101
|
Abstract
The aim of this review is to provide an overview of how person-specific interactions between diet and the gut microbiota could play a role in affecting diet-induced weight loss responses. The highly person-specific gut microbiota, which is shaped by our diet, secretes digestive enzymes and molecules that affect digestion in the colon. Therefore, weight loss responses could in part depend on personal colonic fermentation responses, which affect energy extraction of food and production of microbial metabolites, such as short-chain fatty acids (SCFAs), which exert various effects on host metabolism. Colonic fermentation is the net result of the complex interplay between availability of dietary substrates, the functional capacity of the gut microbiome and environmental (abiotic) factors in the gut such as pH and transit time. While animal studies have demonstrated that the gut microbiota can causally affect obesity, causal and mechanistic evidence from human studies is still largely lacking. However, recent human studies have proposed that the baseline gut microbiota composition may predict diet-induced weight loss-responses. In particular, individuals characterised by high relative abundance of Prevotella have been found to lose more weight on diets rich in dietary fibre compared to individuals with low Prevotella abundance. Although harnessing of personal diet-microbiota interactions holds promise for more personalised nutrition and obesity management strategies to improve human health, there is currently insufficient evidence to unequivocally link the gut microbiota and weight loss in human subjects. To move the field forward, a greater understanding of the mechanistic underpinnings of personal diet-microbiota interactions is needed.
Collapse
|
102
|
Liu H, Liu H, Liu C, Shang M, Wei T, Yin P. Gut Microbiome and the Role of Metabolites in the Study of Graves’ Disease. Front Mol Biosci 2022; 9:841223. [PMID: 35252357 PMCID: PMC8889015 DOI: 10.3389/fmolb.2022.841223] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Graves’ disease (GD) is an autoimmune thyroid disease (AITD), which is one of the most common organ-specific autoimmune disorders with an increasing prevalence worldwide. But the etiology of GD is still unclear. A growing number of studies show correlations between gut microbiota and GD. The dysbiosis of gut microbiota may be the reason for the development of GD by modulating the immune system. Metabolites act as mediators or modulators between gut microbiota and thyroid. The purpose of this review is to summarize the correlations between gut microbiota, microbial metabolites and GD. Challenges in the future study are also discussed. The combination of microbiome and metabolome may provide new insight for the study and put forward the diagnosis, treatment, prevention of GD in the future.
Collapse
Affiliation(s)
- Haihua Liu
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Huiying Liu
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Chang Liu
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Mengxue Shang
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Tianfu Wei
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Peiyuan Yin
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Peiyuan Yin, ,
| |
Collapse
|
103
|
The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging). Nutrients 2022; 14:nu14040747. [PMID: 35215397 PMCID: PMC8879781 DOI: 10.3390/nu14040747] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/29/2022] [Accepted: 02/07/2022] [Indexed: 12/23/2022] Open
Abstract
Aging is typically accompanied by biological and physiological changes that alter cellular functions. Two of the most predominant phenomena in aging include chronic low-grade inflammation (inflammaging) and changes in the gut microbiota composition (dysbiosis). Although a direct causal relationship has not been established, many studies have reported significant reductions in inflammation during aging through well-maintained gut health and microbial balance. Prebiotics and probiotics are known to support gut health and can be easily incorporated into the daily diet. Unfortunately, few studies specifically focus on their significance in reducing inflammation during aging. Therefore, this review summarizes the scientific evidence of the potential roles of probiotics and two types of prebiotics, resistant starch and resistant proteins, in later age. Studies have demonstrated that the oral consumption of bacteria that may contribute to anti-inflammatory response, such as Bifidobacterium spp., Akkermansia munichipilla, and Faecalis praunitzii, contributes significantly to the suppression of pro-inflammatory markers in elderly humans and aged animals. Colonic fermentation of resistant starch and proteins also demonstrates anti-inflammatory activity owing to the production of butyrate and an improvement in the gut microbiota composition. Collectively, probiotics, resistant starch, and resistant proteins have the potential to promote healthy aging.
Collapse
|
104
|
DeMartino P, Johnston EA, Petersen KS, Kris-Etherton PM, Cockburn DW. Additional Resistant Starch from One Potato Side Dish per Day Alters the Gut Microbiota but Not Fecal Short-Chain Fatty Acid Concentrations. Nutrients 2022; 14:nu14030721. [PMID: 35277080 PMCID: PMC8840755 DOI: 10.3390/nu14030721] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 01/11/2023] Open
Abstract
The composition of the gut microbiota and their metabolites are associated with cardiometabolic health and disease risk. Intake of dietary fibers, including resistant starch (RS), has been shown to favorably affect the health of the gut microbiome. The aim of this research was to measure changes in the gut microbiota and fecal short-chain fatty acids as part of a randomized, crossover supplemental feeding study. Fifty participants (68% female, aged 40 ± 13 years, BMI 24.5 ± 3.6 kg/m2) completed this study. Potato dishes (POT) contained more RS than refined grain dishes (REF) (POT: 1.31% wet basis (95% CI: 0.94, 1.71); REF: 0.73% wet basis (95% CI: 0.34, 1.14); p = 0.03). Overall, potato dish consumption decreased alpha diversity, but beta diversity was not impacted. Potato dish consumption was found to increase the abundance of Hungatella xylanolytica, as well as that of the butyrate producing Roseburia faecis, though fecal butyrate levels were unchanged. Intake of one potato-based side dish per day resulted in modest changes in gut microbiota composition and diversity, compared to isocaloric intake of refined grains in healthy adults. Studies examining foods naturally higher in RS are needed to understand microbiota changes in response to dietary intake of RS and associated health effects.
Collapse
Affiliation(s)
- Peter DeMartino
- Department of Food Science, Pennsylvania State University, University Park, PA 16802, USA;
| | - Emily A. Johnston
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA; (E.A.J.); (K.S.P.); (P.M.K.-E.)
| | - Kristina S. Petersen
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA; (E.A.J.); (K.S.P.); (P.M.K.-E.)
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Penny M. Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA; (E.A.J.); (K.S.P.); (P.M.K.-E.)
| | - Darrell W. Cockburn
- Department of Food Science, Pennsylvania State University, University Park, PA 16802, USA;
- Correspondence: ; Tel.: +1-814-863-2950
| |
Collapse
|
105
|
Jiang R, Wang Y, Liu J, Wu Z, Wang D, Deng Q, Yang C, Zhou Q. Gut microbiota is involved in the antidepressant effects of adipose-derived mesenchymal stem cells in chronic social defeat stress mouse model. Psychopharmacology (Berl) 2022; 239:533-549. [PMID: 34981181 DOI: 10.1007/s00213-021-06037-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/23/2021] [Indexed: 10/19/2022]
Abstract
RATIONALE Growing evidence supports the role of microbiota in regulating gut-brain interactions and, thus, contributing to the pathogenesis of depression and the antidepressant actions. Adipose-derived mesenchymal stem cells (ADSCs), as important members of the stem cell family, were demonstrated to alleviate depression behaviors. However, the role of gut microbiota in ADSCs alleviating depression in chronic social defeat stress (CSDS) model is unknown. OBJECTIVES To examine the effects of ADSCs on depression symptoms and detect the changes in the composition of gut microbiota. RESULTS We found that ADSCs administration significantly ameliorated CSDS-induced depression behaviors, which was accompanied by alteration in the gut microbiota. The principal co-ordinates analysis (PCoA) results showed that there was a significant difference between the gut microbiota among the groups. Remarkably, receiver operating characteristic (ROC) curves revealed that order Micrococcales, order Rhizobiales and species Bacteroides acidifaciens are potentially important biomarkers for the antidepressant effects of ADSCs in CSDS model. CONCLUSIONS ADSCs are effective in treating depression behaviors in CSDS model, which might be partly due to the regulation of abnormal composition of gut microbiota. Thus, ADSCs offer a promising therapeutic strategy for treating depression in patients.
Collapse
Affiliation(s)
- Riyue Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junbi Liu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
106
|
Li C, Hu Y. New definition of resistant starch types from the gut microbiota perspectives - a review. Crit Rev Food Sci Nutr 2022; 63:6412-6422. [PMID: 35075962 DOI: 10.1080/10408398.2022.2031101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Current definition of resistant starch (RS) types is largely based on their interactions with digestive enzymes from human upper gastrointestinal tract. However, this is frequently inadequate to reflect their effects on the gut microbiota, which is an important mechanism for RS to fulfill its function to improve human health. Distinct shifts of gut microbiota compositions and alterations of fermented metabolites could be resulted by the consumption of RS from the same type. This review summarized these defects from the current definitions of RS types, while more importantly proposed pioneering concepts for new definitions of RS types from the gut microbiota perspectives. New RS types considered the aspects of RS fermentation rate, fermentation end products, specificity toward gut microbiota and shifts of gut microbiota caused by the consumption of RS. These definitions were depending on the known outcomes from RS-gut microbiota interactions. The application of new RS types in understanding the complex RS-gut microbiota interactions and promoting human health should be focused in the future.
Collapse
Affiliation(s)
- Cheng Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yiming Hu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
107
|
Dietary Alaska Pollock Protein Attenuates the Experimental Colitis Induced by Dextran Sulfate Sodium via Regulation of Gut Microbiota and Its Metabolites in Mice. Metabolites 2022; 12:metabo12010044. [PMID: 35050166 PMCID: PMC8779829 DOI: 10.3390/metabo12010044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/07/2023] Open
Abstract
Protein derived from fish has not only nutritional properties but also health-promoting properties. Few studies have examined the effect of dietary Alaska pollock protein (APP) on the anticolitis effect reported to be associated with metabolic syndrome (MetS). This study investigated the effect of APP intake on colitis symptoms, gut microbiota, and its metabolites in the experimental colitis mouse model induced by dextran sulfate sodium (DSS). Male C57BL/6J mice were divided into three groups: (1) DSS-untreated mice fed an American Institute of Nutrition (AIN) 93G diet (protein source is casein), (2) DSS-treated mice fed an AIN93G diet, and (3) DSS-treated mice fed an APP diet. After the mice were fed the diets for 21 days, experimental colitis was induced by three cycles of 2% DSS administration for 5 days followed by washouts over the course of 5 days. APP-reduced body weight loss increased the disease activity index, and elevated spleen weight and alleviated colon length shortening and colonic tissue damage. Furthermore, APP altered the structure and composition of the microbiota and short-chain fatty acids in feces. Since APP intake alleviates experimental colitis induced by DSS administration through alterations in the gut microbiota and its metabolites, we deduced that APP would inhibit MetS progression via colitis suppression.
Collapse
|
108
|
Dietary ferulic acid and vanillic acid on inflammation, gut barrier function and growth performance in lipopolysaccharide-challenged piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:144-152. [PMID: 34977384 PMCID: PMC8683658 DOI: 10.1016/j.aninu.2021.06.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/23/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022]
Abstract
Ferulic acid (FA) and vanillic acid (VA) are considered as major phenolic metabolites of cyanidin 3-glucoside, a polyphenol that widely exists in plants that possess a protective effect against oxidative stress and inflammation in our previous study. This study aimed to investigate the effect of FA and VA on inflammation, gut barrier function, and growth performance in a weaned piglet model challenged with lipopolysaccharide (LPS). Thirty-six piglets (PIC 337 × C48, 28 d of age) were randomly allocated into 3 treatments with 6 replicate pens (2 piglets per pen). They were fed with a basal diet or a diet containing 4,000 mg/kg of FA or VA. Dietary supplementation of VA significantly increased average daily gain (ADG) (P < 0.05). Both FA and VA decreased serum levels of thiobarbituric acid reactive substances (TBARS), interlukin (IL)-1β, IL-2, IL-6, and tumor necrosis factor (TNF)-α (P < 0.05), and enhanced the expression of tight junction protein oclaudin (P < 0.05). Analysis of gut microbiota indicated that both FA and VA increased the Firmicutes/Bacteroidetes ratio alongside reducing the relative abundance of the Prevotellaceae family including Prevotella 9 and Prevotella 2 genera, but enriched the Lachoiraceaea family including the Lachnospiraceae FCS020 group (P < 0.05). Moreover, VA reduced the relative abundance of Prevotella 7 and Prevotella 1 but enriched Lachnospira, Eubacterium eligens group, and Eubacterium xylanophilum group (P < 0.05), while FA showed a limited effect on these genera. The results demonstrated that both VA and FA could alleviate inflammation and oxidative stress, but only VA has a significant positive effect on the growth performance of LPS-challenged piglets potentially through modulating gut microbiota.
