic, palmitic, stearic, tricosanoic, lignoceric, myristoleic, and linoleic acids. CAF increased palmitic acid and TLR-4 expression in the cerebral cortex while decreasing claudin-5 in the hippocampus. In the social memory test, CAF-fed animals showed greater social interaction with no effect of n3. CONCLUSIONS The lack of n3 effect in some of the evaluated parameters may be due to the severity of the obesity caused by CAF. However, n3 reduced LPS levels, suggesting its ability to reverse endotoxemia.
Collapse
Affiliation(s)
- João Neto
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Fernanda Rodrigues
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Samia Squizani
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Sarah Eller
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Tiago Franco Oliveira
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | | | - José Cláudio Fonseca Moreira
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Departamento de Bioquímica da Universidade Federal do Rio Grande do Sul (UFRGS), Brazil
| | - Marcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Marilene Porawski
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Programa de Pós-Graduação em Medicina: Hepatologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil.
| |
Collapse
|
16
|
Impact of caloric restriction on the gut microbiota. Curr Opin Microbiol 2023; 73:102287. [PMID: 36868081 DOI: 10.1016/j.mib.2023.102287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 03/05/2023]
Abstract
Caloric restriction (CR) and related time-restricted diets have been popularized as means of preventing metabolic disease while improving general well-being. However, evidence as to their long-term efficacy, adverse effects, and mechanisms of activity remains incompletely understood. The gut microbiota is modulated by such dietary approaches, yet causal evidence to its possible downstream impacts on host metabolism remains elusive. Herein, we discuss the positive and adverse influences of restrictive dietary interventions on gut microbiota composition and function, and their collective impacts on host health and disease risk. We highlight known mechanisms of microbiota influences on the host, such as modulation of bioactive metabolites, while discussing challenges in achieving mechanistic dietary-microbiota insights, including interindividual variability in dietary responses as well as other methodological and conceptual challenges. In all, causally understanding the impact of CR approaches on the gut microbiota may enable to better decode their overall influences on human physiology and disease.
Collapse
|
17
|
Kanezawa S, Moriyama M, Kanda T, Fukushima A, Masuzaki R, Sasaki-Tanaka R, Tsunemi A, Ueno T, Fukuda N, Kogure H. Gut-Microbiota Dysbiosis in Stroke-Prone Spontaneously Hypertensive Rats with Diet-Induced Steatohepatitis. Int J Mol Sci 2023; 24:ijms24054603. [PMID: 36902037 PMCID: PMC10002594 DOI: 10.3390/ijms24054603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Metabolic-dysfunction-associated fatty-liver disease (MAFLD) is the principal worldwide cause of liver disease. Individuals with nonalcoholic steatohepatitis (NASH) have a higher prevalence of small-intestinal bacterial overgrowth (SIBO). We examined gut-microbiota isolated from 12-week-old stroke-prone spontaneously hypertensive-5 rats (SHRSP5) fed on a normal diet (ND) or a high-fat- and high-cholesterol-containing diet (HFCD) and clarified the differences between their gut-microbiota. We observed that the Firmicute/Bacteroidetes (F/B) ratio in both the small intestines and the feces of the SHRSP5 rats fed HFCD increased compared to that of the SHRSP5 rats fed ND. Notably, the quantities of the 16S rRNA genes in small intestines of the SHRSP5 rats fed HFCD were significantly lower than those of the SHRSP5 rats fed ND. As in SIBO syndrome, the SHRSP5 rats fed HFCD presented with diarrhea and body-weight loss with abnormal types of bacteria in the small intestine, although the number of bacteria in the small intestine did not increase. The microbiota of the feces in the SHRSP5 rats fed HFCD was different from those in the SHRP5 rats fed ND. In conclusion, there is an association between MAFLD and gut-microbiota alteration. Gut-microbiota alteration may be a therapeutic target for MAFLD.
Collapse
Affiliation(s)
- Shini Kanezawa
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Mitsuhiko Moriyama
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
- Correspondence: (M.M.); (T.K.); Tel.: +81-3-3972-8111 (M.M. & T.K.)
| | - Tatsuo Kanda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
- Correspondence: (M.M.); (T.K.); Tel.: +81-3-3972-8111 (M.M. & T.K.)
| | - Akiko Fukushima
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Ryota Masuzaki
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Reina Sasaki-Tanaka
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Akiko Tsunemi
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Takahiro Ueno
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Noboru Fukuda
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Hirofumi Kogure
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| |
Collapse
|
18
|
Deledda A, Palmas V, Heidrich V, Fosci M, Lombardo M, Cambarau G, Lai A, Melis M, Loi E, Loviselli A, Manzin A, Velluzzi F. Dynamics of Gut Microbiota and Clinical Variables after Ketogenic and Mediterranean Diets in Drug-Naïve Patients with Type 2 Diabetes Mellitus and Obesity. Metabolites 2022; 12:1092. [PMID: 36355175 PMCID: PMC9693465 DOI: 10.3390/metabo12111092] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 07/30/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), the most common form of diabetes, is a progressive chronic metabolic disease that has increasingly spread worldwide, enhancing the mortality rate, particularly from cardiovascular diseases (CVD). Lifestyle improvement through diet and physical activity is, together with drug treatment, the cornerstone of T2DM management. The Mediterranean diet (MD), which favors a prevalence of unprocessed vegetable foods and a reduction in red meats and industrial foods, without excluding any food category, is usually recommended. Recently, scientific societies have promoted a very low-calorie ketogenic diet (VLCKD), a multiphasic protocol that limits carbohydrates and then gradually re-introduces them, with a favorable outcome on body weight and metabolic parameters. Indeed, gut microbiota (GM) modifications have been linked to overweight/obesity and metabolic alterations typical of T2DM. Diet is known to affect GM largely, but only a few studies have investigated the effects of VLCKD on GM, especially in T2DM. In this study, we have compared anthropometric, biochemical, lifestyle parameters, the quality of life, and the GM of eleven patients with recently diagnosed T2DM and overweight or obesity, randomly assigned to two groups of six and five patients who followed the VLCKD (KETO) or hypocaloric MD (MEDI) respectively; parameters were recorded at baseline (T0) and after two (T2) and three months (T3). The results showed that VLCKD had more significant beneficial effects than MD on anthropometric parameters, while biochemical improvements did not statistically differ. As for the GM, despite the lack of significant results regarding the alpha and beta diversity, and the Firmicutes/Bacteroidota ratio between the two groups, in the KETO group, a significant increase in beneficial microbial taxa such as Verrucomicrobiota phylum with its members Verrucomicrobiae, Verrucomicrobiales, Akkermansiaceae, and Akkermansia, Christensenellaceae family, Eubacterium spp., and a reduction in microbial taxa previously associated with obesity (Firmicutes and Actinobacteriota) or other diseases (Alistipes) was observed both at T2 and T3. With regards to the MEDI group, variations were limited to a significant increase in Actinobacteroidota phylum at T2 and T3 and Firmicutes phylum at T3. Moreover, a metagenomic alteration linked to some metabolic pathways was found exclusively in the KETO group. In conclusion, both dietary approaches allowed patients to improve their state of health, but VLCKD has shown better results on body composition as well as on GM profile.
Collapse
Affiliation(s)
- Andrea Deledda
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Vanessa Palmas
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Vitor Heidrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
| | - Michele Fosci
- Endocrinology Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy
| | - Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giulia Cambarau
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Alessio Lai
- Diabetologia, P.O. Binaghi, ASSL Cagliari, 09126 Cagliari, Italy
| | - Marietta Melis
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Elisabetta Loi
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Andrea Loviselli
- Endocrinology Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Fernanda Velluzzi
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
19
|
Helichrysum italicum (Roth) G. Don and Helichrysum arenarium (L.) Moench Infusion Consumption Affects the Inflammatory Status and the Composition of Human Gut Microbiota in Patients with Traits of Metabolic Syndrome: A Randomized Comparative Study. Foods 2022. [PMCID: PMC9601527 DOI: 10.3390/foods11203277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Helichrysum italicum (Roth) G. Don (HI) and Helichrysum arenarium (L.) Moench (HA) are rich in polyphenols and their infusions have beneficial effects for patients with metabolic syndrome. To investigate whether these effects are mediated by the gut microbiota, we analysed the effects of daily consumption of HI or HA infusion on the composition of gut microbiota, inflammatory status, and zonulin, a marker of gut barrier permeability. The study was a randomized, double-blind comparative trial. Thirty participants were randomly assigned to two groups and received either HA or HI tea filter bags, each containing 1 g of dried plant material, for daily consumption lasting 4 weeks. The results show that consumption of both infusions resulted in a reduction of some genera belonging to Firmicutes and in a slight but significant reduction in Shannon diversity index. Consumption of HI infusion significantly reduced serum levels of proinflammatory markers and zonulin alongside with the observed trend of Proteobacteria reduction. It can therefore be concluded that the HI and HA infusions could act as prebiotics and thus improve the intestinal environment. In addition, HI infusion has a positive impact on microbial dysbiosis and gut barrier dysfunction that occur in obesity and metabolic syndrome.
