151
|
Liang X, Zhang Z, Lv Y, Lu H, Liu T, Yi H, Zhao M, Zhang L, Gong P. Krill Oil Combined with Bifidobacterium animalis subsp. lactis F1-7 Alleviates the Atherosclerosis of ApoE -/- Mice. Foods 2021; 10:foods10102374. [PMID: 34681423 PMCID: PMC8535738 DOI: 10.3390/foods10102374] [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] [Received: 08/05/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022] Open
Abstract
There has been an increasing number of studies on the interaction between active substances and probiotics to improve disease. Both krill oil (KO) and probiotics have the effect of improving atherosclerotic cardiovascular disease, but the combined effect has not been explored. Therefore, the purpose of this study was to explore the improvement effect of KO combined with probiotics on atherosclerosis. The atherosclerotic plaque area of ApoE−/− mice was detected after the intervention of KO, Bifidobacterium animalis subsp. lactis F1-7 (Bif. animalis F1-7), and KO combined with Bif. animalis F1-7. The results showed that Bif. animalis F1-7, KO, and KO combined with Bif. animalis F1-7 could significantly reduce the area of atherosclerotic plaque and improve the levels of serum lipids and inflammatory factors. They could regulate the farnesoid X receptor (FXR)/cholesterol 7-alpha hydroxylase (CYP7A1) pathway to reduce lipid accumulation. The intervention groups could also improve the inflammatory response by downregulating the Toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88) pathway. The anti-inflammatory effect of the interaction group was significantly better than that of KO. It proved that Bif. animalis F1-7 might play a synergistic effect in the improvement of inflammation by KO to the alleviation of atherosclerosis.
Collapse
|
152
|
Li HY, Zhou DD, Gan RY, Huang SY, Zhao CN, Shang A, Xu XY, Li HB. Effects and Mechanisms of Probiotics, Prebiotics, Synbiotics, and Postbiotics on Metabolic Diseases Targeting Gut Microbiota: A Narrative Review. Nutrients 2021; 13:nu13093211. [PMID: 34579087 PMCID: PMC8470858 DOI: 10.3390/nu13093211] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/30/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolic diseases are serious threats to public health and related to gut microbiota. Probiotics, prebiotics, synbiotics, and postbiotics (PPSP) are powerful regulators of gut microbiota, thus possessing prospects for preventing metabolic diseases. Therefore, the effects and mechanisms of PPSP on metabolic diseases targeting gut microbiota are worth discussing and clarifying. Generally, PPSP benefit metabolic diseases management, especially obesity and type 2 diabetes mellitus. The underlying gut microbial-related mechanisms are mainly the modulation of gut microbiota composition, regulation of gut microbial metabolites, and improvement of intestinal barrier function. Moreover, clinical trials showed the benefits of PPSP on patients with metabolic diseases, while the clinical strategies for gestational diabetes mellitus, optimal formula of synbiotics and health benefits of postbiotics need further study. This review fully summarizes the relationship between probiotics, prebiotics, synbiotics, postbiotics, and metabolic diseases, presents promising results and the one in dispute, and especially attention is paid to illustrates potential mechanisms and clinical effects, which could contribute to the next research and development of PPSP.
Collapse
Affiliation(s)
- Hang-Yu Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (H.-Y.L.); (D.-D.Z.); (S.-Y.H.); (A.S.); (X.-Y.X.)
| | - Dan-Dan Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (H.-Y.L.); (D.-D.Z.); (S.-Y.H.); (A.S.); (X.-Y.X.)
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu 610213, China;
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Si-Yu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (H.-Y.L.); (D.-D.Z.); (S.-Y.H.); (A.S.); (X.-Y.X.)
| | - Cai-Ning Zhao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China;
| | - Ao Shang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (H.-Y.L.); (D.-D.Z.); (S.-Y.H.); (A.S.); (X.-Y.X.)
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Xiao-Yu Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (H.-Y.L.); (D.-D.Z.); (S.-Y.H.); (A.S.); (X.-Y.X.)
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (H.-Y.L.); (D.-D.Z.); (S.-Y.H.); (A.S.); (X.-Y.X.)
- Correspondence: ; Tel.: +86-20-8733-2391
| |
Collapse
|
153
|
Zhuang T, Li W, Yang L, Wang Z, Ding L, Zhou M. Gut Microbiota: Novel Therapeutic Target of Ginsenosides for the Treatment of Obesity and Its Complications. Front Pharmacol 2021; 12:731288. [PMID: 34512356 PMCID: PMC8429618 DOI: 10.3389/fphar.2021.731288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, generally characterized by excessive lipid accumulation, is a metabolic threat worldwide due to its rapid growth in global prevalence. Ginsenosides are crucial components derived from natural plants that can confer metabolic benefits for obese patients. Considering the low bioavailability and degradable properties of ginsenosides in vivo, it should be admitted that the mechanism of ginsenosides on anti-obesity contribution is still obscure. Recently, studies have indicated that ginsenoside intervention has beneficial metabolic effects on obesity and its complications because it allows for the correction of gut microbiota dysbiosis and regulates the secretion of related endogenous metabolites. In this review, we summarize the role of gut microbiota in the pathogenetic process of obesity, and explore the mechanism of ginsenosides for ameliorating obesity, which can modulate the composition of gut microbiota by improving the metabolism of intestinal endogenous substances and alleviating the level of inflammation. Ginsenosides are expected to become a promising anti-obesity medical intervention in the foreseeable clinical settings.