Collapse
|
109
|
Parker J, O'Brien C, Hawrelak J. A narrative review of the role of gastrointestinal dysbiosis in the pathogenesis of polycystic ovary syndrome. Obstet Gynecol Sci 2022; 65:14-28. [PMID: 34958733 PMCID: PMC8784943 DOI: 10.5468/ogs.21185] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Diet-induced gastrointestinal dysbiosis has been hypothesized to play a significant role in stimulating an increase in gastrointestinal permeability and activating systemic inflammation in women with polycystic ovary syndrome (PCOS). We reviewed the current proof-of-concept studies on the proposed mechanism of dysbiosis in the pathogenesis of PCOS. A literature search was performed to identify articles on changes in the intestinal microbiome (dysbiosis) and increased intestinal mucosal permeability involving lipopolysaccharide (LPS), LPS-binding protein (LPS-BP), and zonulin. We also searched for systematic reviews and meta-analyses that synthesized the results of studies on the therapeutic effects of prebiotics, probiotics, or synbiotics in women with PCOS. Our search was confined to human studies between 2012 and 2021 using the PubMed, Scopus, and Cochrane databases. Thirty-one studies met the inclusion criteria (14 microbiota, 1 LPS, 1 LPS-BP, 1 LPS and LPS-BP, 5 zonulin, 9 systematic reviews). Our analysis revealed that most studies reported reduced alpha diversity and dysbiosis in women with PCOS. Preliminary studies suggest that LPS, LPS-BP, and zonulin may be involved in the pathophysiology of increased intestinal permeability. Treatment of PCOS with prebiotics, probiotics, and synbiotics appears to have a range of beneficial effects on metabolic and biochemical profiles. This review highlights the need for continued research into the pathophysiological mechanisms of dysbiosis and the clinical efficacy of prebiotics, probiotics, and synbiotics in women with PCOS.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia
| | - Claire O'Brien
- Faculty of Science and Technology, University of Canberra, Canberra, Australia
| | - Jason Hawrelak
- College of Health and Medicine, University of Tasmania, Tasmania, Australia
| |
Collapse
|
110
|
Abstract
The prevalence of overweight and obesity has reached epidemic proportions globally over the past few decades. The search for new management approaches continues and among them, targeting the gut microbiota can be envisioned. To date, numerous data showed the involvement of the gut microbes in the regulation and control of host metabolism. There are also increasing evidences highlighting the interactions between environmental factors, intrinsic factors, gut microbiota, and metabolic diseases. Diet emerges as the most relevant factor influencing the gut microbiome. Eating habits, as well as short-term consumption of specific diets, alter the gut microbiota composition. Moreover, nutritional disorders are associated with changes of the gut microbiota composition and/or function, as shown in obesity or type 2 diabetic patients versus healthy lean subjects. Targeting the gut microbiota for improving metabolic health appears as a new approach to manage obesity and cardio-metabolic risk. In this review, we have detailed the results of human interventions targeting the gut microbiome by prebiotic supplementation, prebiotics being defined as "substrates that are selectively utilized by the host microorganisms conferring a health benefit." If the potential benefit of this approach is obvious in preclinical models, the efficacy of prebiotics in humans is less reproducible. The inter-individual variability of response to dietary intervention can be dependent on the gut microbiota and we summarized the basal gut microbiota characteristics driving the metabolic response to dieting, prebiotic and dietary fiber intervention in the context of obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
111
|
Elgarten CW, Tanes C, Lee JJ, Danziger-Isakov LA, Grimley MS, Green M, Michaels MG, Barnum JL, Ardura MI, Auletta JJ, Blumenstock J, Seif AE, Bittinger KL, Fisher BT. Early stool microbiome and metabolome signatures in pediatric patients undergoing allogeneic hematopoietic cell transplantation. Pediatr Blood Cancer 2022; 69:e29384. [PMID: 34709713 PMCID: PMC8629955 DOI: 10.1002/pbc.29384] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/25/2021] [Accepted: 09/16/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The contribution of the gastrointestinal tract microbiome to outcomes after allogeneic hematopoietic cell transplantation (HCT) is increasingly recognized. Investigations of larger pediatric cohorts aimed at defining the microbiome state and associated metabolic patterns pretransplant are needed. METHODS We sought to describe the pretransplant stool microbiome in pediatric allogenic HCT patients at four centers. We performed shotgun metagenomic sequencing and untargeted metabolic profiling on pretransplant stool samples. Samples were compared with normal age-matched controls and by clinical characteristics. We then explored associations between stool microbiome measurements and metabolite concentrations. RESULTS We profiled stool samples from 88 pediatric allogeneic HCT patients, a median of 4 days before transplant. Pretransplant stool samples differed from healthy controls based on indices of alpha diversity and in the proportional abundance of specific taxa and bacterial genes. Relative to stool from healthy patients, samples from HCT patients had decreased proportion of Bacteroides, Ruminococcaeae, and genes involved in butyrate production, but were enriched for gammaproteobacterial species. No systematic differences in stool microbiome or metabolomic profiles by age, transplant indication, or hospital were noted. Stool metabolites demonstrated strong correlations with microbiome composition. DISCUSSION Stool samples from pediatric allogeneic HCT patients demonstrate substantial dysbiosis early in the transplant course. As microbiome disruptions associate with adverse transplant outcomes, pediatric-specific analyses examining longitudinal microbiome and metabolome changes are imperative to identify causal associations and to inform rational design of interventions.
Collapse
Affiliation(s)
- Caitlin W. Elgarten
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia,Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia
| | - Ceylan Tanes
- PennCHOP Microbiome Program, Children’s Hospital of Philadelphia Research Institute
| | - Jung-jin Lee
- PennCHOP Microbiome Program, Children’s Hospital of Philadelphia Research Institute
| | - Lara A. Danziger-Isakov
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children’s Hospital, Medical Center and University of Cincinnati
| | - Michael S. Grimley
- Division of Bone Marrow Transplantation and Immune Deficiency, Department of Pediatrics, Cincinnati Children’s Hospital
| | - Michael Green
- Division of Infectious Diseases, UPMC Children’s Hospital of Pittsburgh
| | | | - Jessie L. Barnum
- Division of Blood and Marrow Transplantation, UPMC Children’s Hospital of Pittsburgh
| | | | - Jeffery J. Auletta
- Division of Infectious Diseases, Nationwide Children’s Hospital,Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital,National Marrow Donor Program/Be The Match
| | - Jesse Blumenstock
- Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia
| | - Alix E. Seif
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia,Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia
| | - Kyle L. Bittinger
- PennCHOP Microbiome Program, Children’s Hospital of Philadelphia Research Institute,Division of Gastroenterology, Department of Pediatrics, Children’s Hospital of Philadelphia
| | - Brian T. Fisher
- Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia,Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia
| |
Collapse
|
112
|
Mutignani M, Penagini R, Gargari G, Guglielmetti S, Cintolo M, Airoldi A, Leone P, Carnevali P, Ciafardini C, Petrocelli G, Mascaretti F, Oreggia B, Dioscoridi L, Cavalcoli F, Primignani M, Pugliese F, Bertuccio P, Soru P, Magistro C, Ferrari G, Speciani MC, Bonato G, Bini M, Cantù P, Caprioli F, Vangeli M, Forti E, Mazza S, Tosetti G, Bonzi R, Vecchi M, La Vecchia C, Rossi M. Blood Bacterial DNA Load and Profiling Differ in Colorectal Cancer Patients Compared to Tumor-Free Controls. Cancers (Basel) 2021; 13:6363. [PMID: 34944982 PMCID: PMC8699505 DOI: 10.3390/cancers13246363] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022] Open
Abstract
Inflammation and immunity are linked to intestinal adenoma (IA) and colorectal cancer (CRC) development. The gut microbiota is associated with CRC risk. Epithelial barrier dysfunction can occur, possibly leading to increased intestinal permeability in CRC patients. We conducted a case-control study including 100 incident histologically confirmed CRC cases, and 100 IA and 100 healthy subjects, matched to cases by center, sex and age. We performed 16S rRNA gene analysis of blood and applied conditional logistic regression. Further analyses were based on negative binomial distribution normalization and Random Forest algorithm. We found an overrepresentation of blood 16S rRNA gene copies in colon cancer as compared to tumor-free controls. For high levels of gene copies, community diversity was higher in colon cancer cases than controls. Bacterial taxa and operational taxonomic unit abundances were different between groups and were able to predict CRC with an accuracy of 0.70. Our data support the hypothesis of a higher passage of bacteria from gastrointestinal tract to bloodstream in colon cancer. This result can be applied on non-invasive diagnostic tests for colon cancer control.
Collapse
Affiliation(s)
- Massimiliano Mutignani
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Roberto Penagini
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Giorgio Gargari
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, 20133 Milan, Italy; (G.G.); (S.G.)
| | - Simone Guglielmetti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, 20133 Milan, Italy; (G.G.); (S.G.)
| | - Marcello Cintolo
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Aldo Airoldi
- Hepatology and Gastroenterology Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (A.A.); (M.V.)
| | - Pierfrancesco Leone
- General Surgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (P.L.); (B.O.)
| | - Pietro Carnevali
- Division of Minimally-Invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (P.C.); (C.M.); (G.F.)
| | - Clorinda Ciafardini
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
| | - Giulio Petrocelli
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
- Associazione Nazionale Operatori Tecniche Endoscopiche (ANOTE), 80061 Massa Lubrense, Italy
| | - Federica Mascaretti
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
- Department of Clinical Science and Community Health, University of Milan, 20133 Milan, Italy; (P.B.); (M.C.S.); (R.B.); (C.L.V.)
| | - Barbara Oreggia
- General Surgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (P.L.); (B.O.)
| | - Lorenzo Dioscoridi
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Federica Cavalcoli
- Diagnostic and Therapeutic Endoscopy Unit, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy;
| | - Massimo Primignani
- Division of Gastroenterology and Hepatology, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (G.T.)
| | - Francesco Pugliese
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Paola Bertuccio
- Department of Clinical Science and Community Health, University of Milan, 20133 Milan, Italy; (P.B.); (M.C.S.); (R.B.); (C.L.V.)
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Pietro Soru
- Division of Endoscopy, IRCCS Istituto Europeo di Oncologia, 20141 Milan, Italy;
| | - Carmelo Magistro
- Division of Minimally-Invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (P.C.); (C.M.); (G.F.)
| | - Giovanni Ferrari
- Division of Minimally-Invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (P.C.); (C.M.); (G.F.)
| | - Michela C. Speciani
- Department of Clinical Science and Community Health, University of Milan, 20133 Milan, Italy; (P.B.); (M.C.S.); (R.B.); (C.L.V.)
| | - Giulia Bonato
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Marta Bini
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Paolo Cantù
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Marcello Vangeli
- Hepatology and Gastroenterology Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (A.A.); (M.V.)
| | - Edoardo Forti
- Digestive and Interventional Endoscopy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (M.M.); (M.C.); (G.P.); (L.D.); (F.P.); (G.B.); (M.B.); (E.F.)
| | - Stefano Mazza
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
- Gastroenterology and Digestive Endoscopy Unit, ASST Cremona, Cremona 26100, Italy
| | - Giulia Tosetti
- Division of Gastroenterology and Hepatology, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (G.T.)
| | - Rossella Bonzi
- Department of Clinical Science and Community Health, University of Milan, 20133 Milan, Italy; (P.B.); (M.C.S.); (R.B.); (C.L.V.)