Collapse
|
20
|
Li A, Wang J, Wang Y, Zhang B, Chen Z, Zhu J, Wang X, Wang S. Tartary Buckwheat (Fagopyrum tataricum) Ameliorates Lipid Metabolism Disorders and Gut Microbiota Dysbiosis in High-Fat Diet-Fed Mice. Foods 2022; 11:foods11193028. [PMID: 36230104 PMCID: PMC9563051 DOI: 10.3390/foods11193028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Jinqiao II, a newly cultivated variety of tartary buckwheat (Fagopyrum tataricum), has been reported to exhibit a higher yield and elevated levels of functional compounds compared to traditional native breeds. We aimed to investigate the potential of Jinqiao II tartary buckwheat to alleviate lipid metabolism disorders by detecting serum biochemistry, pathological symptoms, gene expression profiling, and gut microbial diversity. C57BL/6J mice were provided with either a normal diet; a high-fat diet (HFD); or HFD containing 5%, 10%, and 20% buckwheat for 8 weeks. Our results indicate that Jinqiao II tartary buckwheat attenuated HFD-induced hyperlipidemia, fat accumulation, hepatic damage, endotoxemia, inflammation, abnormal hormonal profiles, and differential lipid-metabolism-related gene expression at mRNA and protein levels in response to the dosages, and high-dose tartary buckwheat exerted optimal outcomes. Gut microbiota sequencing also revealed that the Jinqiao II tartary buckwheat elevated the level of microbial diversity and the abundance of advantageous microbes (Alistipes and Alloprevotella), lowered the abundance of opportunistic pathogens (Ruminococcaceae, Blautia, Ruminiclostridium, Bilophila, and Oscillibacter), and altered the intestinal microbiota structure in mice fed with HFD. These findings suggest that Jinqiao II tartary buckwheat might serve as a competitive candidate in the development of functional food to prevent lipid metabolic abnormalities.
Collapse
Affiliation(s)
- Ang Li
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300350, China
| | - Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300350, China
| | - Yuanyifei Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300350, China
| | - Bowei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300350, China
| | - Zhenjia Chen
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
| | - Junling Zhu
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xiaowen Wang
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
- Institute of Medicinal Plant, Shanxi Agricultural University, Jinzhong 030801, China
| | - Shuo Wang
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300350, China
- Correspondence: ; Tel.: +86-22-8535-8445
| |
Collapse
|
21
|
Attaye I, Warmbrunn MV, Boot ANAF, van der Wolk SC, Hutten BA, Daams JG, Herrema H, Nieuwdorp M. A Systematic Review and Meta-analysis of Dietary Interventions Modulating Gut Microbiota and Cardiometabolic Diseases-Striving for New Standards in Microbiome Studies. Gastroenterology 2022; 162:1911-1932. [PMID: 35151697 DOI: 10.1053/j.gastro.2022.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND & AIMS Cardiometabolic diseases (CMDs) have shared properties and causes. Insulin resistance is a risk factor and characteristic of CMDs and has been suggested to be modulated by plasma metabolites derived from gut microbiota (GM). Because diet is among the most important modulators of GM, we performed a systematic review of the literature to assess whether CMDs can be modulated via dietary interventions targeting the GM. METHODS A systematic review of the literature for clinical studies was performed on Ovid MEDLINE and Ovid Embase. Studies were assessed for risk of bias and patterns of intervention effects. A meta-analysis with random effects models was used to evaluate the effect of dietary interventions on clinical outcomes. RESULTS Our search yielded 4444 unique articles, from which 15 randomized controlled trials and 6 nonrandomized clinical trials were included. The overall risk of bias was high in all studies. In general, most dietary interventions changed the GM composition, but no consistent effect could be found. Results of the meta-analyses showed that only diastolic blood pressure is decreased across interventions compared with controls (mean difference: -3.63 mm Hg; 95% confidence interval, -7.09 to -0.17; I2 = 0%, P = .04) and that a high-fiber diet was associated with reduced triglyceride levels (mean difference: -0.69 mmol/L; 95% confidence interval, -1.36 to -0.02; I2 = 59%, P = .04). Other CMD parameters were not affected. CONCLUSIONS Dietary interventions modulate GM composition, blood pressure, and circulating triglycerides. However, current studies have a high methodological heterogeneity and risk of bias. Well-designed and controlled studies are thus necessary to better understand the complex interaction between diet, microbiome, and CMDs. PROSPERO CRD42020188405.
Collapse
Affiliation(s)
- Ilias Attaye
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Moritz V Warmbrunn
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Aureline N A F Boot
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Suze C van der Wolk
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Barbara A Hutten
- Department of Epidemiology and Data Science, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Joost G Daams
- Medical Library, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Hilde Herrema
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Cantoni C, Dorsett Y, Fontana L, Zhou Y, Piccio L. Effects of dietary restriction on gut microbiota and CNS autoimmunity. Clin Immunol 2022; 235:108575. [PMID: 32822833 PMCID: PMC7889763 DOI: 10.1016/j.clim.2020.108575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 05/12/2020] [Accepted: 08/14/2020] [Indexed: 02/03/2023]
Abstract
Multiple sclerosis (MS) is the most common central nervous system (CNS) autoimmune disease. It is due to the interplay of genetic and environmental factors. Current opinion is that diet could play a pathogenic role in disease onset and development. Dietary restriction (DR) without malnutrition markedly improves health and increases lifespan in multiple model organisms. DR regimens that utilize continuous or intermittent food restriction can induce anti-inflammatory, immuno-modulatory and neuroendocrine adaptations promoting health. These adaptations exert neuroprotective effects in the main MS animal model, experimental autoimmune encephalomyelitis (EAE). This review summarizes the current knowledge on DR-induced changes in gut microbial composition and metabolite production and its impact on underlying functional mechanisms. Studies demonstrating the protective effects of DR regimens on EAE and people with MS are also presented. This is a rapidly developing research field with important clinical implications for personalized dietary interventions in MS prevention and treatment.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yair Dorsett
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia,Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW 2006, Australia,Department of Clinical and Experimental Sciences, Brescia University School of Medicine, Brescia, Italy
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia.,Corresponding author: Laura Piccio, MD PhD, 1) Brain and Mind Centre, University of Sydney, 94 Mallett St Camperdown, NSW, 2050, Australia, , 2) Washington University School of Medicine, Dept. of Neurology, Campus Box 8111; 660 S. Euclid Avenue, St. Louis, MO 63110; USA, Phone: (314) 747-4591; Fax: (314) 747-1345;
| |
Collapse
|
23
|
Liu X, Mao B, Gu J, Wu J, Cui S, Wang G, Zhao J, Zhang H, Chen W. Blautia-a new functional genus with potential probiotic properties? Gut Microbes 2022; 13:1-21. [PMID: 33525961 PMCID: PMC7872077 DOI: 10.1080/19490976.2021.1875796] [Citation(s) in RCA: 785] [Impact Index Per Article: 261.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Blautia is a genus of anaerobic bacteria with probiotic characteristics that occur widely in the feces and intestines of mammals. Based on phenotypic and phylogenetic analyses, some species in the genera Clostridium and Ruminococcus have been reclassified as Blautia, so to date, there are 20 new species with valid published names in this genus. An extensive body of research has recently focused on the probiotic effects of this genus, such as biological transformation and its ability to regulate host health and alleviate metabolic syndrome. This article reviews the origin and biological characteristics of Blautia and the factors that affect its abundance and discusses its role in host health, thus laying a theoretical foundation for the development of new functional microorganisms with probiotic properties.