Collapse
Affiliation(s)
- Tongxi Zhuang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Li
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Li Yang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Zhengtao Wang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Lili Ding
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Mingmei Zhou
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
154
|
Effects of crude Sphallerocarpus gracilis polysaccharides as potential prebiotics on acidifying activity and growth of probiotics in fermented milk. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
155
|
Abstract
PURPOSE OF REVIEW To review the components of the intestinal barrier, the practical measurements of intestinal permeability, and the clinical conditions associated with altered intestinal barrier function, and to summarize the effects of dietary substances that fortify or weaken the intestinal barrier. RECENT FINDINGS The intestinal barrier includes surface mucus, epithelial layer, and immune defense mechanisms. Transport across the epithelium may result from increased paracellular transport, apoptosis, or transcellular permeability. Assessment of the intestinal barrier requires measurements beyond the transport across the epithelial layer or the measurement of tight junction expression. Barrier function is most meaningfully tested in vivo using orally administered probe molecules; other approaches are performed in vitro using mucosal biopsies from humans, or exposing colonic mucosa from rats or mice or cell layers to extracts of colonic mucosa or stool from patients. Dietary factors can influence intestinal leakiness: fortifying the barrier with vitamins A and D, zinc, short-chain fatty acids, methionine, glutamine, and probiotics; weakening of the barrier has been reported with fat, bile acids, emulsifiers, and gliadin. Intestinal mucosal leakiness in 'stress' disorders such as major burns is reversed with enteral glutamine. SUMMARY Inflammatory or ulcerating intestinal diseases result in leakiness of the gut barrier; however, no such disease has been cured by simply normalizing intestinal barrier function. Similarly, it is still unproven that restoring barrier function (reversing 'leaky gut') can ameliorate clinical manifestations in nonulcerating gastrointestinal disease or systemic or neurological diseases. On the other hand, dietary and enteral interventions can fortify the intestinal barrier in stress-associated states.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
156
|
Zhao L, Xie Q, Etareri Evivie S, Liu D, Dong J, Ping L, Liu F, Li B, Huo G. Bifidobacterium dentium N8 with potential probiotic characteristics prevents LPS-induced intestinal barrier injury by alleviating the inflammatory response and regulating the tight junction in Caco-2 cell monolayers. Food Funct 2021; 12:7171-7184. [PMID: 34269367 DOI: 10.1039/d1fo01164b] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The intestinal barrier is vital for preventing inflammatory bowel disease (IBD). This study aimed to investigate the potential mechanism behind the protective effects of B. dentium N8 on the intestinal barrier using the lipopolysaccharide (LPS)-induced Caco-2 cells model. Our probiotic validation results showed that B. dentium N8 had a higher adhesion ability and a more substantial inhibition effect on Escherichia coli ATCC 25922 adhesion to HT-29 cells. Regarding the epithelial integrity, B. dentium N8 significantly increased the trans-epithelial electrical resistance (TEER) value and decreased the paracellular permeability of Caco-2 cells stimulated by lipopolysaccharide (LPS). In addition, B. dentium N8 significantly increased ZO-1, occludin, and claudin-1 mRNA expression. B. dentium N8 downregulated the mRNA expression level of TLR4 and pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). Furthermore, B. dentium N8 had a better protective effect on the intestinal barrier than that of E7. Comparative genomics of B. dentium N8 and E7 showed B. dentium N8 had the specific genes encoding for adhesion ability and immune system regulation. The findings provide the theoretical basis for B. dentium N8 possessing a protective effect on the intestinal barrier, which indicate that it could be used as a novel therapy for IBD.
Collapse
Affiliation(s)
- Li Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Chaiyasut C, Sivamaruthi BS, Kesika P, Khongtan S, Khampithum N, Thangaleela S, Peerajan S, Bumrungpert A, Chaiyasut K, Sirilun S, Sittiprapaporn P. Synbiotic Supplementation Improves Obesity Index and Metabolic Biomarkers in Thai Obese Adults: A Randomized Clinical Trial. Foods 2021; 10:1580. [PMID: 34359450 PMCID: PMC8304233 DOI: 10.3390/foods10071580] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
The cluster of metabolic disorders includes obesity, dyslipidemia, hypertension, and glucose intolerance, increasing the risk of developing cardiovascular diseases and type 2 diabetes. Evolving proofs suggest an essential role of microbiota in human health and disease, including digestion, energy and glucose metabolism, immunomodulation, and brain function. The frequency of overweight is increasing, and the main causes for this are highly processed foods and less active lifestyles. Research is underway to unravel the probable relationship between obesity and intestinal microbiota. Here, we propose a method to understand and elucidate the synergistic function of prebiotics and probiotics in treating obesity. The biomarkers of obesity, such as cholesterol, gut permeability, oxidative stress, bacterial toxins, cytokines, and short-chain fatty acids, were analyzed in Thai obese individuals after being supplemented with a synbiotic preparation containing Lactobacillus paracasei, Bifidobacterium longum, Bifidobacterium breve, inulin, and fructooligosaccharide. The results reveal that the supplementation of synbiotics significantly altered the obesity-associated biomarkers in an appositive way. Further studies are warranted to use synbiotics as an adjuvant therapy for the management of obesity-related health issues.
Collapse
Affiliation(s)
- Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | - Bhagavathi Sundaram Sivamaruthi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | - Periyanaina Kesika
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | - Suchanat Khongtan
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | - Nanticha Khampithum
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | - Subramanian Thangaleela
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | | | - Akkarach Bumrungpert
- Mahidol Nutrition Society, Faculty of Public Health, Mahidol University, Bangkok 10400, Thailand;
- Research Center of Nutraceuticals and Natural Products for Health & Anti-Aging, College of Integrative Medicine, Dhurakij Pundit University, Bangkok 10210, Thailand
| | - Khontaros Chaiyasut
- Institute of Research and Development, Chiang Mai Rajabhat University, Chiangmai 50300, Thailand;
| | - Sasithorn Sirilun
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (P.K.); (S.K.); (N.K.); (S.T.); (S.S.)
| | - Phakkharawat Sittiprapaporn
- Neuropsychological Research Laboratory, Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 11120, Thailand
| |
Collapse
|
158
|
Pontes KSDS, Guedes MR, Cunha MRD, Mattos SDS, Barreto Silva MI, Neves MF, Marques BCAA, Klein MRST. Effects of probiotics on body adiposity and cardiovascular risk markers in individuals with overweight and obesity: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2021; 40:4915-4931. [PMID: 34358838 DOI: 10.1016/j.clnu.2021.06.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Evidence suggests that gut microbiota is a potential factor in the pathophysiology of both obesity and related metabolic disorders. While individual randomized controlled trials (RCTs) have evaluated the effects of probiotics on adiposity and cardiovascular disease (CVD) risk factors in subjects with overweight and obesity, the results are inconsistent. Thus, this systematic review and meta-analysis aimed to evaluate the effects of probiotic supplementation on body weight, body adiposity and CVD risk markers in overweight and obese subjects. METHODS A systematic search for RCTs published up to December 2020 was conducted in MEDLINE (via PubMed), EMBASE, Scopus and LILACS. Meta-analysis using a random-effects model was chosen to analyze the impact of combined trials. RESULTS Twenty-six RCTs (n = 1720) were included. Data pooling showed a significant effect of probiotics in reducing body weight (MD:-0.70 kg; 95%CI:-1.04,-0.35 kg; P < 0.0001), body mass index (BMI) (MD:-0.24 kg/m2; 95%CI:-0.35,-0.12 kg/m2; P = 0.0001), waist circumference (WC) (MD:-1.13 cm; 95%CI:-1.54,-0.73 cm; P < 0.0001), fat mass (MD:-0.71 kg; 95%CI:-1.10,-0.32 kg; P = 0.0004), tumor necrosis factor-α (MD:-0.16 pg/ml; 95%CI:-0.24,-0.08 pg/ml; P = 0.0001), insulin (MD:-0.85mcU/ml; 95%CI:-1.50,-0.21mcU/ml; P = 0.010), total cholesterol (MD:-0.16 mmol/l; 95%CI:-0.26,-0.05 mmol/l; P = 0.003) and LDL (MD:-0.09 mmol/l; 95%CI:-0.16,-0.03 mmol/l; P = 0.006) compared with control groups. There was a significant decrease in body weight, BMI and WC in studies using both single and multi-bacterial species. Decreases in body adiposity parameters were only observed in studies using a probiotic dose of ≥ 1010 CFU and for ≥8 weeks duration. CONCLUSIONS The present meta-analysis suggests that probiotics consumption may be helpful for improving body weight, body adiposity and some CVD risk markers in individuals with overweight and obesity. The review was registered on PROSPERO (International prospective register of systematic reviews): CRD42020183136.