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.P.); (C.C.); (F.M.); (P.C.); (F.C.); (S.M.); (M.V.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Science and Community Health, University of Milan, 20133 Milan, Italy; (P.B.); (M.C.S.); (R.B.); (C.L.V.)
| | - Marta Rossi
- Department of Clinical Science and Community Health, University of Milan, 20133 Milan, Italy; (P.B.); (M.C.S.); (R.B.); (C.L.V.)
| |
Collapse
|
113
|
Kumari M, Singh P, Nataraj BH, Kokkiligadda A, Naithani H, Azmal Ali S, Behare PV, Nagpal R. Fostering next-generation probiotics in human gut by targeted dietary modulation: An emerging perspective. Food Res Int 2021; 150:110716. [PMID: 34865747 DOI: 10.1016/j.foodres.2021.110716] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/07/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022]
Abstract
Emerging evidence and an in-depth understanding of the microbiome have helped in identifying beneficial commensals and their therapeutic potentials. Specific commensal taxa/ strains of the human gut microbiome have been positively associated with human health and recently termed as next-generation probiotics (NGPs). Of these, Akkermansia muciniphila, Ruminococcus bromii, Faecalibacterium prausnitzii, Anaerobutyricum hallii, and Roseburia intestinalis are the five most relevant gut-derived NGPs that have demonstrated therapeutic potential in managing metabolic diseases. Specific and natural dietary interventions can modulate the abundance and activity of these beneficial bacteria in the gut. Hence, the understanding of targeted stimulation of specific NGP by specific probiotic-targeted diets (PTD) is indispensable for the rational application of their combination. The supplementation of NGP with its specific PTD will help the strain(s) to compete with harmful microbes and acquire its niche. This combination would enhance the effectiveness of NGPs to be used as "live biotherapeutic products" or food nutraceuticals. Under the current milieu, we review various PTDs that influence the abundance of specific potential NGPs, and contemplates potential interactions between diet, microbes, and their effects on host health. Taking into account the study mentioned, we propose that combining NGPs will provide an alternate solution for developing the new diet in conjunction with PTD.
Collapse
Affiliation(s)
- Manorama Kumari
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Parul Singh
- Proteomics and Cell Biology Lab, Animal Biotechnology Center, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Basavaprabhu H Nataraj
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Anusha Kokkiligadda
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Harshita Naithani
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Syed Azmal Ali
- Proteomics and Cell Biology Lab, Animal Biotechnology Center, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Pradip V Behare
- Technofunctional Starters Lab, National Collection of Dairy Cultures, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India.
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
114
|
Liu R, Peng C, Jing D, Xiao Y, Zhu W, Zhao S, Zhang J, Chen X, Li J. Biomarkers of Gut Microbiota in Chronic Spontaneous Urticaria and Symptomatic Dermographism. Front Cell Infect Microbiol 2021; 11:703126. [PMID: 34858864 PMCID: PMC8630658 DOI: 10.3389/fcimb.2021.703126] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/22/2021] [Indexed: 01/17/2023] Open
Abstract
Background Chronic urticaria (CU) is a chronic inflammatory skin disease associated with Th2 immune response. The two most common subtypes of CU, i.e., chronic spontaneous urticaria and symptomatic dermographism (CSD), often coexist. However, the pathogenesis of CSD is still unclear. Gut microbiota plays an important role in immune-related inflammatory diseases. The purpose of this study was to explore the correlation between gut microbiota and CSD. Methods A case-control study was conducted on CSD patients as well as gender- and age-matched normal controls (NCs). The 16S ribosomal DNA sequencing of fecal samples was used to detect the gut microbiota of all subjects. QPCR was used to further verify the species with differences between the two groups. Results The alpha diversity of gut microbiota decreased in CSD patients, accompanied by significant changes of the structure of gut microbiota. Subdoligranulum and Ruminococcus bromii decreased significantly in CSD patients and had a potential diagnostic value for CSD according to receiver operating characteristic curve (ROC) analysis. Enterobacteriaceae and Klebsiella were found to be positively correlated with the duration of CSD, while Clostridium disporicum was positively correlated with the dermatology life quality index (DLQI). Conclusions The gut microbiota of CSD patients is imbalanced. Subdoligranulum and Ruminococcus bromii are the gut microbiota biomarkers in CSD.
Collapse
Affiliation(s)
- Runqiu Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, The First People's Hospital of Yancheng, Yancheng, China.,Department of Dermatology, The Fourth Affiliated Hospital of Nantong University, Nantong, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Danrong Jing
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Yangjian Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Jianglin Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| |
Collapse
|
115
|
Wolter M, Grant ET, Boudaud M, Steimle A, Pereira GV, Martens EC, Desai MS. Leveraging diet to engineer the gut microbiome. Nat Rev Gastroenterol Hepatol 2021; 18:885-902. [PMID: 34580480 DOI: 10.1038/s41575-021-00512-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 12/12/2022]
Abstract
Autoimmune diseases, including inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis, have distinct clinical presentations but share underlying patterns of gut microbiome perturbation and intestinal barrier dysfunction. Their potentially common microbial drivers advocate for treatment strategies aimed at restoring appropriate microbiome function, but individual variation in host factors makes a uniform approach unlikely. In this Perspective, we consolidate knowledge on diet-microbiome interactions in local inflammation, gut microbiota imbalance and host immune dysregulation. By understanding and incorporating the effects of individual dietary components on microbial metabolic output and host physiology, we examine the potential for diet-based therapies for autoimmune disease prevention and treatment. We also discuss tools targeting the gut microbiome, such as faecal microbiota transplantation, probiotics and orthogonal niche engineering, which could be optimized using custom dietary interventions. These approaches highlight paths towards leveraging diet for precise engineering of the gut microbiome at a time of increasing autoimmune disease.
Collapse
Affiliation(s)
- Mathis Wolter
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Alex Steimle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | | - Eric C Martens
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg. .,Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
116
|
Arp CG, Correa MJ, Ferrero C. Resistant starches: A smart alternative for the development of functional bread and other starch-based foods. Food Hydrocoll 2021. [DOI: 10.1016/j.foodhyd.2021.106949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
117
|
The Role Transition of Dietary Species Richness in Modulating the Gut Microbial Assembly and Postweaning Performance of a Generalist Herbivore. mSystems 2021; 6:e0097921. [PMID: 34726492 PMCID: PMC8562480 DOI: 10.1128/msystems.00979-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
When facing a food shortage, generalist herbivores can respond by expanding their dietary species richness (DSR) to maximize energy collection, regardless of whether forages are preferred or not. Higher DSR usually indicates higher nutrient adequacy and better health. However, the high-DSR diet containing a large proportion of preferred species or a large proportion of less-preferred species means different things to an animal. It is still unknown how different shift patterns in DSR would affect distinctly the performance of animals via altering gut microbiota. We examined the gut microbial composition, diversity, community assembly processes, and performance of a generalist herbivore, Lasiopodomys brandtii, in a feeding experiment with increased levels of simulated DSR shifting from preferred plant species to less preferred ones. We found the survival rate and body growth of Brandt's voles showed a dome-shaped association with DSR: species performance increased initially with the increase of preferred plant species but declined with the increase of less-preferred food items. Several microbial taxa and functions closely related to the metabolism of amino acids and short-chain fatty acids also showed a dome-shaped association with DSR, which is consistent with the observation of performance change. However, the alpha diversities of gut microbiota increased linearly with DSR. The null model and phylogenetic analysis suggested that stochastic processes dominate at low DSR diets, whereas deterministic processes prevail at high DSR diets. These results suggest that the role of DSR in regulating animal performance by gut microbiota depends on the number of preferred forage items. IMPORTANCE The plant species diversity varies greatly under the influence of both climate change and human disturbance, which may negatively affect the productivity as well as the variability of organisms (e.g., small herbivores) at the next trophic level. It is still unknown how gut microbiota of small herbivores respond to such changes in dietary species richness. Our manipulative food experiment revealed that dietary species richness can affect the composition, functions, and community assembly of gut microbiota of Brandt's vole in a nonlinear way. Given the fast-growing interest in therapeutic diets to treat dysbiosis and to improve health conditions, our study highlights the need to consider not just the variety of consumed food but also the principles of rational nutrition.
Collapse
|
118
|
Intestinal Microbiota as a Contributor to Chronic Inflammation and Its Potential Modifications. Nutrients 2021; 13:nu13113839. [PMID: 34836095 PMCID: PMC8618457 DOI: 10.3390/nu13113839] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is a crucial factor in maintaining homeostasis. The presence of commensal microorganisms leads to the stimulation of the immune system and its maturation. In turn, dysbiosis with an impaired intestinal barrier leads to accelerated contact of microbiota with the host’s immune cells. Microbial structural parts, i.e., pathogen-associated molecular patterns (PAMPs), such as flagellin (FLG), peptidoglycan (PGN), lipoteichoic acid (LTA), and lipopolysaccharide (LPS), induce inflammation via activation of pattern recognition receptors. Microbial metabolites can also develop chronic low-grade inflammation, which is the cause of many metabolic diseases. This article aims to systematize information on the influence of microbiota on chronic inflammation and the benefits of microbiota modification through dietary changes, prebiotics, and probiotic intake. Scientific research indicates that the modification of the microbiota in various disease states can reduce inflammation and improve the metabolic profile. However, since there is no pattern for a healthy microbiota, there is no optimal way to modify it. The methods of influencing microbiota should be adapted to the type of dysbiosis. Although there are studies on the microbiota and its effects on inflammation, this subject is still relatively unknown, and more research is needed in this area.
Collapse
|
119
|
Lin D, Hu B, Li P, Zhao Y, Xu Y, Wu D. Roles of the intestinal microbiota and microbial metabolites in acute GVHD. Exp Hematol Oncol 2021; 10:49. [PMID: 34706782 PMCID: PMC8555140 DOI: 10.1186/s40164-021-00240-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/25/2021] [Indexed: 01/02/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is one of the most curative strategies for the treatment of many hematologic malignancies and diseases. However, acute graft-versus-host disease (GVHD) limits the success of allo-HSCT. The prevention and treatment of acute GVHD is the key issue for improving the efficacy of allo-HSCT and has become a research hotspot. The intestine is the primary organ targeted by acute GVHD, and the intestinal microbiota is critical for maintaining the homeostasis of the intestinal microenvironment and the immune response. Many studies have demonstrated the close association between the intestinal microbiota and the pathogenesis of acute GVHD. Furthermore, dysbiosis of the microbiota, which manifests as alterations in the diversity and composition of the intestinal microbiota, and alterations of microbial metabolites are pronounced in acute GVHD and associated with poor patient prognosis. The microbiota interacts with the host directly via microbial surface antigens or microbiota-derived metabolites to regulate intestinal homeostasis and the immune response. Therefore, intervention strategies targeting the intestinal microbiota, including antibiotics, prebiotics, probiotics, postbiotics and fecal microbiota transplantation (FMT), are potential new treatment options for acute GVHD. In this review, we discuss the alterations and roles of the intestinal microbiota and its metabolites in acute GVHD, as well as interventions targeting microbiota for the prevention and treatment of acute GVHD.
Collapse
Affiliation(s)
- Dandan Lin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, People's Republic of China
| | - Bo Hu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, People's Republic of China
| | - Pengfei Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, People's Republic of China
| | - Ye Zhao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, People's Republic of China.