Collapse
Affiliation(s)
- Xuemei Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,CONTACT Bingyong Mao
| | - Jiayu Gu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiaying Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,Shumao Cui School of Food Science and Technology, Jiangnan University, Lihu Avenue 1800, Wuxi214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China,Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China
| |
Collapse
|
24
|
Rodríguez-Peña MM, Astiarraga B, Seco J, Ceperuelo-Mallafré V, Caballé A, Pérez-Brocal V, Attolini CSO, Moya A, Llauradó G, Megía A, Pellitero S, Vilarrasa N, Fernández-Veledo S, Vendrell J. Changes in glucagon-like peptide 1 and 2 levels in people with obesity after a diet-induced weight-loss intervention are related to a specific microbiota signature: A prospective cohort study. Clin Transl Med 2021; 11:e575. [PMID: 34841718 PMCID: PMC8571947 DOI: 10.1002/ctm2.575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/05/2023] Open
Affiliation(s)
- M-Mar Rodríguez-Peña
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Brenno Astiarraga
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain.,Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Tarragona, Spain
| | - Jesus Seco
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain
| | - Victoria Ceperuelo-Mallafré
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain.,Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Tarragona, Spain
| | - Adrià Caballé
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Vicente Pérez-Brocal
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain.,Biomedical Research Networking Centre for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Andrés Moya
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain.,Biomedical Research Networking Centre for Epidemiology and Public Health (CIBERESP), Madrid, Spain.,Spanish National Research Council (CSIC), Institute for Integrative Systems Biology (I2SysBio), University of Valencia, Valencia, Spain
| | - Gemma Llauradó
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.,Department of Endocrinology and Nutrition, Hospital del Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana Megía
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Pellitero
- Endocrinology and Nutrition Department, Institute Research and Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Núria Vilarrasa
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.,Department of Endocrinology and Nutrition, L'Hospitalet de Llobregat, Bellvitge University-IDIBELL, Barcelona, Spain
| | - Sonia Fernández-Veledo
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Vendrell
- Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.,Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Tarragona, Spain
| |
Collapse
|
25
|
Shu X, Li M, Cao Y, Li C, Zhou W, Ji G, Zhang L. Berberine Alleviates Non-alcoholic Steatohepatitis Through Modulating Gut Microbiota Mediated Intestinal FXR Activation. Front Pharmacol 2021; 12:750826. [PMID: 34603061 PMCID: PMC8484326 DOI: 10.3389/fphar.2021.750826] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Berberine is a natural plant alkaloid isolated from a diverse range of genera, it obtains anti-inflammatory, anti-obesity, and hepatoprotective properties, and is a promising agent for non-alcoholic steatohepatitis (NASH). Farnesoid X receptor (FXR) is a bile acid receptor and a drug target for NASH, however, the underlying mechanisms of berberine on regulating FXR are still unknown. In the present study, we feed mice with a 12-week high-fat diet with interval dextran sulfate sodium (0.5% in drinking water) diet to induce NASH, and treat the mice with berberine (100 mg/kg per day) via oral gavage for additional 4 weeks. We demonstrate that administration of berberine alleviates steatosis and infiltration of inflammatory cells in the liver of NASH mice. We apply 16S ribosomal DNA sequencing to screen the structure of gut microbiota, and ultra-performance liquid chromatography-tandem mass spectrometry analysis to determine the bile acid profiles. The results show that berberine modulates gut dysbiosis, and specifically increases the relative abundance of Clostridiales, Lactobacillaceae, and Bacteroidale. Berberine modulated microbiomes are associated with bile acid de-conjugation and transformation, which are consistent with the altered bile acid species (e.g., deoxycholic acid, ursodeoxycholic acid) upon berberine treatment. BA species that respond to berberine treatment are known FXR agonists, thus we performed quantitative Real Time-PCR and western blot to examine the FXR pathway, and find that berberine up-regulates intestinal FXR and fibroblast growth factor 15 (FGF15) expression, and the secretion of FGF15 further inhibits lipogenesis and nuclear factor-κB activation in the liver. Whereas the beneficial effects of berberine are blunted in FXR knockout mice. Our results reveal that berberine alleviates NASH by modulating the interplay of gut microbiota and bile acid metabolism, as well as the subsequent intestinal FXR activation.
Collapse
Affiliation(s)
- Xiangbing Shu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Geratology, Baoshan Branch of Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Cao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunlin Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Kökten T, Hansmannel F, Ndiaye NC, Heba AC, Quilliot D, Dreumont N, Arnone D, Peyrin-Biroulet L. Calorie Restriction as a New Treatment of Inflammatory Diseases. Adv Nutr 2021; 12:1558-1570. [PMID: 33554240 PMCID: PMC8321869 DOI: 10.1093/advances/nmaa179] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Immoderate calorie intake coupled with a sedentary lifestyle are major determinants of health issues and inflammatory diseases in modern society. The balance between energy consumption and energy expenditure is critical for longevity. Excessive energy intake and adiposity cause systemic inflammation, whereas calorie restriction (CR) without malnutrition, exerts a potent anti-inflammatory effect. The objective of this review was to provide an overview of different strategies used to reduce calorie intake, discuss physiological mechanisms by which CR might lead to improved health outcomes, and summarize the present knowledge about inflammatory diseases. We discuss emerging data of observational studies and randomized clinical trials on CR that have been shown to reduce inflammation and improve human health.
Collapse
Affiliation(s)
- Tunay Kökten
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
| | - Franck Hansmannel
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
| | - Ndeye Coumba Ndiaye
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
| | - Anne-Charlotte Heba
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
| | - Didier Quilliot
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
- Université de Lorraine, Centre Hospitalier Régional Universitaire (CHRU)-Nancy, Department of Diabetology-Endocrinology-Nutrition, Nancy, France
| | - Natacha Dreumont
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
| | - Djésia Arnone
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
| | - Laurent Peyrin-Biroulet
- Université de Lorraine, Inserm U1256 NGERE (Nutrition—Genetics and Exposure to Environmental Risks), Nancy, France
- Université de Lorraine, Centre Hospitalier Régional Universitaire (CHRU)-Nancy, Department of Gastroenterology, Nancy, France
| |
Collapse
|
27
|
Nogal A, Louca P, Zhang X, Wells PM, Steves CJ, Spector TD, Falchi M, Valdes AM, Menni C. Circulating Levels of the Short-Chain Fatty Acid Acetate Mediate the Effect of the Gut Microbiome on Visceral Fat. Front Microbiol 2021; 12:711359. [PMID: 34335546 PMCID: PMC8320334 DOI: 10.3389/fmicb.2021.711359] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background Acetate is a short-chain fatty acid (SCFA) produced by gut bacteria, which has been implicated in cardio-metabolic health. Here we examine the relationships of circulating acetate levels with gut microbiome composition and diversity and with visceral fat in a large population-based cohort. Results Microbiome alpha-diversity was positively correlated with circulating acetate levels (Shannon, Beta [95%CI] = 0.12 [0.06, 0.18], P = 0.002) after adjustment for covariates. Six serum acetate-associated bacterial genera were also identified, including positive correlations with Coprococcus, Barnesiella, Ruminococcus, and Ruminococcaceae NK4A21 and negative correlations were observed with Lachnoclostridium and Bacteroides. We also identified a correlation between visceral fat and serum acetate levels (Beta [95%CI] = −0.07 [−0.11, −0.04], P = 2.8 × 10–4) and between visceral fat and Lachnoclostridium (Beta [95%CI] = 0.076 [0.042, 0.11], P = 1.44 × 10–5). Formal mediation analysis revealed that acetate mediates ∼10% of the total effect of Lachnoclostridium on visceral fat. The taxonomic diversity showed that Lachnoclostridium and Coprococcus comprise at least 18 and 9 species, respectively, including novel bacterial species. By predicting the functional capabilities, we found that Coprococcus spp. present pathways involved in acetate production and metabolism of vitamins B, whereas we identified pathways related to the biosynthesis of trimethylamine (TMA) and CDP-diacylglycerol in Lachnoclostridium spp. Conclusions Our data indicates that gut microbiota composition and diversity may influence circulating acetate levels and that acetate might exert benefits on certain cardio-metabolic disease risk by decreasing visceral fat. Coprococcus may play an important role in host health by its production of vitamins B and SCFAs, whereas Lachnoclostridium might have an opposing effect by influencing negatively the circulating levels of acetate and being involved in the biosynthesis of detrimental lipid compounds.
Collapse
Affiliation(s)
- Ana Nogal
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Panayiotis Louca
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Xinyuan Zhang
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Philippa M Wells
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Ana M Valdes
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom.,Nottingham NIHR Biomedical Research Centre at the School of Medicine, Nottingham City Hospital, University of Nottingham, Nottingham, United Kingdom
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| |
Collapse
|
28
|
Dietary restrictions modulate the gut microbiota: Implications for health and disease. Nutr Res 2021; 89:10-22. [PMID: 33878569 DOI: 10.1016/j.nutres.2021.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/12/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
The health benefits of carefully restricting the energy intake in a strategic manner whilst avoiding malnutrition are widely discussed. In the recent years, the great impact of the gut microbiota on its host has been clarified more and more. Since the gut microbiota produces a number of metabolites and molecules that can affect host metabolism, modulating it with dietary restriction can influence the health and the progression of disease of its host on various levels. This review comprises 15 studies investigating the effect of different variants of fasting and caloric restriction on the gastrointestinal microbiome and its metabolites. The data suggest that changing the gut microbiota composition by dietary restriction has the potential to positively influence the progression of several diseases such as obesity, diabetes, neurological diseases or inflammatory bowel disease. Finally, the relevance of the findings for clinical practice is evaluated and approaches for future research are proposed.