Collapse
Affiliation(s)
- Karine Scanci da Silva Pontes
- Post-Graduation Program in Clinical and Experimental Pathophysiology, State University of Rio de Janeiro (UERJ), Av. Professor Manuel de Abreu, 444, Térreo - Rio de Janeiro, RJ, 20550-170, Brazil.
| | - Marcella Rodrigues Guedes
- Post-Graduation Program in Clinical and Experimental Pathophysiology, State University of Rio de Janeiro (UERJ), Av. Professor Manuel de Abreu, 444, Térreo - Rio de Janeiro, RJ, 20550-170, Brazil.
| | - Michelle Rabello da Cunha
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Samanta de Souza Mattos
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Maria Inês Barreto Silva
- Department of Applied Nutrition, Nutrition Institute, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524 - Pavilhão João Lyra Filho, 12º Andar, Bloco D, Rio de Janeiro, RJ, 20559-900, Brazil; Department of Applied Nutrition, Nutrition School, Federal University of the State of Rio de Janeiro (UNIRIO), Av. Pasteur, 296, Botafogo, 3º Andar, Rio de Janeiro, RJ, 22290-250, Brazil.
| | - Mario Fritsch Neves
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Bianca Cristina Antunes Alves Marques
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil; Department of Nutrition and Dietetics, National Cancer Institute (INCA), Av. Binário do Porto, 831, Rio de Janeiro, RJ, 20081-250, Brazil.
| | - Márcia Regina Simas Torres Klein
- Department of Applied Nutrition, Nutrition Institute, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524 - Pavilhão João Lyra Filho, 12º Andar, Bloco D, Rio de Janeiro, RJ, 20559-900, Brazil.
| |
Collapse
|
159
|
Influence of a High-Impact Multidimensional Rehabilitation Program on the Gut Microbiota of Patients with Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms22137173. [PMID: 34281224 PMCID: PMC8268819 DOI: 10.3390/ijms22137173] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/04/2023] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative inflammatory condition mediated by autoreactive immune processes. Due to its potential to influence host immunity and gut-brain communication, the gut microbiota has been suggested to be involved in the onset and progression of MS. To date, there is no definitive cure for MS, and rehabilitation programs are of the utmost importance, especially in the later stages. However, only a few people generally participate due to poor support, knowledge, and motivation, and no information is available on gut microbiota changes. Herein we evaluated the potential of a brief high-impact multidimensional rehabilitation program (B-HIPE) in a leisure environment to affect the gut microbiota, mitigate MS symptoms and improve quality of life. B-HIPE resulted in modulation of the MS-typical dysbiosis, with reduced levels of pathobionts and the replenishment of beneficial short-chain fatty acid producers. This partial recovery of a eubiotic profile could help counteract the inflammatory tone typically observed in MS, as supported by reduced circulating lipopolysaccharide levels and decreased populations of pro-inflammatory lymphocytes. Improved physical performance and fatigue relief were also found. Our findings pave the way for integrating clinical practice with holistic approaches to mitigate MS symptoms and improve patients’ quality of life.
Collapse
|
160
|
Tsujimoto H, Hirata Y, Ueda Y, Kinoshita N, Tawa H, Tanaka Y, Koshiba R, Ota K, Kojima Y, Kakimoto K, Takeuchi T, Miyazaki T, Nakamura S, Higuchi K. Effect of a proton-pump inhibitor on intestinal microbiota in patients taking low-dose aspirin. Eur J Clin Pharmacol 2021; 77:1639-1648. [PMID: 34085115 DOI: 10.1007/s00228-021-03167-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIM Low-dose aspirin (LDA) administration prevents cerebral infarction and myocardial infarction, but many studies found an association with mucosal injury. Proton-pump inhibitors (PPIs) can prevent gastric and duodenal mucosal damage, but they may exacerbate small-intestinal mucosal injury by altering the microbiota. We aimed to assess the effect of PPIs on the intestinal flora of LDA users. METHODS Thirty-two recruited patients, who received LDA (100 mg/day) but did not take PPIs, were divided into 15 patients additionally receiving esomeprazole (20 mg/day) and 17 patients additionally receiving vonoprazan (10 mg/day). On days 0, 30, 90, and 180, the microbiota of each patient was examined by terminal restriction fragment length polymorphism analysis, and the serum gastrin, hemoglobin, and hematocrit levels were measured. RESULTS Additional PPI administration increased the proportion of Lactobacillales in the microbiota of LDA users. This trend was more prevalent in the vonoprazan group (p < 0.0001) than in the esomeprazole group (p = 0.0024). The Lactobacillales proportion was positively correlated with the gastrin level (r = 0.5354). No significant hemoglobin or hematocrit level reduction was observed in subjects receiving LDA with additional PPI. CONCLUSIONS Additional PPI administration increased the Lactobacillales proportion in the microbiota of LDA users. The positive correlation between the gastrin level and the proportion of Lactobacillales suggested that the change in the intestinal flora was associated with the degree of suppression of gastric acid secretion. Additional oral PPI did not significantly promote anemia, but the risk of causing PPI-induced small-intestinal mucosal injury in LDA users should be considered.