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
120
|
Wang S, De Paepe K, Van de Wiele T, Fu X, Yuan Y, Zhang B, Huang Q. Starch Microspheres Entrapped with Chitosan Delay In Vitro Fecal Fermentation and Regulate Human Gut Microbiota Composition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12323-12332. [PMID: 34623811 DOI: 10.1021/acs.jafc.1c04039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A slow dietary fiber fermentation rate is desirable to obtain a steady metabolite release and even distribution throughout the entire colon, ensuring to meet the energy needs in the distal colon. In this study, we prepared starch-entrapped microspheres with a variable chitosan-to-starch ratio by means of electrospraying and investigated the fermentability by human fecal microbiota in an in vitro batch system. Starch encapsulation reduced microbial gas production and the concentration of short-chain fatty acids. Butyrate production, in particular, gradually decreased with increasing chitosan proportions. Moreover, the starch and chitosan composites induced a synergistic effect on the gut microbiota composition. Roseburia, Lachnospiraceae, and Clostridiales were promoted by all of the microspheres, and the abundance of the aforementioned health-promoting taxa reached a maximum in chitosan/starch microspheres with a 1:6 (w/w) ratio. Our findings highlight the possible benefits of rationally designing functional foods targeting functional and taxonomic gut microbiota modulation.
Collapse
Affiliation(s)
- Shaokang Wang
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Kim De Paepe
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Xiong Fu
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, (111 Center), Guangzhou 510640, China
| | - Yang Yuan
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, China
| | - Bin Zhang
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, (111 Center), Guangzhou 510640, China
| | - Qiang Huang
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, (111 Center), Guangzhou 510640, China
| |
Collapse
|
121
|
Fujiwara H. Crosstalk Between Intestinal Microbiota Derived Metabolites and Tissues in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2021; 12:703298. [PMID: 34512627 PMCID: PMC8429959 DOI: 10.3389/fimmu.2021.703298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an evidence based- cellular immunotherapy for hematological malignancies. Immune reactions not only promote graft-versus-tumor effects that kill hematological malignant cells but also graft-versus-host disease (GVHD) that is the primary complication characterized by systemic organ damages consisting of T-cells and antigen presenting cells (APCs) activation. GVHD has long been recognized as an immunological reaction that requires an immunosuppressive treatment targeting immune cells. However immune suppression cannot always prevent GVHD or effectively treat it once it has developed. Recent studies using high-throughput sequencing technology investigated the impact of microbial flora on GVHD and provided profound insights of the mechanism of GVHD other than immune cells. Allo-HSCT affects the intestinal microbiota and microbiome-metabolome axis that can alter intestinal homeostasis and the severity of experimental GVHD. This axis can potentially be manipulated via dietary intervention or metabolites produced by intestinal bacteria affected post-allo-HSCT. In this review, we discuss the mechanism of experimental GVHD regulation by the complex microbial community-metabolites-host tissue axis. Furthermore, we summarize the major findings of microbiome-based immunotherapeutic approaches that protect tissues from experimental GVHD. Understanding the complex relationships between gut microbiota-metabolites-host tissues axis provides crucial insight into the pathogenesis of GVHD and advances the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
122
|
Müller B, Rasmusson AJ, Just D, Jayarathna S, Moazzami A, Novicic ZK, Cunningham JL. Fecal Short-Chain Fatty Acid Ratios as Related to Gastrointestinal and Depressive Symptoms in Young Adults. Psychosom Med 2021; 83:693-699. [PMID: 34267089 PMCID: PMC8428857 DOI: 10.1097/psy.0000000000000965] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 04/14/2021] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Short-chain fatty acids (SCFAs) are produced by the gut microbiota and may reflect health. Gut symptoms are common in individuals with depressive disorders, and recent data indicate relationships between gut microbiota and psychiatric health. We aimed to investigate potential associations between SCFAs and self-reported depressive and gut symptoms in young adults. METHODS Fecal samples from 164 individuals (125 were patients with psychiatric disorders: mean [standard deviation] age = 21.9 [2.6] years, 14% men; 39 nonpsychiatric controls: age = 28.5 [9.5] years, 38% men) were analyzed for the SCFA acetate, butyrate, and propionate by nuclear magnetic resonance spectroscopy. We then compared SCFA ratios with dimensional measures of self-reported depressive and gut symptoms. RESULTS Depressive symptoms showed a positive association with acetate levels (ρ = 0.235, p = .003) and negative associations with both butyrate (ρ = -0.195, p = .014) and propionate levels (ρ = -0.201, p = .009) in relation to total SCFA levels. Furthermore, symptoms of diarrhea showed positive associations with acetate (ρ = 0.217, p = .010) and negative associations with propionate in relation to total SCFA levels (ρ = 0.229, p = 0-007). Cluster analysis revealed a heterogeneous pattern where shifts in SCFA ratios were observed in individuals with elevated levels of depressive symptoms, elevated levels of gut symptoms, or both. CONCLUSIONS Shifts in SCFAs are associated with both depressive symptoms and gut symptoms in young adults and may have of relevance for treatment.
Collapse
|
123
|
Okada Y, Park SY, Wilkens LR, Maskarinec G, Shvetsov YB, Haiman C, Le Marchand L. White Rice Consumption and Risk for Colorectal Cancer among Japanese Americans: The Multiethnic Cohort Study. J Epidemiol 2021; 33:170-176. [PMID: 34380917 PMCID: PMC9939926 DOI: 10.2188/jea.je20200611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND White rice is a staple food for Japanese, a population at high risk for colorectal cancer (CRC). We investigated the association between white rice intake and CRC among Japanese Americans in the Multiethnic Cohort (MEC) study. METHODS The Multiethnic Cohort Study is a prospective study established in Hawaii and California in 1993-1996. Usual dietary intake was assessed by a validated quantitative food frequency questionnaire at baseline. Cox proportional hazards models were used to compute hazard ratios (HR) and 95% confidence intervals (CI) for quartiles of intake and to perform trend tests across sex-specific quartiles with adjustment for relevant confounders. RESULTS We identified 1,553 invasive CRC cases among 49,136 Japanese Americans (23,595 men and 25,541 women) during a mean follow-up of 19 years. White rice consumption was not associated with overall CRC incidence in men (p-trend = 0.11) or women (p-trend = 0.56). After excluding participants with a history of diabetes, the inverse associations were significant for CRC (p-trend = 0.03, HR for quartile 4 (Q4) vs. 1 = 0.81; 95% CI: 0.64-1.03) and tumors of the distal colon (p-trend = 0.006, HR for Q4 vs. Q1: 0.66; 0.44-0.99) among men but not women. CONCLUSIONS White rice consumption was not associated with an increased risk of overall CRC among Japanese Americans. An inverse association was observed with risk of CRC and distal colon cancer in men without a history of diabetes.
Collapse
Affiliation(s)
- Yuito Okada
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center.,Office of Public Health Studies, University of Hawaii
| | - Song-Yi Park
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center
| | - Lynne R Wilkens
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center
| | - Gertraud Maskarinec
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center
| | - Yurii B Shvetsov
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California
| | - Loïc Le Marchand
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center
| |
Collapse
|
124
|
Adesina PA, Isayama K, Sitolo GC, Yamamoto Y, Suzuki T. Propionate and Dietary Fermentable Fibers Upregulate Intestinal Heat Shock protein70 in Intestinal Caco-2 Cells and Mouse Colon. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8460-8470. [PMID: 34291640 DOI: 10.1021/acs.jafc.1c03036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Short-chain fatty acids (SCFAs), including propionate, are major metabolites of intestinal microorganisms and play an essential role in regulating intestinal epithelial integrity. Heat shock proteins (HSPs) promote cellular homeostasis under physiological and stressed conditions. This study aimed to investigate the regulation of intestinal HSP70 by propionate in human intestinal Caco-2 cells and the colon of fermentable dietary fiber (DF)-fed mice and germ-free mice. The results showed that propionate increased Hspa1a (HSP70 mRNA) level in Caco-2 cells, upregulated HSP70 protein, and phosphorylation of heat shock factor 1; however, the latter two were reduced by mitogen-activated protein kinases and the mechanistic target of rapamycin inhibitors. Feeding fermentable DFs, such as guar gum (GG) and partially hydrolyzed GG, increased both cecal SCFAs and colonic HSP70 expression, both of which were reduced in germ-free mice than in specific-pathogen-free mice. Collectively, the propionate-induced HSP70 expression was shown to be possibly involved in intestinal homeostasis.
Collapse
Affiliation(s)
- Precious Adedayo Adesina
- Graduate School of Biosphere Science, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Kana Isayama
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Gertrude Cynthia Sitolo
- Graduate School of Biosphere Science, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
- Department of Physics and Biochemical Sciences, Malawi University of Business and Applied Sciences, Private Bag 303, Chichiri Blantyre 3, Malawi
| | - Yoshinari Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Takuya Suzuki
- Graduate School of Biosphere Science, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| |
Collapse
|
125
|
Gual-Grau A, Guirro M, Crescenti A, Boqué N, Arola L. In vitro fermentability of a broad range of natural ingredients by fecal microbiota from lean and obese individuals: potential health benefits. Int J Food Sci Nutr 2021; 73:195-209. [PMID: 34294012 DOI: 10.1080/09637486.2021.1954144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The prevalence of obesity and related complications is continuously increasing while the gut microbiota might have a significant role to address this challenge. In this context, the food industry generates large amounts of residues that could be likely revalorised as functional ingredients. Hence, we evaluated the fermentability of food skins, husks, shells, trimming residues, mosses and mushrooms, which were subjected to in vitro fermentation with faecal microbiota from lean and obese adults. We demonstrated for the first time that pumpkin skin is highly fermented by human faecal microbiota showing pH-lowering effects and promoting gas and SCFA production. Furthermore, brewers' spent grain generated an inulin-like SCFA profile after microbial fermentation, whereas Irish moss, plum skin, quinoa husk and mushrooms, including Armillaria mellea and Boletus edulis, showed high fermentation rates. Remarkably, although propionate production was significantly higher in obese individuals, the fermentability of the ingredients was similar between lean and obese conditions.
Collapse
Affiliation(s)
- Andreu Gual-Grau
- Department of Biochemistry and Biotechnology, Nutrigenomics Research Group, Universitat Rovira i Virgili, Tarragona, Spain
| | - Maria Guirro
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (Joint Unit Eurecat-Universitat Rovira i Virgili), Unique Scientific and Technical Infrastructures (ICTS), Reus, Spain
| | - Anna Crescenti
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus, Spain
| | - Noemí Boqué
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus, Spain
| | - Lluís Arola
- Department of Biochemistry and Biotechnology, Nutrigenomics Research Group, Universitat Rovira i Virgili, Tarragona, Spain
| |
Collapse
|
126
|
Bongiovanni T, Yin MOL, Heaney L. The Athlete and Gut Microbiome: Short-chain Fatty Acids as Potential Ergogenic Aids for Exercise and Training. Int J Sports Med 2021; 42:1143-1158. [PMID: 34256388 DOI: 10.1055/a-1524-2095] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Short-chain fatty acids (SCFAs) are metabolites produced in the gut via microbial fermentation of dietary fibers referred to as microbiota-accessible carbohydrates (MACs). Acetate, propionate, and butyrate have been observed to regulate host dietary nutrient metabolism, energy balance, and local and systemic immune functions. In vitro and in vivo experiments have shown links between the presence of bacteria-derived SCFAs and host health through the blunting of inflammatory processes, as well as purported protection from the development of illness associated with respiratory infections. This bank of evidence suggests that SCFAs could be beneficial to enhance the athlete's immunity, as well as act to improve exercise recovery via anti-inflammatory activity and to provide additional energy substrates for exercise performance. However, the mechanistic basis and applied evidence for these relationships in humans have yet to be fully established. In this narrative review, we explore the existing knowledge of SCFA synthesis and the functional importance of the gut microbiome composition to induce SCFA production. Further, changes in gut microbiota associated with exercise and various dietary MACs are described. Finally, we provide suggestions for future research and practical applications, including how these metabolites could be manipulated through dietary fiber intake to optimize immunity and energy metabolism.