Collapse
|
29
|
Liao J, Xie X, Gao J, Zhang Z, Qu F, Cui H, Cao Y, Han X, Zhao J, Wen W, Wang H. Jian-Gan-Xiao-Zhi Decoction Alleviates Inflammatory Response in Nonalcoholic Fatty Liver Disease Model Rats through Modulating Gut Microbiota. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5522755. [PMID: 33824675 PMCID: PMC8007356 DOI: 10.1155/2021/5522755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Jian-Gan-Xiao-Zhi decoction (JGXZ), composed of Salvia miltiorrhiza Bunge, Panax notoginseng, Curcuma zedoaria, and other 9 types of herbs, has demonstrated beneficial effects on nonalcoholic fatty liver disease (NAFLD). However, the mechanisms behind JGXZ's impact on NAFLD remain unknown. METHODS In this study, a NAFLD rat model induced by a high-fat diet (HFD) received oral treatment of JGXZ (8 or 16 g crude herb/kg) for 12 weeks. The therapeutic effects of JGXZ on NAFLD model rats were investigated through blood lipid levels and pathological liver changes. 16S rRNA analysis was used to study the changes in gut microbiota after JGXZ treatment. The expressions of occludin and tight junction protein 1 (ZO-1) in the colon were investigated using immunostaining to study the effects of JGXZ on gut permeability. The anti-inflammatory effects of JGXZ were also studied through measuring the levels of IL-1β, IL-6, and TNF-α in the serum and liver. RESULTS JGXZ treatment could decrease body weight and ameliorate dyslipidemia in NAFLD model rats. H&E and Oil Red O staining indicated that JGXZ reduced steatosis and infiltration of inflammatory cells in the liver. 16S rRNA analysis showed that JGXZ impacted the diversity of gut microbiota, decreasing the Firmicutes-to-Bacteroidetes ratio, and increasing the relative abundance of probiotics, such as Alloprevotella, Lactobacillus, and Turicibacter. Gut permeability evaluation found that the expressions of ZO-1 and occludin in the colon were increased after JGXZ treatment. Moreover, JGXZ treatment could decrease the levels of IL-1β, IL-6, and TNF-α in the serum and liver. CONCLUSIONS Our study illustrated that JGXZ could ameliorate NAFLD through modulating gut microbiota, decreasing gut permeability, and alleviating inflammatory response.
Collapse
Affiliation(s)
- Jiabao Liao
- Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
| | - Xuehua Xie
- Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Jinmei Gao
- Fujian People's Hospital of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zhaiyi Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fei Qu
- Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
| | - Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yongjun Cao
- Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| | - Xue Han
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Jie Zhao
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Weibo Wen
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Hongwu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
30
|
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
|
31
|
Wang M, Wang B, Wang S, Lu H, Wu H, Ding M, Ying L, Mao Y, Li Y. Effect of Quercetin on Lipids Metabolism Through Modulating the Gut Microbial and AMPK/PPAR Signaling Pathway in Broilers. Front Cell Dev Biol 2021; 9:616219. [PMID: 33634119 PMCID: PMC7900412 DOI: 10.3389/fcell.2021.616219] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
The present study was conducted to investigate effects and mechanism of quercetin on lipids metabolism in broilers. 480 AA broilers were randomly allotted to four treatments (0, 0.2, 0.4, and 0.6 g/kg quercetin) for 42 days. Compared with the control, 0.6 g/kg quercetin significantly decreased percentage of abdominal fat (P < 0.05); 0.2, 0.4, and 0.6 g/kg quercetin significantly decreased relative abundance of Lachnospiraceae and Desulfovibrionaceae (P < 0.05, P < 0.05, P < 0.01; P < 0.01, P < 0.01, P < 0.01); 0.2 g/kg quercetin significantly increased mRNA expression of PI3K, AMPKα1, AMPKα2, AMPKβ2, LKB1 (P < 0.01, P < 0.01, P < 0.05, P < 0.01, P < 0.05), and significantly reduced mRNA expression of SREBP1 and PPARγ (P < 0.01, P < 0.05); 0.4 g/kg quercetin significantly increased mRNA expression of LKB1 and PKB (P < 0.05, P < 0.01) and significantly reduced mRNA expression of ACC, HMGR, PPARγ, and SREBP1 (P < 0.05, P < 0.01, P < 0.01, P < 0.01); 0.6 g/kg quercetin significantly increased mRNA expression of AMPKγ, LKB1, CPT1, PPARα, PKB (P < 0.01, P < 0.01, P < 0.01, P < 0.05, P < 0.05), and significantly reduced the mRNA expression of PI3K, ACC, HMGR, PPARγ, SREBP1 (P < 0.05, P < 0.05, P < 0.01, P < 0.01, P < 0.01); 0.2 g/kg quercetin significantly increased protein expression of AMPK (P < 0.01); 0.6 g/kg quercetin significantly increased protein expression of LKB1 (P < 0.01), 0.2 and 0.6 g/kg quercetin significantly increased protein expression of PI3K, PKB, CPT1 (P < 0.05, P < 0.01, P < 0.05, P < 0.01, P < 0.01, P < 0.01), and significantly reduced protein expression of ACC and SREBP1 (P < 0.01, P < 0.01, P < 0.01, P < 0.01). In conclusion, quercetin improved lipid metabolism by modulating gut microbial and AMPK/PPAR signaling pathway in broilers.
Collapse
Affiliation(s)
- Mi Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China.,College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Bo Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Shanshan Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Han Lu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Hao Wu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Manyi Ding
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Linlin Ying
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Yanjun Mao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Yao Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
32
|
Haidari F, Hojhabrimanesh A, Helli B, Seyedian SS, Ahmadi-Angali K, Abiri B. A hypocaloric high-protein diet supplemented with β-cryptoxanthin improves non-alcoholic fatty liver disease: a randomized controlled trial. BMC Gastroenterol 2020; 20:349. [PMID: 33081717 PMCID: PMC7576825 DOI: 10.1186/s12876-020-01502-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Background Despite promising animal data, there is no randomized controlled trial (RCT) on the effects of high protein (HP)-diet and/or β-cryptoxanthin in non-alcoholic fatty liver disease (NAFLD). Aims: Safety and efficacy assessment of a hypocaloric HP-diet supplemented with β-cryptoxanthin in NAFLD. Methods Ninety-two Iranian NAFLD outpatients were recruited for this 12-week, single-center, parallel-group, double-blind RCT and randomized into 4 arms (n = 23): HP-diet and β-cryptoxanthin (hypocaloric HP-diet + β-cryptoxanthin), HP-diet (hypocaloric HP-diet + placebo), β-cryptoxanthin (standard hypocaloric diet + β-cryptoxanthin), and control (standard hypocaloric diet + placebo). Serum levels of liver enzymes and grade of hepatic steatosis were assessed at baseline and study endpoint as outcome measures. Results In the intention-to-treat population (N = 92), HP-diet and β-cryptoxanthin group experienced greater 12-week reductions in serum levels of liver enzymes than control group (mean difference for alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and gamma-glutamyl transferase: − 27.2, − 7.2, − 39.2, and − 16.3 IU/L, respectively; all p < 0.010). Clinical remission rate (achieving grade 0 hepatic steatosis) in HP-diet and β-cryptoxanthin group (82.6%) was also higher than other groups (13.0%, 17.4%, and 0.0% in HP-diet, β-cryptoxanthin, and control groups, respectively; p < 0.001). Sixteen patients reported minor adverse events. Conclusion A hypocaloric HP-diet supplemented with β-cryptoxanthin safely and efficaciously improves NAFLD. Trial registration number This trial was registered at https://www.irct.ir as IRCT2017060210181N10.