Collapse
Affiliation(s)
- Hiroyuki Tsujimoto
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Yuki Hirata
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan.
| | - Yasuhiro Ueda
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Naohiko Kinoshita
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Hideki Tawa
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Yasuyoshi Tanaka
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Ryoji Koshiba
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kazuhiro Ota
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Yuichi Kojima
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kazuki Kakimoto
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Toshihisa Takeuchi
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Takako Miyazaki
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Shiro Nakamura
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kazuhide Higuchi
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| |
Collapse
|
161
|
Méndez-Salazar EO, Vázquez-Mellado J, Casimiro-Soriguer CS, Dopazo J, Çubuk C, Zamudio-Cuevas Y, Francisco-Balderas A, Martínez-Flores K, Fernández-Torres J, Lozada-Pérez C, Pineda C, Sánchez-González A, Silveira LH, Burguete-García AI, Orbe-Orihuela C, Lagunas-Martínez A, Vazquez-Gomez A, López-Reyes A, Palacios-González B, Martínez-Nava GA. Taxonomic variations in the gut microbiome of gout patients with and without tophi might have a functional impact on urate metabolism. Mol Med 2021; 27:50. [PMID: 34030623 PMCID: PMC8142508 DOI: 10.1186/s10020-021-00311-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Objective To evaluate the taxonomic composition of the gut microbiome in gout patients with and without tophi formation, and predict bacterial functions that might have an impact on urate metabolism. Methods Hypervariable V3–V4 regions of the bacterial 16S rRNA gene from fecal samples of gout patients with and without tophi (n = 33 and n = 25, respectively) were sequenced and compared to fecal samples from 53 healthy controls. We explored predictive functional profiles using bioinformatics in order to identify differences in taxonomy and metabolic pathways. Results We identified a microbiome characterized by the lowest richness and a higher abundance of Phascolarctobacterium, Bacteroides, Akkermansia, and Ruminococcus_gnavus_group genera in patients with gout without tophi when compared to controls. The Proteobacteria phylum and the Escherichia-Shigella genus were more abundant in patients with tophaceous gout than in controls. Fold change analysis detected nine genera enriched in healthy controls compared to gout groups (Bifidobacterium, Butyricicoccus, Oscillobacter, Ruminococcaceae_UCG_010, Lachnospiraceae_ND2007_group, Haemophilus, Ruminococcus_1, Clostridium_sensu_stricto_1, and Ruminococcaceae_UGC_013). We found that the core microbiota of both gout groups shared Bacteroides caccae, Bacteroides stercoris ATCC 43183, and Bacteroides coprocola DSM 17136. These bacteria might perform functions linked to one-carbon metabolism, nucleotide binding, amino acid biosynthesis, and purine biosynthesis. Finally, we observed differences in key bacterial enzymes involved in urate synthesis, degradation, and elimination. Conclusion Our findings revealed that taxonomic variations in the gut microbiome of gout patients with and without tophi might have a functional impact on urate metabolism. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00311-5.
Collapse
Affiliation(s)
- Eder Orlando Méndez-Salazar
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM-INMEGEN, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Tlalpan, 14610, Mexico City, Mexico.,Programa de Doctorado en ICES, Facultad de Química, UNAM, Mexico City, Mexico
| | - Janitzia Vázquez-Mellado
- Rheumatology Department, Hospital General de México Eduardo Liceaga Mexico City, Mexico City, Mexico
| | - Carlos S Casimiro-Soriguer
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, 41013, Sevilla, Spain.,Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013, Sevilla, Spain
| | - Joaquin Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, 41013, Sevilla, Spain.,Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013, Sevilla, Spain.,Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, 41013, Sevilla, Spain.,FPS/ELIXIR-Es, Hospital Virgen del Rocío, 42013, Sevilla, Spain
| | - Cankut Çubuk
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calz México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Adriana Francisco-Balderas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calz México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Karina Martínez-Flores
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calz México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Javier Fernández-Torres
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calz México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico
| | - Carlos Lozada-Pérez
- Servicio de Reumatología, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Carlos Pineda
- División de Enfermedades Musculo-Esqueléticas y Reumáticas, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | | | - Luis H Silveira
- Departamento de Reumatología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Ana I Burguete-García
- Departamento de Epidemiología Genética, Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Morelos, Mexico
| | - Citlalli Orbe-Orihuela
- Departamento de Epidemiología Genética, Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Morelos, Mexico
| | - Alfredo Lagunas-Martínez
- Departamento de Epidemiología Genética, Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Morelos, Mexico
| | - Alonso Vazquez-Gomez
- Hospital General Regional No. 1 "Ignacio García Tellez", Instituto Mexicano del Seguro Social, Mérida, Yucatán, Mexico
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Berenice Palacios-González
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM-INMEGEN, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Tlalpan, 14610, Mexico City, Mexico.
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calz México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico
| |
Collapse
|
162
|
Stevens Y, Pinheiro I, Salden B, Duysburgh C, Bolca S, Degroote J, Majdeddin M, Van Noten N, Gleize B, Caris-Veyrat C, Michiels J, Jonkers D, Troost F, Possemiers S, Masclee A. Effect of a carotenoid-producing Bacillus strain on intestinal barrier integrity and systemic delivery of carotenoids: A randomised trial in animals and humans. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
163
|
Rodríguez-Sorrento A, Castillejos L, López-Colom P, Cifuentes-Orjuela G, Rodríguez-Palmero M, Moreno-Muñoz JA, Luise D, Trevisi P, Martín-Orúe SM. Effects of the Administration of Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001 and Their Synbiotic Combination With Galacto-Oligosaccharides Against Enterotoxigenic Escherichia coli F4 in an Early Weaned Piglet Model. Front Microbiol 2021; 12:642549. [PMID: 33935999 PMCID: PMC8086512 DOI: 10.3389/fmicb.2021.642549] [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] [Received: 12/16/2020] [Accepted: 03/23/2021] [Indexed: 01/09/2023] Open
Abstract
We evaluated the potential of multi-strain probiotic (Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001) with or without galacto-oligosaccharides against enterotoxigenic Escherichia coli (ETEC) F4 infection in post-weaning pigs. Ninety-six piglets were distributed into 32 pens assigned to five treatments: one non-challenged (CTR+) and four challenged: control diet (CTR-), with probiotics (>3 × 1010 CFU/kg body weight each, PRO), prebiotic (5%, PRE), or their combination (SYN). After 1 week, animals were orally inoculated with ETEC F4. Feed intake, weight, and clinical signs were recorded. On days 4 and 8 post-inoculation (PI), one animal per pen was euthanized and samples from blood, digesta, and tissues collected. Microbiological counts, ETEC F4 real-time PCR (qPCR) quantification, fermentation products, serum biomarkers, ileal histomorphometry, and genotype for mucin 4 (MUC4) polymorphism were determined. Animals in the PRO group had similar enterobacteria and coliform numbers to the CTR+ group, and the ETEC F4 prevalence, the number of mitotic cells at day 4 PI, and villus height at day 8 PI were between that observed in the CTR+ and CTR- groups. The PRO group exhibited reduced pig major acute-phase protein (Pig-MAP) levels on day 4 PI. The PRE diet group presented similar reductions in ETEC F4 and Pig-MAP, but there was no effect on microbial groups. The SYN group showed reduced fecal enterobacteria and coliform counts after the adaptation week but, after the inoculation, the SYN group showed lower performance and more animals with high ETEC F4 counts at day 8 PI. SYN treatment modified the colonic fermentation differently depending on the MUC4 polymorphism. These results confirm the potential of the probiotic strains and the prebiotic to fight ETEC F4, but do not show any synergy when administered together, at least in this animal model.