Collapse
Affiliation(s)
| | | | - Liam Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
127
|
Dietary Fiber Hierarchical Specificity: the Missing Link for Predictable and Strong Shifts in Gut Bacterial Communities. mBio 2021; 12:e0102821. [PMID: 34182773 PMCID: PMC8262931 DOI: 10.1128/mbio.01028-21] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Most dietary fibers used to shape the gut microbiota present different and unpredictable responses, presumably due to the diverse microbial communities of people. Recently, we proposed that fibers can be classified in a hierarchical way where fibers of high specificity (i.e., structurally complex and utilized by a narrow group of gut bacteria) could have more similar interindividual responses than those of low specificity (i.e., structurally simple and utilized by many gut bacteria). To test this hypothesis, we evaluated microbiota fermentation of fibers tentatively classified as low (fructooligosaccharides), low-to-intermediate (type 2 resistant starch), intermediate (pectin), and high (insoluble β-1,3-glucan) specificity, utilizing fecal inoculum from distinct subjects, regarding interindividual similarity/dissimilarity in fiber responses. Individual shifts in target bacteria (as determined by linear discriminant analysis) confirmed that divergent fiber responses occur when utilizing both of the low-specificity dietary fibers, but fibers of intermediate and high specificity lead to more similar responses across subjects in support of targeted bacteria. The high-specificity insoluble β-glucan promoted a large increase of the target bacteria (from 0.3 to 16.5% average for Anaerostipes sp. and 2.5 to 17.9% average for Bacteroides uniformis), which were associated with increases in ratios of related metabolites (butyrate and propionate, respectively) in every microbial community in which these bacteria were present. Also, high-specificity dietary fibers promoted more dramatic changes in microbial community structure than low-specificity ones relative to the initial microbial communities.
Collapse
|
128
|
The Microbiota and the Gut-Brain Axis in Controlling Food Intake and Energy Homeostasis. Int J Mol Sci 2021; 22:ijms22115830. [PMID: 34072450 PMCID: PMC8198395 DOI: 10.3390/ijms22115830] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity currently represents a major societal and health challenge worldwide. Its prevalence has reached epidemic proportions and trends continue to rise, reflecting the need for more effective preventive measures. Hypothalamic circuits that control energy homeostasis in response to food intake are interesting targets for body-weight management, for example, through interventions that reinforce the gut-to-brain nutrient signalling, whose malfunction contributes to obesity. Gut microbiota-diet interactions might interfere in nutrient sensing and signalling from the gut to the brain, where the information is processed to control energy homeostasis. This gut microbiota-brain crosstalk is mediated by metabolites, mainly short chain fatty acids, secondary bile acids or amino acids-derived metabolites and subcellular bacterial components. These activate gut-endocrine and/or neural-mediated pathways or pass to systemic circulation and then reach the brain. Feeding time and dietary composition are the main drivers of the gut microbiota structure and function. Therefore, aberrant feeding patterns or unhealthy diets might alter gut microbiota-diet interactions and modify nutrient availability and/or microbial ligands transmitting information from the gut to the brain in response to food intake, thus impairing energy homeostasis. Herein, we update the scientific evidence supporting that gut microbiota is a source of novel dietary and non-dietary biological products that may beneficially regulate gut-to-brain communication and, thus, improve metabolic health. Additionally, we evaluate how the feeding time and dietary composition modulate the gut microbiota and, thereby, the intraluminal availability of these biological products with potential effects on energy homeostasis. The review also identifies knowledge gaps and the advances required to clinically apply microbiome-based strategies to improve the gut-brain axis function and, thus, combat obesity.
Collapse
|
129
|
Murga-Garrido SM, Hong Q, Cross TWL, Hutchison ER, Han J, Thomas SP, Vivas EI, Denu J, Ceschin DG, Tang ZZ, Rey FE. Gut microbiome variation modulates the effects of dietary fiber on host metabolism. MICROBIOME 2021; 9:117. [PMID: 34016169 PMCID: PMC8138933 DOI: 10.1186/s40168-021-01061-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/24/2021] [Indexed: 05/11/2023]
Abstract
BACKGROUND There is general consensus that consumption of dietary fermentable fiber improves cardiometabolic health, in part by promoting mutualistic microbes and by increasing production of beneficial metabolites in the distal gut. However, human studies have reported variations in the observed benefits among individuals consuming the same fiber. Several factors likely contribute to this variation, including host genetic and gut microbial differences. We hypothesized that gut microbial metabolism of dietary fiber represents an important and differential factor that modulates how dietary fiber impacts the host. RESULTS We examined genetically identical gnotobiotic mice harboring two distinct complex gut microbial communities and exposed to four isocaloric diets, each containing different fibers: (i) cellulose, (ii) inulin, (iii) pectin, (iv) a mix of 5 fermentable fibers (assorted fiber). Gut microbiome analysis showed that each transplanted community preserved a core of common taxa across diets that differentiated it from the other community, but there were variations in richness and bacterial taxa abundance within each community among the different diet treatments. Host epigenetic, transcriptional, and metabolomic analyses revealed diet-directed differences between animals colonized with the two communities, including variation in amino acids and lipid pathways that were associated with divergent health outcomes. CONCLUSION This study demonstrates that interindividual variation in the gut microbiome is causally linked to differential effects of dietary fiber on host metabolic phenotypes and suggests that a one-fits-all fiber supplementation approach to promote health is unlikely to elicit consistent effects across individuals. Overall, the presented results underscore the importance of microbe-diet interactions on host metabolism and suggest that gut microbes modulate dietary fiber efficacy. Video abstract.
Collapse
Affiliation(s)
- Sofia M Murga-Garrido
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Dr., Madison, WI, 53706, USA
- PECEM (MD/PhD), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - Qilin Hong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Tzu-Wen L Cross
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Dr., Madison, WI, 53706, USA
- Present Address: Department of Nutrition Science, Purdue University, 700 W. State Street, Stone Hall 205, West Lafayette, IN, 47907, USA
| | - Evan R Hutchison
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Dr., Madison, WI, 53706, USA
| | - Jessica Han
- Wisconsin Institute for Discovery, Madison, WI, USA
| | | | - Eugenio I Vivas
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Dr., Madison, WI, 53706, USA
| | - John Denu
- Wisconsin Institute for Discovery, Madison, WI, USA
| | - Danilo G Ceschin
- Unidad de Bioinformática Traslacional, Centro de Investigación en Medicina Traslacional Severo Amuchástegui, Instituto Universitario de Ciencias Biomédicas de Córdoba, Av. Naciones Unidas 420, 5000, Córdoba, CP, Argentina
| | - Zheng-Zheng Tang
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA.
- Wisconsin Institute for Discovery, Madison, WI, USA.
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Dr., Madison, WI, 53706, USA.
| |
Collapse
|
130
|
Jaagura M, Viiard E, Karu-Lavits K, Adamberg K. Low-carbohydrate high-fat weight reduction diet induces changes in human gut microbiota. Microbiologyopen 2021; 10:e1194. [PMID: 34180599 PMCID: PMC8123914 DOI: 10.1002/mbo3.1194] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 12/23/2022] Open
Abstract
Obesity has become a major public health problem in recent decades. More effective interventions may result from a better understanding of microbiota alterations caused by weight loss and diet. Our objectives were (a) to calculate the fiber composition of a specially designed low‐calorie weight loss diet (WLD), and (b) to evaluate changes in the composition of gut microbiota and improvements in health characteristics during WLD. A total of 19 overweight/obese participants were assigned to 20%–40% reduced calories low‐carbohydrate high‐fat diet for four weeks. Protein and fat content in the composed diet was 1.5 times higher compared to that in the average diet of the normal weight reference group, while carbohydrate content was 2 times lower. Food consumption data were obtained from the assigned meals. Microbial composition was analyzed before and after WLD intervention from two sequential samples by 16S rRNA gene sequencing. During WLD, body mass index (BMI) was reduced on average 2.5 ± 0.6 kg/m2 and stool frequency was normalized. The assigned diet induced significant changes in fecal microbiota. The abundance of bile‐resistant bacteria (Alistipes, Odoribacter splanchnicus), Ruminococcus bicirculans, Butyricimonas, and Enterobacteriaceae increased. Importantly, abundance of bacteria often associated with inflammation such as Collinsella and Dorea decreased in parallel with a decrease in BMI. Also, we observed a reduction in bifidobacteria, which can be attributed to the relatively low consumption of grains. In conclusion, weight loss results in significant alteration of the microbial community structure.
Collapse
Affiliation(s)
- Madis Jaagura
- Center of Food and Fermentation Technologies, Tallinn, 12618, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, 12618, Estonia
| | - Ene Viiard
- Center of Food and Fermentation Technologies, Tallinn, 12618, Estonia
| | | | - Kaarel Adamberg
- Center of Food and Fermentation Technologies, Tallinn, 12618, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, 12618, Estonia
| |
Collapse
|
131
|
Prebiotics protect against acute graft-versus-host disease and preserve the gut microbiota in stem cell transplantation. Blood Adv 2021; 4:4607-4617. [PMID: 32991720 DOI: 10.1182/bloodadvances.2020002604] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Therefore, management of aGVHD is important for successful transplantation. Mucosal damage and alteration of the gut microbiota after allo-HSCT are key factors in the development of aGVHD. We conducted a prospective study to evaluate the ability of prebiotics, which can alleviate mucosal damage and manipulate the gut microbiota, to mitigate posttransplantation complications, including aGVHD. Resistant starch (RS) and a commercially available prebiotics mixture, GFO, were administered to allo-HSCT recipients from pretransplantation conditioning to day 28 after allo-HSCT. Prebiotic intake mitigated mucosal injury and reduced the incidence of all aGVHD grades combined and of aGVHD grades 2 to 4. The cumulative incidence of skin aGVHD was markedly decreased by prebiotics intake. Furthermore, the gut microbial diversity was well maintained and butyrate-producing bacterial population were preserved by prebiotics intake. In addition, the posttransplantation fecal butyrate concentration was maintained or increased more frequently in the prebiotics group. These observations indicate that prebiotic intake may be an effective strategy for preventing aGVHD in allo-HSCT, thereby improving treatment outcomes and the clinical utility of stem cell transplantation approaches. This study was registered on the University Hospital Medical Information Network (UMIN) clinical trials registry (https://www.umin.ac.jp/ctr/index.htm) as #UMIN000027563.
Collapse
|
132
|
Cronin P, Joyce SA, O’Toole PW, O’Connor EM. Dietary Fibre Modulates the Gut Microbiota. Nutrients 2021; 13:nu13051655. [PMID: 34068353 PMCID: PMC8153313 DOI: 10.3390/nu13051655] [Citation(s) in RCA: 342] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Dietary fibre has long been established as a nutritionally important, health-promoting food ingredient. Modern dietary practices have seen a significant reduction in fibre consumption compared with ancestral habits. This is related to the emergence of low-fibre “Western diets” associated with industrialised nations, and is linked to an increased prevalence of gut diseases such as inflammatory bowel disease, obesity, type II diabetes mellitus and metabolic syndrome. The characteristic metabolic parameters of these individuals include insulin resistance, high fasting and postprandial glucose, as well as high plasma cholesterol, low-density lipoprotein (LDL) and high-density lipoprotein (HDL). Gut microbial signatures are also altered significantly in these cohorts, suggesting a causative link between diet, microbes and disease. Dietary fibre consumption has been hypothesised to reverse these changes through microbial fermentation and the subsequent production of short-chain fatty acids (SCFA), which improves glucose and lipid parameters in individuals who harbour diseases associated with dysfunctional metabolism. This review article examines how different types of dietary fibre can differentially alter glucose and lipid metabolism through changes in gut microbiota composition and function.
Collapse
Affiliation(s)
- Peter Cronin
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (S.A.J.); (P.W.O.)
| | - Susan A. Joyce
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (S.A.J.); (P.W.O.)