Collapse
Affiliation(s)
- Fatemeh Haidari
- Department of Nutrition Sciences, Nutrition and Metabolic Diseases Research Center, School of Paramedical Sciences, Ahvaz Jundishapur University of Medical Sciences, 61357-15794, Ahvaz, Iran
| | - Abdollah Hojhabrimanesh
- Department of Nutrition Sciences, Nutrition and Metabolic Diseases Research Center, School of Paramedical Sciences, Ahvaz Jundishapur University of Medical Sciences, 61357-15794, Ahvaz, Iran.
| | - Bizhan Helli
- Department of Nutrition Sciences, Nutrition and Metabolic Diseases Research Center, School of Paramedical Sciences, Ahvaz Jundishapur University of Medical Sciences, 61357-15794, Ahvaz, Iran
| | - Seyed-Saeed Seyedian
- Department of Gastroenterology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kambiz Ahmadi-Angali
- Department of Epidemiology and Biostatistics, School of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Behnaz Abiri
- Department of Nutrition, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
33
|
Pisanu S, Palmas V, Madau V, Casula E, Deledda A, Cusano R, Uva P, Vascellari S, Boi F, Loviselli A, Manzin A, Velluzzi F. Impact of a Moderately Hypocaloric Mediterranean Diet on the Gut Microbiota Composition of Italian Obese Patients. Nutrients 2020; 12:nu12092707. [PMID: 32899756 PMCID: PMC7551852 DOI: 10.3390/nu12092707] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/21/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Although it is known that the gut microbiota (GM) can be modulated by diet, the efficacy of specific dietary interventions in determining its composition and diversity in obese patients remains to be ascertained. The present work aims to evaluate the impact of a moderately hypocaloric Mediterranean diet on the GM of obese and overweight patients (OB). The GM of 23 OB patients (F/M = 20/3) was compared before (T0) and after 3 months (T3) of nutritional intervention (NI). Fecal samples were analyzed by Illumina MiSeq sequencing of the 16S rRNA gene. At baseline, GM characterization confirmed typical obesity-associated dysbiosis. After 3 months of NI, patients presented a statistically significant reduction in body weight and fat mass, along with changes in the relative abundance of many microbial patterns. In fact, an increase in the abundance of several Bacteroidetes taxa (i.e., Sphingobacteriaceae, Sphingobacterium, Bacteroides spp., Prevotella stercorea) and a depletion of many Firmicutes taxa (i.e., Lachnospiraceae members, Ruminococcaceae and Ruminococcus, Veillonellaceae, Catenibacterium, Megamonas) were observed. In addition, the phylum Proteobacteria showed an increased abundance, while the genus Sutterella, within the same phylum, decreased after the intervention. Metabolic pathways, predicted by bioinformatic analyses, showed a decrease in membrane transport and cell motility after NI. The present study extends our knowledge of the GM profiles in OB, highlighting the potential benefit of moderate caloric restriction in counteracting the gut dysbiosis.
Collapse
Affiliation(s)
- Silvia Pisanu
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy; (S.P.); (V.P.); (V.M.); (E.C.); (S.V.); (A.M.)
| | - Vanessa Palmas
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy; (S.P.); (V.P.); (V.M.); (E.C.); (S.V.); (A.M.)
| | - Veronica Madau
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy; (S.P.); (V.P.); (V.M.); (E.C.); (S.V.); (A.M.)
| | - Emanuela Casula
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy; (S.P.); (V.P.); (V.M.); (E.C.); (S.V.); (A.M.)
| | - Andrea Deledda
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (A.D.); (F.B.); (F.V.)
| | - Roberto Cusano
- CRS4, Science and Technology Park Polaris, Piscina Manna, Pula, 09010 Cagliari, Italy; (R.C.); (P.U.)
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Piscina Manna, Pula, 09010 Cagliari, Italy; (R.C.); (P.U.)
| | - Sarah Vascellari
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy; (S.P.); (V.P.); (V.M.); (E.C.); (S.V.); (A.M.)
| | - Francesco Boi
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (A.D.); (F.B.); (F.V.)
| | - Andrea Loviselli
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (A.D.); (F.B.); (F.V.)
- Correspondence: ; Tel.: +39-070-675-4268
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy; (S.P.); (V.P.); (V.M.); (E.C.); (S.V.); (A.M.)
| | - Fernanda Velluzzi
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (A.D.); (F.B.); (F.V.)
| |
Collapse
|
34
|
Wei L, Yue F, Xing L, Wu S, Shi Y, Li J, Xiang X, Lam SM, Shui G, Russell R, Zhang D. Constant Light Exposure Alters Gut Microbiota and Promotes the Progression of Steatohepatitis in High Fat Diet Rats. Front Microbiol 2020; 11:1975. [PMID: 32973715 PMCID: PMC7472380 DOI: 10.3389/fmicb.2020.01975] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) poses a significant health concern worldwide. With the progression of urbanization, light pollution may be a previously unrecognized risk factor for NAFLD/NASH development. However, the role of light pollution on NAFLD is insufficiently understood, and the underlying mechanism remains unclear. Interestingly, recent studies indicate the gut microbiota affects NAFLD/NASH development. Therefore, the present study explored effects of constant light exposure on NAFLD and its related microbiotic mechanisms. Materials and Methods Twenty-eight SD male rats were divided into four groups (n = 7 each): rats fed a normal chow diet, and exposed to standard light-dark cycle (ND-LD); rats fed a normal chow diet, and exposed to constant light (ND-LL); rats fed a high fat diet, and exposed to standard light-dark cycle (HFD-LD); and rats on a high fat diet, and exposed to constant light (HFD-LL). Body weight, hepatic pathophysiology, gut microbiota, and short/medium chain fatty acids in colon contents, serum lipopolysaccharide (LPS), and liver LPS-binding protein (LBP) mRNA expression were documented post intervention and compared among groups. Result In normal chow fed groups, rats exposed to constant light displayed glucose abnormalities and dyslipidemia. In HFD-fed rats, constant light exposure exacerbated glucose abnormalities, insulin resistance, inflammation, and liver steatohepatitis. Constant light exposure altered composition of gut microbiota in both normal chow and HFD fed rats. Compared with HFD-LD group, HFD-LL rats displayed less Butyricicoccus, Clostridium, and Turicibacter, butyrate levels in colon contents, decreased colon expression of occludin-1 and zonula occluden−1 (ZO-1), and increased serum LPS and liver LBP mRNA expression. Conclusion Constant light exposure impacts gut microbiota and its metabolic products, impairs gut barrier function and gut-liver axis, promotes NAFLD/NASH progression in HFD rats.
Collapse
Affiliation(s)
- Lin Wei
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Fangzhi Yue
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Xing
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanyu Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinchen Li
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xingwei Xiang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Ryan Russell
- Cardiomatabolic Exercise Lab Director, Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
35
|
Shah RA, Alkhouri N, Kowdley KV. Emerging drugs for the treatment of non-alcoholic steatohepatitis: a focused review of farnesoid X receptor agonists. Expert Opin Emerg Drugs 2020; 25:251-260. [DOI: 10.1080/14728214.2020.1796968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Zhou L, Xiao X, Li M, Zhang Q, Yu M, Zheng J, Deng M. Maternal Exercise Improves High-Fat Diet-Induced Metabolic Abnormalities and Gut Microbiota Profiles in Mouse Dams and Offspring. Front Cell Infect Microbiol 2020; 10:292. [PMID: 32626663 PMCID: PMC7311581 DOI: 10.3389/fcimb.2020.00292] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Early-life overnutrition programs increased risks of metabolic disorders in adulthood. Regular exercise has been widely accepted to be an effective measure to maintain metabolic health. However, the intergenerational effects of maternal exercise and the specific mechanism are largely unclear. Our objective was to investigate whether maternal exercise could alleviate the metabolic disturbances induced by early-life overnutrition in both dams and offspring and to explore the role of gut microbiota in mediating the effects. C57BL/6 female mice were randomly divided into three groups: the control group, which were fed a normal control diet; high-fat group, which received a high-fat diet; and high-fat with exercise intervention group, which was fed a high-fat diet and received a voluntary wheel running training. The diet intervention started from 3 weeks prior to mating and lasted throughout pregnancy and lactation. The exercise intervention was only prior to and during pregnancy. The male offspring got free access to normal chow diet from weaning to 24 weeks of age. Glucose tolerance test and biochemical parameters were detected in dams at weaning and offspring at 8 and 24 weeks of age. Their cecal contents were collected for the 16 s rDNA amplicon sequencing. The results showed that maternal high-fat diet resulted in significant glucose intolerance, insulin resistance, and lipid profiles disorders in both dams and offspring. Maternal exercise markedly improved insulin sensitivity in dams and metabolic disorders in offspring from young into adulthood. The decrease in unfavorable bacteria and the persistent enrichment of short-chain fatty acids-producers from mothers to adult offspring, particularly the genus Odoribacter, were all associated with the improvement of metabolism by maternal exercise. Overall, maternal exercise could significantly mitigate the detrimental effects of a maternal high-fat diet on metabolism in both dams and male offspring. The continuous alterations in gut microbiota might be a critical factor in deciphering the metabolic benefits of maternal exercise, which provides some novel evidence and targets for combating metabolic diseases.