Collapse
Affiliation(s)
- Agustina Rodríguez-Sorrento
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lorena Castillejos
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Paola López-Colom
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | - Diana Luise
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Susana María Martín-Orúe
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
164
|
Warda AK, Clooney AG, Ryan F, de Almeida Bettio PH, Di Benedetto G, Ross RP, Hill C. A postbiotic consisting of heat-treated lactobacilli has a bifidogenic effect in pure culture and in human fermented faecal communities. Appl Environ Microbiol 2021; 87:AEM.02459-20. [PMID: 33579683 PMCID: PMC8091120 DOI: 10.1128/aem.02459-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/05/2021] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota has a significant impact on host health. Dietary interventions using probiotics, prebiotics and postbiotics have the potential to alter microbiota composition and function. Other therapeutic interventions such as antibiotics and faecal microbiota transplantation have also been shown to significantly alter the microbiota and its metabolites. Supplementation of a faecal fermentation model of the human gut with a postbiotic product Lactobacillus LB led to changes in microbiome composition (i.e. increase in beneficial bifidobacteria) and associated metabolic changes (i.e. increased acid production). Lactobacillus LB is a heat-treated preparation of cellular biomass and a fermentate generated by Limosilactobacillus fermentum CNCM MA65/4E-1b (formerly known as Lactobacillus fermentum CNCM MA65/4E-1b) and Lactobacillus delbrueckii ssp. delbrueckii CNCM MA65/4E-2z, medically relevant strains used to produce antidiarrheal preparations. In pure culture, Lactobacillus LB also stimulates the growth of a range of bifidobacterial species and strains. Lactobacillus LB-like preparations generated using other Lactobacillaceae, including commercially available probiotic bacteria, did not have the same impact on a model strain (Bifidobacterium longum subsp. infantis ATCC 15697). This bifidogenic activity is heat- and enzyme-stable and cannot be attributed to lactose, which is a major constituent of Lactobacillus LB. L fermentum CNCM MA65/4E-1b is largely responsible for the observed activity and there is a clear role for compounds smaller than 1 kDa.Importance In general, disruptions to the gut microbiota are associated with multiple disorders in humans. The presence of high levels of Bifidobacterium spp. in the human gut is commonly considered to be beneficial. Bifidobacteria can be supplemented in the diet (as probiotics) or those bifidobacteria already present in the gut can be stimulated by the consumption of prebiotics such as inulin. We demonstrate that Lactobacillus LB (a product consisting of two heat-killed lactic acid bacteria and their metabolites) can stimulate the growth of bifidobacteria in human fermented faecal communities and in pure culture. Given the heat-treatment applied during the production process, there is no risk of the lactic acid bacteria colonising (or causing bacteraemia) in vulnerable consumers (infants, immunocompromised, etc). Lactobacillus LB has the potential to affect human health by selectively promoting the growth of beneficial bacteria.
Collapse
Affiliation(s)
- Alicja K Warda
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Adam G Clooney
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Feargal Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | - Reynolds P Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
165
|
Xiao Y, Zhai Q, Zhang H, Chen W, Hill C. Gut Colonization Mechanisms of Lactobacillus and Bifidobacterium: An Argument for Personalized Designs. Annu Rev Food Sci Technol 2021; 12:213-233. [DOI: 10.1146/annurev-food-061120-014739] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lactobacillus and Bifidobacterium spp. are best understood for their applications as probiotics, which are often transient, but as commensals it is probable that stable colonization in the gut is important for their beneficial roles. Recent research suggests that the establishment and persistence of strains of Lactobacillus and Bifidobacterium in the gut are species- and strain-specific and affected by natural history, genomic adaptability, and metabolic interactions of the bacteria and the microbiome and immune aspects of the host but also regulated by diet. This provides new perspectives on the underlying molecular mechanisms. With an emphasis on host–microbe interaction, this review outlines how the characteristics of individual Lactobacillus and Bifidobacterium bacteria, the host genotype and microbiome structure,diet, and host–microbe coadaptation during bacterial gut transition determine and influence the colonization process. The diet-tuned and personally tailored colonization can be achieved via a machine learning prediction model proposed here.