- School of Biochemistry and Cell Biology, University College Cork, T12 K8AF Cork, Ireland
| | - Paul W. O’Toole
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (S.A.J.); (P.W.O.)
- Department of Microbiology, University College Cork, T12 K8AF Cork, Ireland
| | - Eibhlís M. O’Connor
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (S.A.J.); (P.W.O.)
- Health Research Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Correspondence:
| |
Collapse
|
133
|
Liu H, Zhang M, Ma Q, Tian B, Nie C, Chen Z, Li J. Health beneficial effects of resistant starch on diabetes and obesity via regulation of gut microbiota: a review. Food Funct 2021; 11:5749-5767. [PMID: 32602874 DOI: 10.1039/d0fo00855a] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Resistant starch (RS) is well known to prevent type 2 diabetes mellitus (T2DM) and obesity. Recently, attention has been paid to gut microbiota which mediates the RS's impact on T2DM and obesity, while a mechanistic understanding of how RS prevents T2DM and obesity through gut microbiota is not clear yet. Therefore, this review aims at exploring the underlying mechanisms of it. RS prevents T2DM and obesity through gut microbiota by modifying selective microbial composition to produce starch-degrading enzymes, promoting the production of intestinal metabolites, and improving gut barrier function. Therefore, RS possessing good functional features can be used to increase the fiber content of healthier food. Furthermore, achieving highly selective effects on gut microbiota based on the slight differences of RS's chemical structure and focusing on the effects of RS on strain-levels are essential to manipulate the microbiota for human health.
Collapse
Affiliation(s)
- Huicui Liu
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Min Zhang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Qingyu Ma
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Baoming Tian
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Chenxi Nie
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Zhifei Chen
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Juxiu Li
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China.
| |
Collapse
|
134
|
Jiang W, Yu X, Kosik RO, Song Y, Qiao T, Tong J, Liu S, Fan S, Luo Q, Chai L, Lv Z, Li D. Gut Microbiota May Play a Significant Role in the Pathogenesis of Graves' Disease. Thyroid 2021; 31:810-820. [PMID: 33234057 PMCID: PMC8110022 DOI: 10.1089/thy.2020.0193] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Gut microbiota are considered to be intrinsic regulators of thyroid autoimmunity. We designed a cross-sectional study to examine the makeup and metabolic function of microbiota in Graves' disease (GD) patients, with the ultimate aim of offering new perspectives on the diagnosis and treatment of GD. Methods: The 16S ribosomal RNA (rRNA) V3-V4 DNA regions of microbiota were obtained from fecal samples collected from 45 GD patients and 59 controls. Microbial differences between the two groups were subsequently analyzed based on high-throughput sequencing. Results: Compared with controls, GD patients had reduced alpha diversity (p < 0.05). At the phylum level, GD patients had a significantly lower proportion of Firmicutes (p = 0.008) and a significantly higher proportion of Bacteroidetes (p = 0.002) compared with the controls. At the genus level, GD patients had greater numbers of Bacteroides and Lactobacillus, although fewer Blautia, [Eubacterium]_hallii_group, Anaerostipes, Collinsella, Dorea, unclassified_f_Peptostreptococcaceae, and [Ruminococcus]_torques_group than controls (all p < 0.05). Subgroup analysis of GD patients revealed that Lactobacillus may play a key role in the pathogenesis of autoimmune thyroid diseases. Nine distinct genera showed significant correlations with certain thyroid function tests. Functional prediction revealed that Blautia may be an important microbe in certain metabolic pathways that occur in the hyperthyroid state. In addition, linear discriminant analysis (LDA) and effect size (LEfSe) analysis showed that there were significant differences in the levels of 18 genera between GD patients and controls (LDA >3.0, all p < 0.05). A diagnostic model using the top nine genera had an area under the curve of 0.8109 [confidence interval: 0.7274-0.8945]. Conclusions: Intestinal microbiota are different in GD patients. The microbiota we identified offer an alternative noninvasive diagnostic methodology for GD. Microbiota may also play a role in thyroid autoimmunity, and future research is needed to further elucidate the role.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaqing Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Russell Oliver Kosik
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingchun Song
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tingting Qiao
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junyu Tong
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Simin Liu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Suyun Fan
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiong Luo
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Chai
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Address correspondence to: Zhongwei Lv, PhD, MD, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Dan Li
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Address correspondence to: Dan Li, PhD, MD, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
135
|
Teichmann J, Cockburn DW. In vitro Fermentation Reveals Changes in Butyrate Production Dependent on Resistant Starch Source and Microbiome Composition. Front Microbiol 2021; 12:640253. [PMID: 33995299 PMCID: PMC8117019 DOI: 10.3389/fmicb.2021.640253] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
One of the primary benefits associated with dietary resistant starch (RS) is the production of butyrate by the gut microbiome during fermentation of this fiber in the large intestine. The ability to degrade RS is a relatively rare trait among microbes in the gut, seemingly confined to only a few species, none of which are butyrate producing organisms. Thus, production of butyrate during RS fermentation requires a network of interactions between RS degraders and butyrate producers. This is further complicated by the fact that there are multiple types of RS that differ in their structural properties and impacts on the microbiome. Human dietary intervention trials with RS have shown increases in fecal butyrate levels at the population level but with individual to individual differences. This suggests that interindividual differences in microbiome composition dictate butyrate response, but the factors driving this are still unknown. Furthermore, it is unknown whether a lack of increase in butyrate production upon supplementation with one RS is indicative of a lack of butyrate production with any RS. To shed some light on these issues we have undertaken an in vitro fermentation approach in an attempt to mimic RS fermentation in the colon. Fecal samples from 10 individuals were used as the inoculum for fermentation with 10 different starch sources. Butyrate production was heterogeneous across both fecal inocula and starch source, suggesting that a given microbiome is best suited to produce butyrate only from a subset of RS sources that differs between individuals. Interestingly, neither the total amount of RS degraders nor butyrate producers seemed to be limiting for any individual, rather the membership of these sub-populations was more important. While none of the RS degrading organisms were correlated with butyrate levels, Ruminococcus bromii was strongly positively correlated with many of the most important butyrate producers in the gut, though total butyrate production was strongly influenced by factors such as pH and lactate levels. Together these results suggest that the membership of the RS degrader and butyrate producer communities rather than their abundances determine the RS sources that will increase butyrate levels for a given microbiome.
Collapse
Affiliation(s)
- June Teichmann
- Department of Food Science, The Pennsylvania State University, University Park, PA, United States
| | - Darrell W Cockburn
- Department of Food Science, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
136
|
Bacillus amyloliquefaciens ameliorates high-carbohydrate diet-induced metabolic phenotypes by restoration of intestinal acetate-producing bacteria in Nile Tilapia. Br J Nutr 2021; 127:653-665. [PMID: 33858522 DOI: 10.1017/s0007114521001318] [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] [Indexed: 11/06/2022]
Abstract
Poor utilisation efficiency of carbohydrate always leads to metabolic phenotypes in fish. The intestinal microbiota plays an important role in carbohydrate degradation. Whether the intestinal bacteria could alleviate high-carbohydrate diet (HCD)-induced metabolic phenotypes in fish remains unknown. Here, a strain affiliated to Bacillus amyloliquefaciens was isolated from the intestine of Nile tilapia. A basal diet (CON), HCD or HCD supplemented with B. amy SS1 (HCB) was used to feed fish for 10 weeks. The beneficial effects of B. amy SS1 on weight gain and protein accumulation were observed. Fasting glucose and lipid deposition were decreased in the HCB group compared with the HCD group. High-throughput sequencing showed that the abundance of acetate-producing bacteria was increased in the HCB group relative to the HCD group. Gas chromatographic analysis indicated that the concentration of intestinal acetate was increased dramatically in the HCB group compared with that in the HCD group. Glucagon-like peptide-1 was also increased in the intestine and serum of the HCB group. Thus, fish were fed with HCD, HCD supplemented with sodium acetate at 900 mg/kg (HLA), 1800 mg/kg (HMA) or 3600 mg/kg (HHA) diet for 8 weeks, and the HMA and HHA groups mirrored the effects of B. amy SS1. This study revealed that B. amy SS1 could alleviate the metabolic phenotypes caused by HCD by enriching acetate-producing bacteria in fish intestines. Regulating the intestinal microbiota and their metabolites might represent a powerful strategy for fish nutrition modulation and health maintenance in future.
Collapse
|
137
|
Saarenpää M, Roslund MI, Puhakka R, Grönroos M, Parajuli A, Hui N, Nurminen N, Laitinen OH, Hyöty H, Cinek O, Sinkkonen A, the ADELE Research Group. Do Rural Second Homes Shape Commensal Microbiota of Urban Dwellers? A Pilot Study among Urban Elderly in Finland. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073742. [PMID: 33918486 PMCID: PMC8038225 DOI: 10.3390/ijerph18073742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022]
Abstract
According to the hygiene and biodiversity hypotheses, increased hygiene levels and reduced contact with biodiversity can partially explain the high prevalence of immune-mediated diseases in developed countries. A disturbed commensal microbiota, especially in the gut, has been linked to multiple immune-mediated diseases. Previous studies imply that gut microbiota composition is associated with the everyday living environment and can be modified by increasing direct physical exposure to biodiverse materials. In this pilot study, the effects of rural-second-home tourism were investigated on the gut microbiota for the first time. Rural-second-home tourism, a popular form of outdoor recreation in Northern Europe, North America, and Russia, has the potential to alter the human microbiota by increasing exposure to nature and environmental microbes. The hypotheses were that the use of rural second homes is associated with differences in the gut microbiota and that the microbiota related to health benefits are more diverse or common among the rural-second-home users. Based on 16S rRNA Illumina MiSeq sequencing of stool samples from 10 urban elderly having access and 15 lacking access to a rural second home, the first hypothesis was supported: the use of rural second homes was found to be associated with lower gut microbiota diversity and RIG-I-like receptor signaling pathway levels. The second hypothesis was not supported: health-related microbiota were not more diverse or common among the second-home users. The current study encourages further research on the possible health outcomes or causes of the observed microbiological differences. Activities and diet during second-home visits, standard of equipment, surrounding environment, and length of the visits are all postulated to play a role in determining the effects of rural-second-home tourism on the gut microbiota.
Collapse
Affiliation(s)
- Mika Saarenpää
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
| | - Marja I. Roslund
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
| | - Riikka Puhakka
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
| | - Mira Grönroos
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
| | - Anirudra Parajuli
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
- Department of Medicine, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Nan Hui
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan RD. Minhang District, Shanghai 200240, China
| | - Noora Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland; (N.N.); (O.H.L.); (H.H.)
| | - Olli H. Laitinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland; (N.N.); (O.H.L.); (H.H.)
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland; (N.N.); (O.H.L.); (H.H.)
| | - Ondrej Cinek
- Second Faculty of Medicine, Charles University, V Úvalu 84, 150 06 Prague 5, Czech Republic;
| | - Aki Sinkkonen
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; (M.S.); (M.I.R.); (R.P.); (M.G.); (A.P.); (N.H.)
- Natural Resources Institute Finland, Itäinen Pitkäkatu 4 A, 20520 Turku, Finland
- Correspondence:
| | | |
Collapse
|
138
|
Henig I, Yehudai-Ofir D, Zuckerman T. The clinical role of the gut microbiome and fecal microbiota transplantation in allogeneic stem cell transplantation. Haematologica 2021; 106:933-946. [PMID: 33241674 PMCID: PMC8017815 DOI: 10.3324/haematol.2020.247395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 08/28/2020] [Indexed: 12/26/2022] Open
Abstract
Outcomes of allogeneic hematopoietic stem cell transplantation (allo- HSCT) have improved in the recent decade; however, infections and graft-versus-host disease remain two leading complications significantly contributing to early transplant-related mortality. In past years, the human intestinal microbial composition (microbiota) has been found to be associated with various disease states, including cancer, response to cancer immunotherapy and to modulate the gut innate and adaptive immune response. In the setting of allo-HSCT, the intestinal microbiota diversity and composition appear to have an impact on infection risk, mortality and overall survival. Microbial metabolites have been shown to contribute to the health and integrity of intestinal epithelial cells during inflammation, thus mitigating graft-versus-host disease in animal models. While the cause-andeffect relationship between the intestinal microbiota and transplant-associated complications has not yet been fully elucidated, the above findings have already resulted in the implementation of various interventions aiming to restore the intestinal microbiota diversity and composition. Among others, these interventions include the administration of fecal microbiota transplantation. The present review, based on published data, is intended to define the role of the latter approach in the setting of allo-HSCT.