Collapse
Affiliation(s)
- Liyuan Zhou
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Li
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Miao Yu
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia Zheng
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingqun Deng
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Translational Medicine Center, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Jian C, Luukkonen P, Sädevirta S, Yki-Järvinen H, Salonen A. Impact of short-term overfeeding of saturated or unsaturated fat or sugars on the gut microbiota in relation to liver fat in obese and overweight adults. Clin Nutr 2020; 40:207-216. [PMID: 32536582 DOI: 10.1016/j.clnu.2020.05.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUNDS & AIMS Intestinal microbiota may be causally involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). We aimed to study the effect of short-term overfeeding on human gut microbiota in relation to baseline and overfeeding-induced liver steatosis. We also asked whether the baseline microbiota composition is associated to the overfeeding-induced increase in liver fat. METHODS In a randomized trial, 38 overweight and obese subjects were assigned to consume an excess of 1000 kcal/day of diets rich in either saturated fat, unsaturated fat, or simple sugars for 3 weeks. Fasting blood samples and 1H-MR spectroscopy were used for extensive clinical phenotyping as previously reported (PMID: 29844096). Fecal samples were collected for the analysis of the gut microbiota using 16S rRNA amplicon sequencing, imputed metagenomics and qPCR. Microbiota results were correlated with dietary intakes and clinical measurements before and during overfeeding. RESULTS The overall community structure of the microbiota remained highly stable and personalized during overfeeding based on between-sample Bray-Curtis dissimilarity, but the relative abundances of individual taxa were altered in a diet-specific manner: overfeeding saturated fat increased Proteobacteria, while unsaturated fat increased butyrate producers. Sugar overfeeding increased Lactococcus and Escherichia coli. Imputed functions of the gut microbiota were not affected by overfeeding. Several taxa affected by overfeeding significantly correlated with the changes in host metabolic markers. The baseline levels of proteobacterial family Desulfovibrionaceae, and especially genus Bilophila, were significantly associated to overfeeding-induced liver fat increase independently of the diet arm. In general, limited overlap was observed between the overfeeding-induced microbiota changes and the liver fat-associated microbiota features at baseline. CONCLUSIONS Our work indicates that the human gut microbiota is resilient to short-term overfeeding on community level, but specific taxa are altered on diet composition-dependent manner. Generalizable microbiota signatures directly associated with liver steatosis could not be identified. Instead, the carriage of Bilophila was identified as a potential novel risk factor for diet-induced liver steatosis in humans. Clinical trial registry number: NCT02133144 listed on NIH website: ClinicalTrials.gov.
Collapse
Affiliation(s)
- Ching Jian
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Panu Luukkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Sanja Sädevirta
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Hannele Yki-Järvinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
38
|
Abstract
INTRODUCTION NAFLD has grown to become the most prevalent liver disease in the world, with a quarter of the general population estimated to have the disease. NASH, characterized as NAFLD with inflammation, is associated with worsening fibrosis along with increased incidence of HCC. Despite high prevalence of this disease, no pharmacologic treatments approved by regulatory agencies are available. AREAS COVERED This review briefly discusses present understanding of NASH pathology and currently available treatments. We also discuss data on the role of OCA as an FXR agonist in modulating disease in NASH. A comprehensive literature search of review articles, original research articles, and prospective clinical trials from 1998 to the present was performed. EXPERT OPINION Based on 18-month interim findings of the REGENERATE trial, OCA likely improves fibrosis in NASH and therefore may have a beneficial effect in delaying or even preventing cirrhosis. The side effect of an atherogenic lipoprotein profile may adversely affect long-term outcomes, though studies have shown that co-administration of statins is able to mitigate this effect. OCA is likely to become an option for treatment, but the specific context within which it may be prescribed still needs to be clarified.
Collapse
Affiliation(s)
- Raj A Shah
- Liver Institute Northwest , Seattle, WA, USA
| | | |
Collapse
|
39
|
Alyousif Z, Mendoza DR, Auger J, De Carvalho V, Amos S, Sims C, Dahl WJ. Gastrointestinal Tolerance and Microbiome Response to Snacks Fortified with Pea Hull Fiber: A Randomized Trial in Older Adults. Curr Dev Nutr 2020; 4:nzaa005. [PMID: 32025615 PMCID: PMC6994441 DOI: 10.1093/cdn/nzaa005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/31/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Consuming foods with added fiber may help older adults achieve fiber recommendations; however, many high-fiber ingredients have little effect on laxation and may contribute to unpleasant gastrointestinal side effects. OBJECTIVES The aim of the study was to determine the effects of consuming snacks fortified with pea hull fiber (PHF) on stool frequency and form, gastrointestinal symptoms, and appetite in older adults. An exploratory aim was to determine if PHF altered the microbiota profile. METHODS A 10-wk, randomized, blinded, crossover study was carried out. Following a 2-wk baseline period, participants [aged (mean ± SD) 69.7 ± 6.5 y; n = 31; 14 men, 17 women] consumed snacks providing 10 g/d of PHF or a control, each for 2-wk periods followed by 2-wk washouts. Participants used the Bristol Stool Form Scale (BSFS) to record daily stool frequency and gastrointestinal symptoms, and completed the Gastrointestinal Symptom Rating Scale (GSRS) and Simplified Nutritional Appetite Questionnaire (SNAQ) biweekly. One stool was collected per period for 16S ribosomal RNA high-throughput amplicon sequencing of the fecal microbiota profile. RESULTS Participants reported 1.63 ± 0.05 stools/d and 76.6% normal transit stool form at baseline and no change with PHF. GSRS syndrome scores were similarly unchanged. Daily abdominal noises and bloating were higher for PHF versus control, and flatulence was higher for PHF versus baseline, suggesting fermentation in some individuals. There was no evidence to suggest a common PHF-induced microbiome response for the group as a whole; however, a subgroup of participants (n = 7) who responded with increased flatulence (fermenters), harbored many different taxa than nonfermenters, and demonstrated lower abundance of Clostridiales with PHF. Appetite was unchanged with PHF. CONCLUSIONS PHF did not modulate stool form or frequency in older adults with normal bowel habits. Because snacks fortified with PHF did not suppress appetite, PHF may be an appropriate fiber source for older adults at nutritional risk. Microbiome profile may be predictive of gastrointestinal symptom response to PHF. This trial was registered at www.clinicaltrials.gov as NCT02778230.
Collapse
Affiliation(s)
- Zainab Alyousif
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Daniela Rivero Mendoza
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Jérémie Auger
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec, Canada
| | | | - Samantha Amos
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Charles Sims
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Wendy J Dahl
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| |
Collapse
|
40
|
Song M, Chan AT, Sun J. Influence of the Gut Microbiome, Diet, and Environment on Risk of Colorectal Cancer. Gastroenterology 2020; 158:322-340. [PMID: 31586566 PMCID: PMC6957737 DOI: 10.1053/j.gastro.2019.06.048] [Citation(s) in RCA: 493] [Impact Index Per Article: 98.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 02/07/2023]
Abstract
Researchers have discovered associations between elements of the intestinal microbiome (including specific microbes, signaling pathways, and microbiota-related metabolites) and risk of colorectal cancer (CRC). However, it is unclear whether changes in the intestinal microbiome contribute to the development of sporadic CRC or result from it. Changes in the intestinal microbiome can mediate or modify the effects of environmental factors on risk of CRC. Factors that affect risk of CRC also affect the intestinal microbiome, including overweight and obesity; physical activity; and dietary intake of fiber, whole grains, and red and processed meat. These factors alter microbiome structure and function, along with the metabolic and immune pathways that mediate CRC development. We review epidemiologic and laboratory evidence for the influence of the microbiome, diet, and environmental factors on CRC incidence and outcomes. Based on these data, features of the intestinal microbiome might be used for CRC screening and modified for chemoprevention and treatment. Integrated prospective studies are urgently needed to investigate these strategies.