Collapse
Affiliation(s)
- Yue Xiao
- State Key Laboratory of Food Science and Technology and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China;, , ,
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China;, , ,
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China;, , ,
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China;, , ,
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- Beijing Advanced Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| | - Colin Hill
- School of Microbiology and APC Microbiome Institute, University College Cork, Cork T12 YN60, Ireland
| |
Collapse
|
166
|
Bifidobacterium adolescentis Isolated from Different Hosts Modifies the Intestinal Microbiota and Displays Differential Metabolic and Immunomodulatory Properties in Mice Fed a High-Fat Diet. Nutrients 2021; 13:nu13031017. [PMID: 33801119 PMCID: PMC8004121 DOI: 10.3390/nu13031017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/07/2023] Open
Abstract
The incidence of obesity, which is closely associated with the gut microbiota and chronic inflammation, has rapidly increased in the past 40 years. Therefore, the probiotic-based modification of the intestinal microbiota composition has been developed as a strategy for the treatment of obesity. In this study, we selected four Bifidobacterium adolescentis strains isolated from the feces of newborn and elderly humans to investigate whether supplementation with B. adolescentis of various origins could alleviate obesity in mice. Male C57BL/6J mice fed a high-fat diet (HFD, 60% energy as fat) received one of the following 14-week interventions: (i) B. adolescentis N4_N3, (ii) B. adolescentis Z25, (iii) B. adolescentis 17_3, (iv) B. adolescentis 2016_7_2, and (v) phosphate-buffered saline. The metabolic parameters, thermogenesis, and immunity of all treated mice were measured. Cecal and colonic microbial profiles were determined by 16S rRNA gene sequencing. Intestinal concentrations of short-chain fatty acids (SCFAs) were measured by gas chromatography-mass spectrometry (GC-MS). The B. adolescentis strains isolated from the feces of elderly humans (B. adolescentis Z25, 17_3, and 2016_7_2) decreased the body weight or weight gain of mice, whilst the strain isolated from the newborn (B. adolescentis N4_N3) increased the body weight of mice. The B. adolescentis strains isolated from the elderly also increased serum leptin concentrations and induced the expression of thermogenesis- and lipid metabolism-related genes in brown adipose tissue. All the B. adolescentis strains alleviated inflammations in the spleen and brain and modified the cecal and colonic microbiota. Particularly, all strains reversed the HFD-induced depletion of Bifidobacterium and reduced the development of beta-lactam resistance. In addition, the B. adolescentis strains isolated from the elderly increased the relative abundances of potentially beneficial genera, such as Bacteroides, Parabacteroides, and Faecalibaculum. We speculate that such increased abundance of commensal bacteria may have mediated the alleviation of obesity, as B. adolescentis supplementation decreased the intestinal production of SCFAs, thereby reducing energy delivery to the host mice. Our results revealed that certain strains of B. adolescentis can alleviate obesity and modify the gut microbiota of mice. The tested strains of B. adolescentis showed different effects on lipid metabolism and immunity regulation, with these effects related to whether they had been isolated from the feces of newborn or elderly humans. This indicates that B. adolescentis from different sources may have disparate effects on host health possibly due to the transmission of origin-specific functions to the host.
Collapse
|
167
|
Cai J, Chen Z, Wu W, Lin Q, Liang Y. High animal protein diet and gut microbiota in human health. Crit Rev Food Sci Nutr 2021; 62:6225-6237. [PMID: 33724115 DOI: 10.1080/10408398.2021.1898336] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The role of the intestinal flora in health and disease has become a research hotspot. Compared with carbohydrates and fats, proteins are metabolized primarily by microbial fermentation in the intestine. The production of protein fermentation products and metabolites depends on the composition, diversity, and metabolism of the gut microbiota. Several protein fermentation products, including indoles, phenols, polyamines, hydrogen sulfide (H2S), amines, and carnitine, are toxic. This study analyzes the relationship between high-protein diets (HPDs), the intestinal microbiota, and human health and disease. Long-term HPDs increase the risk of intestinal diseases, type 2 diabetes (T2DM), obesity, central nervous system (CNS) diseases, and cardiovascular diseases (CVD) by producing toxic metabolites in the colon, including amines, H2S, and ammonia. Short-term HPDs have little effect on the metabolism of healthy individuals under 65 years old. However, meeting the protein requirements of individuals over 65 years old using HPDs is more challenging. The adverse effects of HPDs on athletes are minimal. Natural compounds (plant extracts, whose main constituents are polysaccharides and polyphenols), prebiotics, probiotics, and regular physical exercise improve gut dysbiosis and reduce disease risk.
Collapse
Affiliation(s)
- Jie Cai
- Molecular Nutrition Branch, National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Zhongxu Chen
- Molecular Nutrition Branch, National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Wei Wu
- Molecular Nutrition Branch, National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Qinlu Lin
- Molecular Nutrition Branch, National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Ying Liang
- Molecular Nutrition Branch, National Engineering Laboratory for Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
168
|
Ruggiero AD, Key CCC, Kavanagh K. Adipose Tissue Macrophage Polarization in Healthy and Unhealthy Obesity. Front Nutr 2021; 8:625331. [PMID: 33681276 PMCID: PMC7925825 DOI: 10.3389/fnut.2021.625331] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Over 650 million adults are obese (body mass index ≥ 30 kg/m2) worldwide. Obesity is commonly associated with several comorbidities, including cardiovascular disease and type II diabetes. However, compiled estimates suggest that from 5 to 40% of obese individuals do not experience metabolic or cardiovascular complications. The existence of the metabolically unhealthy obese (MUO) and the metabolically healthy obese (MHO) phenotypes suggests that underlying differences exist in both tissues and overall systemic function. Macrophage accumulation in white adipose tissue (AT) in obesity is typically associated with insulin resistance. However, as plastic cells, macrophages respond to stimuli in their microenvironments, altering their polarization between pro- and anti-inflammatory phenotypes, depending on the state of their surroundings. The dichotomous nature of MHO and MUO clinical phenotypes suggests that differences in white AT function dictate local inflammatory responses by driving changes in macrophage subtypes. As obesity requires extensive AT expansion, we posit that remodeling capacity with adipose expansion potentiates favorable macrophage profiles in MHO as compared with MUO individuals. In this review, we discuss how differences in adipogenesis, AT extracellular matrix deposition and breakdown, and AT angiogenesis perpetuate altered AT macrophage profiles in MUO compared with MHO. We discuss how non-autonomous effects of remote organ systems, including the liver, gastrointestinal tract, and cardiovascular system, interact with white adipose favorably in MHO. Preferential AT macrophage profiles in MHO stem from sustained AT function and improved overall fitness and systemic health.