Collapse
Affiliation(s)
- Israel Henig
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa
| | - Dana Yehudai-Ofir
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa
| | - Tsila Zuckerman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa.
| |
Collapse
|
139
|
Wang S, Dhital S, Wang K, Fu X, Zhang B, Huang Q. Side-by-side and exo-pitting degradation mechanism revealed from in vitro human fecal fermentation of granular starches. Carbohydr Polym 2021; 263:118003. [PMID: 33858585 DOI: 10.1016/j.carbpol.2021.118003] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022]
Abstract
The in vitro fecal fermentation characteristics and microbiota responses to A- and B-type polymorphic starches as model (whole) foods enriched with resistant starch was investigated. Marked difference in fermentation rate as well as microbial genera was observed during fermentation, the degradation pattern as well as structural evolution during fermentation was almost similar. The final butyrate concentrations of both HAMS and PS (ca. 38 mM) were significantly higher than that of WMS (23 mM) and NMS (33 mM), which was associated with the increase of the relative abundance of Roseburia, Blautia, and Lachnospiraceae. A-type polymorphic starches, on the other hand had remarkably faster fermentation rate and promoted Megamonas. X-ray diffraction and size-exclusion chromatography of residual starch during the fermentation course demonstrated the "side-by-side" fermentation pattern. Based on the structural changes observed, we conclude that in vitro fecal fermentation of starch granules predominantly controlled by the surface features rather than the molecular and supra-molecular structure.
Collapse
Affiliation(s)
- Shaokang Wang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, 510640, China; Sino-Singapore International Research Institute, Guangzhou, 510555, China
| | - Sushil Dhital
- Department of Chemical Engineering, Monash University, Clayton Campus, VIC, 3800, Australia
| | - Kai Wang
- School of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiong Fu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, 510640, China; Sino-Singapore International Research Institute, Guangzhou, 510555, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, 510640, China
| | - Bin Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, 510640, China; Sino-Singapore International Research Institute, Guangzhou, 510555, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, 510640, China.
| | - Qiang Huang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, 510640, China; Sino-Singapore International Research Institute, Guangzhou, 510555, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, 510640, China.
| |
Collapse
|
140
|
Xie Z, Ding L, Huang Q, Fu X, Liu F, Dhital S, Zhang B. In vitro colonic fermentation profiles and microbial responses of propionylated high-amylose maize starch by individual Bacteroides-dominated enterotype inocula. Food Res Int 2021; 144:110317. [PMID: 34053522 DOI: 10.1016/j.foodres.2021.110317] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/13/2021] [Accepted: 03/14/2021] [Indexed: 12/28/2022]
Abstract
The concept of "enterotype" has been proposed to differentiate the gut microbiota between individual humans, and different dominant bacteria utilize fiber substrates with different fermentation properties and microbial changes. In this study, we made propionylated high-amylose maize starch and investigated both in vitro fecal fermentation properties and microbial responses by individual Bacteroides-dominated enterotype inocula. Propionyl group substitution of HAMS did not significantly change gas production profiles, suggesting that the gas production during fermentation is independent of propionylation. The final concentration of released propionate significantly increased (10.26-12.60 mM) as a function of propionylation degree, suggesting that the introduced propionyl groups can increase the concentration of short-chain fatty acids (SCFA) during colonic fermentation. At the genus level, Bacteroides was obviously promoted for all donors with the final abundance in the range of 0.1-0.24, indicating that propionylated high-amylose maize starch changed the structure and abundance of microbiota compared to unmodified starch. Besides, the non-metric dimensional scoring (NMDS) plots showed that those changes were related to the initial microbiota composition. The results may offer useful information for the design of personalized food products and relevant therapies at least within Bacteroides-dominated enterotype.
Collapse
Affiliation(s)
- Zhuqing Xie
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Li Ding
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qiang Huang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China; SCUT-Zhuhai Institute of Modern Industrial Innovation, Zhuhai 519175, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| | - Xiong Fu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China; SCUT-Zhuhai Institute of Modern Industrial Innovation, Zhuhai 519175, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| | - Feitong Liu
- H&H Group Global Research and Technology Center, Guangzhou 510700, China
| | - Sushil Dhital
- Department of Chemical Engineering, Monash University, Clayton Campus, VIC 3800, Australia
| | - Bin Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China; SCUT-Zhuhai Institute of Modern Industrial Innovation, Zhuhai 519175, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| |
Collapse
|
141
|
Qin R, Wang J, Chao C, Yu J, Copeland L, Wang S, Wang S. RS5 Produced More Butyric Acid through Regulating the Microbial Community of Human Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3209-3218. [PMID: 33630575 DOI: 10.1021/acs.jafc.0c08187] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The objective of this research was to compare the in vitro fermentability of three resistant starches (RS2, RS3, and RS5). Structural analyses showed that there were small changes in the long- and short-range ordered structure of three RSs after fermentation by human gut microbiota. The fermentation of RSs by gut microbiota produced large amounts of short-chain fatty acids, with RS5 producing more butyric acid and RS3 producing more lactic acid. RS3 and RS5 decreased the pH of the fermentation culture to a greater extent compared with RS2. Moreover, RS5 increased significantly the relative abundance of Bifidobacterium, Dialister, Collinsella, Romboutsia, and Megamonas. The results suggested that the form of RS was the main factor affecting the physiological function of RS and that RS5, as a recently recognized form of resistant starch, could be a better functional ingredient to improve health compared with RS2 and RS3.
Collapse
Affiliation(s)
- Renbing Qin
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- School of Food Science and Technology Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Chen Chao
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- School of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jinglin Yu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Les Copeland
- Sydney Institute of Agriculture, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Shujun Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- School of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Shuo Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
142
|
Targeting the Gut Microbiome to Mitigate Immunotherapy-Induced Colitis in Cancer. Trends Cancer 2021; 7:583-593. [PMID: 33741313 DOI: 10.1016/j.trecan.2021.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have been a transformational advance in cancer therapy in the past decade. However, ICIs can produce immune-related adverse effects (irAEs), which can lead to both morbidity and premature termination of therapy. Recent studies suggest that the gut microbiota and its metabolites affect ICI efficacy and toxicity. Herein, we review such evidence in the context of ICI-induced colitis. In particular, the short-chain fatty acid butyrate, a microbial metabolite, has known protective effects on the intestine. We discuss how the use of dietary prebiotics, which can be metabolized by bacteria to produce butyrate, can be an intriguing new investigational approach to prevent ICI-associated colitis and lead to improved patient outcomes.
Collapse
|
143
|
High-Fiber, Whole-Food Dietary Intervention Alters the Human Gut Microbiome but Not Fecal Short-Chain Fatty Acids. mSystems 2021; 6:6/2/e00115-21. [PMID: 33727392 PMCID: PMC8546969 DOI: 10.1128/msystems.00115-21] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Dietary shifts can have a direct impact on the gut microbiome by preferentially selecting for microbes capable of utilizing the various dietary nutrients. The intake of dietary fiber has decreased precipitously in the last century, while consumption of processed foods has increased. Fiber, or microbiota-accessible carbohydrates (MACs), persist in the digestive tract and can be metabolized by specific bacteria encoding fiber-degrading enzymes. The digestion of MACs results in the accumulation of short-chain fatty acids (SCFAs) and other metabolic by-products that are critical to human health. Here, we implemented a 2-week dietary fiber intervention aiming for 40 to 50 g of fiber per day within the context of a course-based undergraduate research experience (CURE) (n = 20). By coupling shotgun metagenomic sequencing and targeted gas chromatography-mass spectrometry (GC-MS), we found that the dietary intervention significantly altered the composition of individual gut microbiomes, accounting for 8.3% of the longitudinal variability within subjects. Notably, microbial taxa that increased in relative abundance as a result of the diet change included known MAC degraders (i.e., Bifidobacterium and Lactobacillus). We further assessed the genetic diversity within Bifidobacterium, assayed by amplification of the groEL gene. Concomitant with microbial composition changes, we show an increase in the abundance of genes involved in inositol degradation. Despite these changes in gut microbiome composition, we did not detect a consistent shift in SCFA abundance. Collectively, our results demonstrate that on a short-term timescale of 2 weeks, increased fiber intake can induce compositional changes of the gut microbiome, including an increase in MAC-degrading bacteria. IMPORTANCE A profound decrease in the consumption of dietary fiber in many parts of the world in the last century may be associated with the increasing prevalence of type II diabetes, colon cancer, and other health problems. A typical U.S. diet includes about ∼15 g of fiber per day, far less fiber than the daily recommended allowance. Changes in dietary fiber intake affect human health not only through the uptake of nutrients directly but also indirectly through changes in the microbial community and their associated metabolism. Here, we conducted a 2-week diet intervention in healthy young adults to investigate the impact of fiber consumption on the gut microbiome. Participants increased their average fiber consumption by 25 g/day on average for 2 weeks. The high-fiber diet intervention altered the gut microbiome of the study participants, including increases in known fiber-degrading microbes, such as Bifidobacterium and Lactobacillus.
Collapse
|
144
|
Shanahan ER, McMaster JJ, Staudacher HM. Conducting research on diet-microbiome interactions: A review of current challenges, essential methodological principles, and recommendations for best practice in study design. J Hum Nutr Diet 2021; 34:631-644. [PMID: 33639033 DOI: 10.1111/jhn.12868] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/21/2022]
Abstract
Diet is one of the strongest modulators of the gut microbiome. However, the complexity of the interactions between diet and the microbial community emphasises the need for a robust study design and continued methodological development. This review aims to summarise considerations for conducting high-quality diet-microbiome research, outline key challenges unique to the field, and provide advice for addressing these in a practical manner useful to dietitians, microbiologists, gastroenterologists and other diet-microbiome researchers. Searches of databases and references from relevant articles were conducted using the primary search terms 'diet', 'diet intervention', 'dietary analysis', 'microbiome' and 'microbiota', alone or in combination. Publications were considered relevant if they addressed methods for diet and/or microbiome research, or were a human study relevant to diet-microbiome interactions. Best-practice design in diet-microbiome research requires appropriate consideration of the study population and careful choice of trial design and data collection methodology. Ongoing challenges include the collection of dietary data that accurately reflects intake at a timescale relevant to microbial community structure and metabolism, measurement of nutrients in foods pertinent to microbes, improving ability to measure and understand microbial metabolic and functional properties, adequately powering studies, and the considered analysis of multivariate compositional datasets. Collaboration across the disciplines of nutrition science and microbiology is crucial for high-quality diet-microbiome research. Improvements in our understanding of the interaction between nutrient intake and microbial metabolism, as well as continued methodological innovation, will facilitate development of effective evidence-based personalised dietary treatments.