Collapse
Affiliation(s)
- Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, Microbiology/Immunology, UIC Cancer Center, University of Illinois at Chicago, Illinois.
| |
Collapse
|
41
|
An energy-restricted high-protein diet supplemented with β-cryptoxanthin alleviated oxidative stress and inflammation in nonalcoholic fatty liver disease: a randomized controlled trial. Nutr Res 2020; 73:15-26. [DOI: 10.1016/j.nutres.2019.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/12/2019] [Accepted: 08/28/2019] [Indexed: 12/26/2022]
|
42
|
Fang S, Chen X, Zhou L, Wang C, Chen Q, Lin R, Xiao T, Gan Q. Faecal microbiota and functional capacity associated with weaning weight in meat rabbits. Microb Biotechnol 2019; 12:1441-1452. [PMID: 31571427 PMCID: PMC6801154 DOI: 10.1111/1751-7915.13485] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/15/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022] Open
Abstract
Weaning weight is an important economic trait in the meat rabbit industry. Evidence has linked the gut microbiota to health and production performance in rabbits. However, the effect of gut microbiota on meat rabbit weaning weight remains unclear. In this study, we performed 16S rRNA gene sequencing analysis of 135 faecal samples from commercial Ira rabbits. We detected 50 OTUs significantly associated with weaning weight. OTUs that showed positive associations with weaning weight were mostly members of the family Ruminococcaceae which are important in degrading dietary fibres and producing butyrate. On the contrary, OTUs annotated to genera Blautia, Lachnoclostridium and Butyricicoccus correlated with fat deposition were negatively associated with weaning weight. Predicted functional capacity analysis revealed that 91 KOs and 26 KEGG pathways exhibited potential correlations with weaning weight. We found that gut microbiota involved in the metabolism of amino acids, butanoate, energy and monosaccharides affected weaning weight. Additionally, cross-validation analysis indicated that 16.16% of the variation in weaning weight was explained by the gut microbiome. Our findings provide important information to improve weaning weight of meat rabbits by modulating their gut microbiome.
Collapse
Affiliation(s)
- Shaoming Fang
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Xuan Chen
- College of Life ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Liwen Zhou
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Chongchong Wang
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Qiaohui Chen
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Ruiyi Lin
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Tianfang Xiao
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - QianFu Gan
- College of Animal ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| |
Collapse
|
43
|
Duarte SMB, Stefano JT, Vanni DS, Carrilho FJ, Oliveira CPMSD. IMPACT OF CURRENT DIET AT THE RISK OF NON-ALCOHOLIC FATTY LIVER DISEASE (NAFLD). ARQUIVOS DE GASTROENTEROLOGIA 2019; 56:431-439. [PMID: 31721969 DOI: 10.1590/s0004-2803.201900000-67] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/24/2019] [Indexed: 12/15/2022]
Abstract
The nonalcoholic fatty liver disease (NAFLD) affects approximately 20%-30% of general population and is even more prevalent among obese individuals. The risk factors mainly associated with NAFLD are diseases related to the metabolic syndrome, genetics and environment. In this review, we provide a literature compilation evaluating the evidence behind dietary components, including calories intake, fat, protein, fibers and carbohydrate, especially fructose which could be a trigger to development and progression of the NAFLD. In fact, it has been demonstrated that diet is an important factor for the development of NAFLD and its association is complex and extends beyond total energy intake.
Collapse
Affiliation(s)
| | - José Tadeu Stefano
- Universidade de São Paulo, Hospital das Clínicas, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07) do Departamento de Gastroenterologia da FMUSP, São Paulo, SP, Brasil
| | - Denise Siqueira Vanni
- Universidade de São Paulo, Hospital das Clínicas, Divisão de Gastroenterologia e Hepatologia Clínica e Departamento de Gastroenterologia da FMUSP, São Paulo, SP, Brasil
| | - Flair José Carrilho
- Universidade de São Paulo, Faculdade de Medicina, São Paulo, SP, Brasil
- Universidade de São Paulo, Hospital das Clínicas, Divisão de Gastroenterologia e Hepatologia Clínica e Departamento de Gastroenterologia da FMUSP, São Paulo, SP, Brasil
| | - Claudia Pinto Marques Souza de Oliveira
- Universidade de São Paulo, Faculdade de Medicina, São Paulo, SP, Brasil
- Universidade de São Paulo, Hospital das Clínicas, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07) do Departamento de Gastroenterologia da FMUSP, São Paulo, SP, Brasil
| |
Collapse
|
44
|
Wang W, Li Q, Chai W, Sun C, Zhang T, Zhao C, Yuan Y, Wang X, Liu H, Ye H. Lactobacillus paracasei Jlus66 extenuate oxidative stress and inflammation via regulation of intestinal flora in rats with non alcoholic fatty liver disease. Food Sci Nutr 2019; 7:2636-2646. [PMID: 31428351 PMCID: PMC6694609 DOI: 10.1002/fsn3.1118] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
The nonalcoholic fatty liver disease (NAFLD) is a progressive liver disease that affects the health of people in an increasing rate. In the current research, we investigated the beneficial effect of a novel probiotic strain L. paracasei Jlus66 (Jlus66) on rats with high-fat diet (HFD)-induced NAFLD. The intestinal flora of rats was analyzed based on V3-V4 region 16S rDNA sequencing. Moreover, we measured the oxidative stress and inflammation factors in the liver using commercial ELISA kit, and the lipopolysaccharide (LPS) in serum with chromogenic end-point tachypheus amebocyte lysate. Compared with the HFD-induced group, Jlus66 treatment significantly decreased the malondialdehyde (MDA) level in the serum (p < 0.05). Additionally, Jlus66 significantly enhanced the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in the liver and serum (p < 0.05). Jlus66 administration also reduced the levels of tumor necrosis factor (TNF-α) and interleukin-6 (IL-6), and inversely increased the interleukin-10 (IL-10) level in serum (p < 0.05). Intestinal flora analysis results showed that Jlus66 can improve intestinal flora structure by increasing the abundance of gram-positive flora such as Firmicutes, and decreasing gram-negative flora such as Bacteroidetes, Proteobacteria, and Fusobacteria, and then reduced LPS concentration in the serum. So we concluded that Jlus66 can improve NAFLD by modulating the intestinal flora and followed reduction of oxidative stress (OxS) and inflammation.
Collapse
Affiliation(s)
- Wei Wang
- College of Food Science and Engineering, Jilin UniversityChangchunChina
- Jilin Provincial People's HospitalChangchunChina
| | - Qian Li
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Wenhui Chai
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Chunyan Sun
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Yuan Yuan
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Xinyu Wang
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Huiqin Liu
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin UniversityChangchunChina
| |
Collapse
|
45
|
Frost F, Storck LJ, Kacprowski T, Gärtner S, Rühlemann M, Bang C, Franke A, Völker U, Aghdassi AA, Steveling A, Mayerle J, Weiss FU, Homuth G, Lerch MM. A structured weight loss program increases gut microbiota phylogenetic diversity and reduces levels of Collinsella in obese type 2 diabetics: A pilot study. PLoS One 2019; 14:e0219489. [PMID: 31318902 PMCID: PMC6638920 DOI: 10.1371/journal.pone.0219489] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 06/23/2019] [Indexed: 12/17/2022] Open
Abstract
The global obesity epidemic constitutes a major cause of morbidity and mortality challenging public health care systems worldwide. Thus, a better understanding of its pathophysiology and the development of novel therapeutic options are urgently needed. Recently, alterations of the intestinal microbiome in the obese have been discussed as a promoting factor in the pathophysiology of obesity and as a contributing factor to related diseases such as type 2 diabetes and metabolic syndrome. The present pilot study investigated the effect of a structured weight loss program on fecal microbiota in obese type 2 diabetics. Twelve study subjects received a low-calorie formula diet for six weeks, followed by a nine week food reintroduction and stabilization period. Fecal microbiota were determined by 16S rRNA gene sequencing of stool samples at baseline, after six weeks and at the end of the study after fifteen weeks. All study subjects lost weight continuously throughout the program. Changes in fecal microbiota were most pronounced after six weeks of low-calorie formula diet, but reverted partially until the end of the study. However, the gut microbiota phylogenetic diversity increased persistently. The abundance of Collinsella, which has previously been associated with atherosclerosis, decreased significantly during the weight loss program. This study underlines the impact of dietary changes on the intestinal microbiome and further demonstrates the beneficial effects of weight loss on gut microbiota. Trial registration: ClinicalTrials.gov NCT02970838.