Collapse
Affiliation(s)
- Alistaire D Ruggiero
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Chia-Chi Chuang Key
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kylie Kavanagh
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.,Department of Biomedicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
169
|
Cortés-Martín A, Iglesias-Aguirre CE, Meoro A, Selma MV, Espín JC. Pharmacological Therapy Determines the Gut Microbiota Modulation by a Pomegranate Extract Nutraceutical in Metabolic Syndrome: A Randomized Clinical Trial. Mol Nutr Food Res 2021; 65:e2001048. [PMID: 33458928 DOI: 10.1002/mnfr.202001048] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/04/2020] [Indexed: 12/13/2022]
Abstract
SCOPE Poly-pharmacological therapy shapes the gut microbiota (GM) in metabolic syndrome (MetS) patients. The effects of polyphenols in poly-medicated MetS patients are unknown. METHODS AND RESULTS A randomized, placebo-controlled, double-blinded, and crossover trial in poly-medicated MetS patients (n=50) explored whether the effects of a pomegranate extract nutraceutical (PE, 320 mg phenolics/day for 1 month) are affected by the drug therapy. Considering the lipid-lowering (LL-), anti-hypertensive (HP-) and(or) anti-diabetic (AD-) treatments: GM (16S rRNA sequencing), short-chain fatty acids, 40 inflammatory-metabolic and endotoxemia-related biomarkers, associations between biomarkers and GM with 53 cardiometabolic dysfunctions-related single-nucleotide polymorphisms (SNPs), and urolithin metabotypes (UMs) influence are evaluated. Representative SNPs-GM associations after PE include Lactococcus and ClostridiumXIVa with rs5443-GNB3 (G-protein-β-polypeptide-3) and ClostridiumXIVa with rs7903146-TCF7L2 (transcription-factor-7-like-2) and rs1137101-LEPR (leptin-receptor). PE decreases sICAM-1 in LL-patients and the lipopolysaccharide-binding protein in all the patients. PE does not affect the other patients' markers as a group or stratifying by UMs. After PE, Lactococcus increases in AD-, LL-, and HP-patients, Bifidobacterium increases in LL- and AD-, while Clostridium XIVa decreases in non-LL- and non-HP-patients. CONCLUSION The prebiotic effect of PE depends on the medication, mainly on HP-treatments. Targeting GM can complement MetS therapy, but the patients' drug therapy should be considered individually.
Collapse
Affiliation(s)
- Adrián Cortés-Martín
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia, 30100, Spain
| | - Carlos Eduardo Iglesias-Aguirre
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia, 30100, Spain
| | - Amparo Meoro
- Service of Endocrinology, Reina Sofía University Hospital, Avda. Intendente Jorge Palacios s/n, Murcia, 30003, Spain
| | - María Victoria Selma
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia, 30100, Spain
| | - Juan Carlos Espín
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia, 30100, Spain
| |
Collapse
|
170
|
An overview of the level of dietary support in the gut microbiota at different stages of life: A systematic review. Clin Nutr ESPEN 2021; 42:41-52. [PMID: 33745615 DOI: 10.1016/j.clnesp.2021.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS The gut microbiome is an essential factor for the health of the host. Several factors may alter the gut's microbiota composition, including genetic factors, lifestyle, aging, and dietary intervention. This process can be an essential element in the prevention and treatment of diseases associated with microbiome dysfunction through appropriate dietary interventions. Based on this context, a systematic review was carried out in order to assess the effect of dietary intervention on the profile of the gut microbiota throughout different stages of life. METHODS The systematic review was conducted following the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA), with the eligibility criteria following the principle of PICOS. The literature search was carried out in 2019 throughout PubMed/MEDLINE, Scopus, and Science Direct. Thus, 1237 studies were selected, and 40 articles were included by criteria. RESULTS According to the level of evidence of Centre for Evidence-Based Medicine (OCEBM), 21 studies reached the level of evidence B1, 15 articles were classified with B2, and four articles with B3. No dietary intervention was applied at all stages of life, nor with similar proportions of intervention. No dietary intervention was applied at all stages of life, nor with similar proportions of intervention. On the other hand, dietary interventions alter the intestinal microbiota in different pathological realities. CONCLUSIONS Different dietary interventions change the microbiome composition at all stages of life in healthy and pathological individuals. However, more clinical studies are needed to identify the specifics of each stage in response to interventions.
Collapse
|
171
|
Spiller R. Impact of Diet on Symptoms of the Irritable Bowel Syndrome. Nutrients 2021; 13:nu13020575. [PMID: 33572262 PMCID: PMC7915127 DOI: 10.3390/nu13020575] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/08/2023] Open
Abstract
Irritable bowel syndrome (IBS), with its key features of abdominal pain and disturbed bowel habit, is thought by both patients and clinicians to be strongly influenced by diet. However, the complexities of diet have made identifying specific food intolerances difficult. Eating disorders can masquerade as IBS and may need specialist treatment. While typical food allergy is readily distinguished from IBS, the mechanisms of gut-specific adverse reactions to food are only just being defined. These may include gut-specific mast cell activation as well as non-specific activation by stressors and certain foods. Visceral hypersensitivity, in some cases mediated by mast cell activation, plays a key part in making otherwise innocuous gut stimuli painful. Rapidly fermented poorly absorbed carbohydrates produce gaseous distension as well as short-chain fatty acids and lowering of colonic pH which may cause symptoms in IBS patients. Limiting intake of these in low FODMAP and related diets has proven popular and apparently successful in many patients. Existing diet, colonic microbiota and their metabolic products may be helpful in predicting who will respond. Wheat intolerance may reflect the fact that wheat is often a major source of dietary FODMAPs. It may also be either a forme fruste of coeliac disease or non-specific immune activation. Wheat exclusion can be successful in some of these patients. More research is needed to fully understand the mechanisms of food intolerances and how to best ameliorate them in a personalised medicine approach to diet in IBS.
Collapse
Affiliation(s)
- Robin Spiller
- NIHR Nottingham Biomedical Research Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
172
|
Liu C, Wu H, Fan H. Progress in understanding of mechanism of dietary therapy for ulcerative colitis with regard to intestinal microbiota. Shijie Huaren Xiaohua Zazhi 2021; 29:146-151. [DOI: 10.11569/wcjd.v29.i3.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of ulcerative colitis is closely related to the complex interaction between heredity, environment, and intestinal microbiota. Intestinal dysbiosis is not only the cause of ulcerative colitis, but also the pathological result of ulcerative colitis. Dietary therapies have been found to modulate the microbiota to alter the effects of environmental factors on ulcerative colitis. Dietary pattern is related to the pathogenesis, development, and prognosis of ulcerative colitis, and the role of diet in ulcerative colitis has attracted more and more attention. This article reviews the mechanisms by which dietary therapy treats ulcerative colitis with regard to regulating the brain-gut functional axis, regulating the immune function, and protecting the intestinal mucosal barrier by modulating intestinal microbiota.