Collapse
Affiliation(s)
- Erin R Shanahan
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | | | - Heidi M Staudacher
- IMPACT (The Institute for Mental and Physical Health and Clinical Translation) Food & Mood Centre, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
145
|
Deehan EC, Colin-Ramirez E, Triador L, Madsen KL, Prado CM, Field CJ, Ball GDC, Tan Q, Orsso C, Dinu I, Pakseresht M, Rubin D, Sharma AM, Tun H, Walter J, Newgard CB, Freemark M, Wine E, Haqq AM. Efficacy of metformin and fermentable fiber combination therapy in adolescents with severe obesity and insulin resistance: study protocol for a double-blind randomized controlled trial. Trials 2021; 22:148. [PMID: 33596993 PMCID: PMC7890810 DOI: 10.1186/s13063-021-05060-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023] Open
Abstract
Background Accumulating evidence suggests that the metabolic effects of metformin and fermentable fibers are mediated, in part, through diverging or overlapping effects on the composition and metabolic functions of the gut microbiome. Pre-clinical animal models have established that the addition of fiber to metformin monotherapy improves glucose tolerance. However, possible synergistic effects of combination therapy (metformin plus fiber) have not been investigated in humans. Moreover, the underlying mechanisms of synergy have yet to be elucidated. The aim of this study is to compare in adolescents with obesity the metabolic effects of metformin and fermentable fibers in combination with those of metformin or fiber alone. We will also determine if therapeutic responses correlate with compositional and functional features of the gut microbiome. Methods This is a parallel three-armed, double-blinded, randomized controlled trial. Adolescents (aged 12–18 years) with obesity, insulin resistance (IR), and a family history of type 2 diabetes mellitus (T2DM) will receive either metformin (850 mg p.o. twice/day), fermentable fibers (35 g/day), or a combination of metformin plus fiber for 12 months. Participants will be seen at baseline, 3, 6, and 12 months, with a phone follow-up at 1 and 9 months. Primary and secondary outcomes will be assessed at baseline, 6, and 12 months. The primary outcome is change in IR estimated by homeostatic model assessment of IR; key secondary outcomes include changes in the Matsuda index, oral disposition index, body mass index z-score, and fat mass to fat-free mass ratio. To gain mechanistic insight, endpoints that reflect host-microbiota interactions will also be assessed: obesity-related immune, metabolic, and satiety markers; humoral metabolites; and fecal microbiota composition, short-chain fatty acids, and bile acids. Discussion This study will compare the potential metabolic benefits of fiber with those of metformin in adolescents with obesity, determine if metformin and fiber act synergistically to improve IR, and elucidate whether the metabolic benefits of metformin and fiber associate with changes in fecal microbiota composition and the output of health-related metabolites. This study will provide insight into the potential role of the gut microbiome as a target for enhancing the therapeutic efficacy of emerging treatments for T2DM prevention. Trial registration ClinicalTrials.gov NCT04578652. Registered on 8 October 2020. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05060-8.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | | | - Lucila Triador
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, T6G 2C2, AB, Canada
| | - Carla M Prado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Geoff D C Ball
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Qiming Tan
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Camila Orsso
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Irina Dinu
- School of Public Health, University of Alberta, Edmonton, T6G 1C9, AB, Canada
| | - Mohammadreza Pakseresht
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Daniela Rubin
- California State University Fullerton, Fullerton, USA
| | - Arya M Sharma
- Department of Medicine, University of Alberta, Edmonton, T6G 2C2, AB, Canada
| | - Hein Tun
- University of Hong Kong School of Public Health, Hong Kong, China
| | - Jens Walter
- DNational University of Ireland University College Cork, University College Cork, Cork, Ireland
| | | | - Michael Freemark
- Duke University Medical Center, Duke University Hospital, Durham, NC, USA
| | - Eytan Wine
- Department of Pediatrics and Physiology, University of Alberta, Edmonton, T6G 1C9, BA, Canada
| | - Andrea M Haqq
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada.
| |
Collapse
|
146
|
Resistant Starch Type 2 from Wheat Reduces Postprandial Glycemic Response with Concurrent Alterations in Gut Microbiota Composition. Nutrients 2021; 13:nu13020645. [PMID: 33671147 PMCID: PMC7922998 DOI: 10.3390/nu13020645] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/03/2023] Open
Abstract
The majority of research on the physiological effects of dietary resistant starch type 2 (RS2) has focused on sources derived from high-amylose maize. In this study, we conduct a double-blind, randomized, placebo-controlled, crossover trial investigating the effects of RS2 from wheat on glycemic response, an important indicator of metabolic health, and the gut microbiota. Overall, consumption of RS2-enriched wheat rolls for one week resulted in reduced postprandial glucose and insulin responses relative to conventional wheat when participants were provided with a standard breakfast meal containing the respective treatment rolls (RS2-enriched or conventional wheat). This was accompanied by an increase in the proportions of bacterial taxa Ruminococcus and Gemmiger in the fecal contents, reflecting the composition in the distal intestine. Additionally, fasting breath hydrogen and methane were increased during RS2-enriched wheat consumption. However, although changes in fecal short-chain fatty acid (SCFA) concentrations were not significant between control and RS-enriched wheat roll consumption, butyrate and total SCFAs were positively correlated with relative abundance of Faecalibacterium, Ruminoccocus, Roseburia, and Barnesiellaceae. These effects show that RS2-enriched wheat consumption results in a reduction in postprandial glycemia, altered gut microbial composition, and increased fermentation activity relative to wild-type wheat.
Collapse
|
147
|
Gut Microbiota Bacterial Species Associated with Mediterranean Diet-Related Food Groups in a Northern Spanish Population. Nutrients 2021; 13:nu13020636. [PMID: 33669303 PMCID: PMC7920039 DOI: 10.3390/nu13020636] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/05/2021] [Accepted: 02/13/2021] [Indexed: 12/11/2022] Open
Abstract
The MD (Mediterranean diet) is recognized as one of the healthiest diets worldwide and is associated with the prevention of cardiovascular and metabolic diseases. Dietary habits are considered one of the strongest modulators of gut microbiota, which seem to play a significant role in health status of the host. The purpose of the present study was to evaluate interactive associations between gut microbiota composition and habitual dietary intake in 360 Spanish adults from the Obekit cohort (normal weight, overweight, and obese participants). Dietary intake and adherence to the MD tests were administered and fecal samples were collected from each participant. Fecal 16S rRNA (ribosomal Ribonucleic Acid) gene sequencing was performed and checked against the dietary habits. MetagenomeSeq was the statistical tool applied to analyze data at the species taxonomic level. Results from this study identified several beneficial bacteria that were more abundant in the individuals with higher adherence to the MD. Bifidobacterium animalis was the species with the strongest association with the MD. Some SCFA (Short Chain Fatty Acids) -producing bacteria were also associated with MD. In conclusion, this study showed that MD, fiber, legumes, vegetable, fruit, and nut intake are associated with an increase in butyrate-producing taxa such as Roseburia faecis, Ruminococcus bromii, and Oscillospira (Flavonifractor) plautii.
Collapse
|
148
|
Abstract
Resistant starch, microbiome, and precision modulation. Mounting evidence has positioned the gut microbiome as a nexus of health. Modulating its phylogenetic composition and function has become an attractive therapeutic prospect. Resistant starches (granular amylase-resistant α-glycans) are available as physicochemically and morphologically distinguishable products. Attempts to leverage resistant starch as microbiome-modifying interventions in clinical studies have yielded remarkable inter-individual variation. Consequently, their utility as a potential therapy likely depends predominantly on the selected resistant starch and the subject's baseline microbiome. The purpose of this review is to detail i) the heterogeneity of resistant starches, ii) how resistant starch is sequentially degraded and fermented by specialized gut microbes, and iii) how resistant starch interventions yield variable effects on the gut microbiome.
Collapse
Affiliation(s)
- Peter A. Dobranowski
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
149
|
Copeland JK, Chao G, Vanderhout S, Acton E, Wang PW, Benchimol EI, El-Sohemy A, Croitoru K, Gommerman JL, Guttman DS, the GEMINI Research Team. The Impact of Migration on the Gut Metagenome of South Asian Canadians. Gut Microbes 2021; 13:1-29. [PMID: 33794735 PMCID: PMC8023248 DOI: 10.1080/19490976.2021.1902705] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023] Open
Abstract
South Asian (SA) Canadian immigrants have a higher risk of developing certain immune-mediated inflammatory diseases compared to non-migrant SAs. We sought to investigate the effect of migration on the gut metagenome and to identify microbiological associations between migration and conditions that may influence the development of immune-mediated inflammatory diseases. Metagenomic analysis of 58 first-generation (GEN1) SA immigrants and 38 unrelated Canadian born children-of-immigrants (GEN2) determined that the time lived in Canada was associated with continued changes in gut microbial communities. Migration of GEN1 to Canada early in life results in a gut community with similarities to GEN2 SA Canadians and non-SA North Americans. Conversely, GEN1 immigrants who arrived recently to Canada exhibited pronounced differences from GEN2, while displaying microbial similarities to a non-migrating SA cohort. Multivariate analysis identified that community composition was primarily influenced by high abundance taxa. Prevotella copri dominated in GEN1 and non-migrant SAs. Clostridia and functionally related Bacteroidia spp. replaced P. copri dominance over generations in Canada. Mutually exclusive Dialister species occurred at differing relative abundances over time and generations in Canada. This shift in species composition is accompanied by a change in genes associated with carbohydrate utilization and short-chain fatty acid production. Total energy derived from carbohydrates compared to protein consumption was significantly higher for GEN1 recent immigrants, which may influence the functional requirements of the gut community. This study demonstrates the associations between migration and the gut microbiome, which may be further associated with the altered risk of immune-mediated inflammatory diseases observed for SA Canadians.
Collapse
Affiliation(s)
- Julia K. Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
| | - Gary Chao
- Department of Immunology, University of Toronto, Toronto, CA, Canada
| | - Shelley Vanderhout
- Nutrigenomix, Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
| | - Erica Acton
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
| | - Pauline W. Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
| | - Eric I. Benchimol
- Department of Pediatrics, and School of Epidemiology and Public Health, University of Ottawa, Ottawa, CA, Canada
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
| | - Ken Croitoru
- Department of Medicine, University of Toronto and Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, CA, Canada
| | | | - David S. Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
| | - the GEMINI Research Team
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
- Department of Immunology, University of Toronto, Toronto, CA, Canada
- Nutrigenomix, Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
- Department of Pediatrics, and School of Epidemiology and Public Health, University of Ottawa, Ottawa, CA, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
- Department of Medicine, University of Toronto and Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, CA, Canada
| |
Collapse
|
150
|
Cockburn DW, Cerqueira FM, Bahr C, Koropatkin NM. The structures of the GH13_36 amylases from Eubacterium rectale and Ruminococcus bromii reveal subsite architectures that favor maltose production. ACTA ACUST UNITED AC 2020. [DOI: 10.1515/amylase-2020-0003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractBacteria in the human gut including Ruminococcus bromii and Eubacterium rectale encode starch-active enzymes that dictate how these bacteria interact with starch to initiate a metabolic cascade that leads to increased butyrate. Here, we determined the structures of two predicted secreted glycoside hydrolase 13 subfamily 36 (GH13_36) enzymes: ErAmy13B complexed with maltotetraose from E. rectale and RbAmy5 from R. bromii. The structures show a limited binding pocket extending from –2 through +2 subsites with limited possibilities for substrate interaction beyond this, which contributes to the propensity for members of this family to produce maltose as their main product. The enzyme structures reveal subtle differences in the +1/+2 subsites that may restrict the recognition of larger starch polymers by ErAmy13B. Our bioinformatic analysis of the biochemically characterized members of the GH13_36 subfamily, which includes the cell-surface GH13 SusG from Bacteroides thetaiotaomicron, suggests that these maltogenic amylases (EC 3.2.1.133) are usually localized to the outside of the cell, display a range of substrate preferences, and most likely contribute to maltose liberation at the cell surface during growth on starch. A broader comparison between GH13_36 and other maltogenic amylase subfamilies explain how the activity profiles of these enzymes are influenced by their structures.
Collapse
Affiliation(s)
- Darrell W. Cockburn
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Food Science, Pennsylvania State University, University Park, PA 16802, USA
| | - Filipe M. Cerqueira
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Constance Bahr
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicole M Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|