Collapse
Affiliation(s)
- Fabian Frost
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Lena J. Storck
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
- Department of Medicine, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Tim Kacprowski
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Simone Gärtner
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Malte Rühlemann
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Ali A. Aghdassi
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Antje Steveling
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Frank U. Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Markus M. Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
- * E-mail:
| |
Collapse
|
46
|
Basal Diet Determined Long-Term Composition of the Gut Microbiome and Mouse Phenotype to a Greater Extent than Fecal Microbiome Transfer from Lean or Obese Human Donors. Nutrients 2019; 11:nu11071630. [PMID: 31319545 PMCID: PMC6682898 DOI: 10.3390/nu11071630] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 12/23/2022] Open
Abstract
The Western dietary pattern can alter the gut microbiome and cause obesity and metabolic disorders. To examine the interactions between diet, the microbiome, and obesity, we transplanted gut microbiota from lean or obese human donors into mice fed one of three diets for 22 weeks: (1) a control AIN93G diet; (2) the total Western diet (TWD), which mimics the American diet; or (3) a 45% high-fat diet-induced obesity (DIO) diet. We hypothesized that a fecal microbiome transfer (FMT) from obese donors would lead to an obese phenotype and aberrant glucose metabolism in recipient mice that would be exacerbated by consumption of the TWD or DIO diets. Prior to the FMT, the native microbiome was depleted using an established broad-spectrum antibiotic protocol. Interestingly, the human donor body type microbiome did not significantly affect final body weight or body composition in mice fed any of the experimental diets. Beta diversity analysis and linear discriminant analysis with effect size (LEfSe) showed that mice that received an FMT from obese donors had a significantly different microbiome compared to mice that received an FMT from lean donors. However, after 22 weeks, diet influenced the microbiome composition irrespective of donor body type, suggesting that diet is a key variable in the shaping of the gut microbiome after FMT.
Collapse
|
47
|
Gu Y, Liu C, Zheng N, Jia W, Zhang W, Li H. Metabolic and Gut Microbial Characterization of Obesity-Prone Mice under a High-Fat Diet. J Proteome Res 2019; 18:1703-1714. [PMID: 30793608 DOI: 10.1021/acs.jproteome.8b00945] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is characterized with high heterogeneity due to genetic abnormality, energy imbalance, gut dysbiosis, or a combination of all three. Obesity-prone (OP) and -resistant (OR) phenotypes are frequently observed in rodents, even in those given a high-fat diet (HFD). However, the underlying mechanisms are largely unknown. Male C57BL/6J mice were fed with chow or a HFD for 8 weeks. OP and OR mice were defined based on body weight gain, and integrated serum metabolic and gut microbial profiling was performed by the gas chromatography-mass spectroscopy-based metabolomic sequencing and pyrosequencing of 16S rDNA of cecum contents. A total of 60 differential metabolites were identified in comparisons among Con, OP, and OR groups, in which 27 were OP-related. These differential metabolites are mainly involved in glycolysis, lipids, and amino acids metabolism and the TCA cycle. Meanwhile, OP mice had a distinct profile in gut microbiota compared to those of OR or Con mice, which showed a reduced ratio of Firmicutes to Bacteroidetes and increased Proteobacteria. Moreover, the gut microbial alteration of OP mice was correlated with the changes of the key serum metabolites. OP-enriched Parasutterella from the Proteobacteria phylum correlated to most of metabolites, suggesting that it was essential in obesity. OP mice are distinct in metabolic and gut microbial profiles, and OP-related metabolites and bacteria are of significance for understanding obesity development.
Collapse
Affiliation(s)
- Yu Gu
- Institute of Interdisciplinary Integrative Medicine Research , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Can Liu
- Department of Biochemistry and Molecular Biology , Bengbu Medical College , Anhui Province 233030 , China
| | - Ningning Zheng
- Institute of Interdisciplinary Integrative Medicine Research , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Wei Jia
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine , Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233 , China.,University of Hawaii Cancer Center , Honolulu , Hawaii 96813 , United States
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China.,Department of Phytochemistry, College of Pharmacy , Second Military Medical University , Shanghai 200433 , China
| | - Houkai Li
- Institute of Interdisciplinary Integrative Medicine Research , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| |
Collapse
|
48
|
Wang Z, Lam K, Hu J, Ge S, Zhou A, Zheng B, Zeng S, Lin S. Chlorogenic acid alleviates obesity and modulates gut microbiota in high-fat-fed mice. Food Sci Nutr 2019; 7:579-588. [PMID: 30847137 PMCID: PMC6392816 DOI: 10.1002/fsn3.868] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 01/05/2023] Open
Abstract
To evaluate the anti-obesity effects of chlorogenic acid (CGA), the mice were fed a high-fat diet (HFD) upon chlorogenic acid treatment for 6 weeks. The results showed administration of chlorogenic acid (150 mg per kg per day) remarkably promoted body loss, reduced lipid levels in plasma and altered mRNA expression of lipogenesis and lipolysis related genes in adipose tissue. Moreover, chlorogenic acid also reversed the HFD-induced gut microbiota dysbiosis, including significantly inhibiting the growth of Desulfovibrionaceae, Ruminococcaceae, Lachnospiraceae, Erysipelotrichaceae, and raising the growth of Bacteroidaceae, Lactobacillaceae. Overall, the amelioration of HFD-induced gut microbiota dysbiosis by chlorogenic acid may contribute, at least partially, to its beneficial effects on ameliorating HFD-induced obesity.
Collapse
Affiliation(s)
- Zhengyu Wang
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special StarchFujian Agriculture and Forestry UniversityFuzhouChina
| | - Ka‐Lung Lam
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong S.A.R.China
| | - Jiamiao Hu
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special StarchFujian Agriculture and Forestry UniversityFuzhouChina
| | - Shenghan Ge
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special StarchFujian Agriculture and Forestry UniversityFuzhouChina
| | - Arong Zhou
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special StarchFujian Agriculture and Forestry UniversityFuzhouChina
| | - Baodong Zheng
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special StarchFujian Agriculture and Forestry UniversityFuzhouChina
| | - Shaoxiao Zeng
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
| | - Shaoling Lin
- College of Food ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special StarchFujian Agriculture and Forestry UniversityFuzhouChina
| |
Collapse
|
49
|
Calorie restriction and its impact on gut microbial composition and global metabolism. Front Med 2018; 12:634-644. [PMID: 30446879 DOI: 10.1007/s11684-018-0670-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/27/2018] [Indexed: 02/08/2023]
Abstract
Calorie restriction (CR) is a dietary regimen that reduces calorie intake without incurring malnutrition or a reduction in essential nutrients. It has long been recognized as a natural strategy for promoting health, extending longevity, and prevents the development of metabolic and age-related diseases. In the present review, we focus on the general effect of CR on gut microbiota composition and global metabolism. We also propose mechanisms for its beneficial effect. Results showed that probiotic and butyrate-producing microbes increased their relative abundance, whereas proinflammatory strains exhibited suppressed relative abundance following CR. Analyses of the gut microbial and host metabolisms revealed that most host microbial co-metabolites were changed due to CR. Examples of dramatic CR-induced changes in host metabolism included a decrease in the rate of lipid biosynthesis and an increase in the rates of fatty acid catabolism, β-oxidation, glycogenolysis, and gluconeogenesis. The observed phenotypes and the further verification of the direct link between gut microbiota and metabolome may benefit patients that are at risk for developing metabolic disease. Thus, improved gut microbiota composition and metabolome are potential biomarkers for determining the effectiveness of dietary interventions for age-related and metabolic diseases.
Collapse
|
50
|
Telle-Hansen VH, Holven KB, Ulven SM. Impact of a Healthy Dietary Pattern on Gut Microbiota and Systemic Inflammation in Humans. Nutrients 2018; 10:E1783. [PMID: 30453534 PMCID: PMC6267105 DOI: 10.3390/nu10111783] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota have recently been suggested to play a part in low-grade systemic inflammation, which is considered a key risk factor for cardiometabolic disorders. Diet is known to affect gut microbiota; however, the effects of diet and dietary components on gut microbiota and inflammation are not fully understood. In the present review, we summarize recent research on human dietary intervention studies, investigating the effects of healthy diets or dietary components on gut microbiota and systemic inflammation. We included 18 studies that reported how different dietary components altered gut microbiota composition, short-chain fatty acid levels, and/or inflammatory markers. However, the heterogeneity among the intervention studies makes it difficult to conclude whether diets or dietary components affect gut microbiota homeostasis and inflammation. More appropriately designed studies are needed to better understand the effects of diet on the gut microbiota, systemic inflammation, and risk of cardiometabolic disorders.
Collapse
Affiliation(s)
- Vibeke H Telle-Hansen
- Faculty of Health Sciences, Oslo Metropolitan University, P.O. Box 4, St. Olavsplass, 0130 Oslo, Norway.
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046, Blindern, 0317 Oslo, Norway.
- National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Rikshospitalet, P.O. Box 4950, Nydalen, 0424 Oslo, Norway.
| | - Stine M Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046, Blindern, 0317 Oslo, Norway.
| |
Collapse
|