Collapse
Affiliation(s)
- Chang Liu
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Hui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
173
|
Olivier S, Pochard C, Diounou H, Castillo V, Divoux J, Alcantara J, Leclerc J, Guilmeau S, Huet C, Charifi W, Varin TV, Daniel N, Foretz M, Neunlist M, Salomon BL, Ghosh P, Marette A, Rolli-Derkinderen M, Viollet B. Deletion of intestinal epithelial AMP-activated protein kinase alters distal colon permeability but not glucose homeostasis. Mol Metab 2021; 47:101183. [PMID: 33548500 PMCID: PMC7921883 DOI: 10.1016/j.molmet.2021.101183] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/21/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Objective The intestinal epithelial barrier (IEB) restricts the passage of microbes and potentially harmful substances from the lumen through the paracellular space, and rupture of its integrity is associated with a variety of gastrointestinal disorders and extra-digestive diseases. Increased IEB permeability has been linked to disruption of metabolic homeostasis leading to obesity and type 2 diabetes. Interestingly, recent studies have uncovered compelling evidence that the AMP-activated protein kinase (AMPK) signaling pathway plays an important role in maintaining epithelial cell barrier function. However, our understanding of the function of intestinal AMPK in regulating IEB and glucose homeostasis remains sparse. Methods We generated mice lacking the two α1 and α2 AMPK catalytic subunits specifically in intestinal epithelial cells (IEC AMPK KO) and determined the physiological consequences of intestinal-specific deletion of AMPK in response to high-fat diet (HFD)-induced obesity. We combined histological, functional, and integrative analyses to ascertain the effects of gut AMPK loss on intestinal permeability in vivo and ex vivo and on the development of obesity and metabolic dysfunction. We also determined the impact of intestinal AMPK deletion in an inducible mouse model (i-IEC AMPK KO) by measuring IEB function, glucose homeostasis, and the composition of gut microbiota via fecal 16S rRNA sequencing. Results While there were no differences in in vivo intestinal permeability in WT and IEC AMPK KO mice, ex vivo transcellular and paracellular permeability measured in Ussing chambers was significantly increased in the distal colon of IEC AMPK KO mice. This was associated with a reduction in pSer425 GIV phosphorylation, a marker of leaky gut barrier. However, the expression of tight junction proteins in intestinal epithelial cells and pro-inflammatory cytokines in the lamina propria were not different between genotypes. Although the HFD-fed AMPK KO mice displayed suppression of the stress polarity signaling pathway and a concomitant increase in colon permeability, loss of intestinal AMPK did not exacerbate body weight gain or adiposity. Deletion of AMPK was also not sufficient to alter glucose homeostasis or the acute glucose-lowering action of metformin in control diet (CD)- or HFD-fed mice. CD-fed i-IEC AMPK KO mice also presented higher permeability in the distal colon under homeostatic conditions but, surprisingly, this was not detected upon HFD feeding. Alteration in epithelial barrier function in the i-IEC AMPK KO mice was associated with a shift in the gut microbiota composition with higher levels of Clostridiales and Desulfovibrionales. Conclusions Altogether, our results revealed a significant role of intestinal AMPK in maintaining IEB integrity in the distal colon but not in regulating glucose homeostasis. Our data also highlight the complex interaction between gut microbiota and host AMPK. Deletion of intestinal AMPKα1 and α2 suppresses the stress-polarity signaling (SPS) pathway. Loss of the SPS pathway is associated with increased paracellular permeability in the distal colon. Intestinal AMPK is dispensable for the acute glucose-lowering action of metformin. Loss of intestinal AMPK alters the gut microbiota composition.
Collapse
Affiliation(s)
- Séverine Olivier
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Camille Pochard
- University of Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Hanna Diounou
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jordane Divoux
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Joshua Alcantara
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jocelyne Leclerc
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Sandra Guilmeau
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Camille Huet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Wafa Charifi
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Thibault V Varin
- Québec Heart and Lung Research Institute (IUCPQ) & Institute for Nutrition and Functional Foods (INAF), Laval University Québec, Québec, Canada
| | - Noëmie Daniel
- Québec Heart and Lung Research Institute (IUCPQ) & Institute for Nutrition and Functional Foods (INAF), Laval University Québec, Québec, Canada
| | - Marc Foretz
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Michel Neunlist
- University of Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Benoit L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - André Marette
- Québec Heart and Lung Research Institute (IUCPQ) & Institute for Nutrition and Functional Foods (INAF), Laval University Québec, Québec, Canada
| | - Malvyne Rolli-Derkinderen
- University of Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.
| | - Benoit Viollet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France.
| |
Collapse
|
174
|
Impacts of gut microbiota on gestational diabetes mellitus: a comprehensive review. Eur J Nutr 2021; 60:2343-2360. [PMID: 33512587 DOI: 10.1007/s00394-021-02483-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a condition that seriously threatens mother and child health. The incidence of GDM has increased worldwide in the past decades. In addition, the complications of GDM such as type 2 diabetes (T2DM) and neonatal malformations could negatively affect the living quality of mothers and their children. AIM It has been widely known that the imbalance of gut microbiota or called 'gut dysbiosis' plays a key role in the development of insulin resistance and chronic low-grade inflammation in T2DM patients. However, the impacts of gut microbiota on GDM remain controversial. Here, we aim to comprehensively review the alterations of gut microbiota in GDM mothers and their offspring. RESULTS The alterations of Firmicutes/Bacteroidetes (F/B) ratio, short-chain fatty acid (SCFA)-producing bacteria, bacteria with probiotics properties and gram-negative lipopolysaccharide (LPS)-producing bacteria play a vital role in the development of GDM. The beneficial roles of gut microbiota modification (probiotics, synbiotics and lifestyle modification) as a treatment of GDM were found in some, but not all studies. CONCLUSION In the near future, gut microbiota modification may be considered as one of the standard treatments for GDM. Moreover, further studies regarding the specific gut microbiota that are associated with the early development of GDM are required. This may contribute to the novel diagnostic markers for early stages of GDM.
Collapse
|
175
|
Camilleri M. Human Intestinal Barrier: Effects of Stressors, Diet, Prebiotics, and Probiotics. Clin Transl Gastroenterol 2021; 12:e00308. [PMID: 33492118 PMCID: PMC7838004 DOI: 10.14309/ctg.0000000000000308] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
The objectives of this article are to understand the effects of stressors (nonsteroidal antiinflammatory drug, exercise, and pregnancy) and components in the diet, specifically prebiotics and probiotics, on intestinal barrier function. Stressors generally reduce barrier function, and these effects can be reversed by supplements such as zinc or glutamine that are among the substances that enhance the barrier. Other dietary factors in the diet that improve the barrier are vitamins A and D, tryptophan, cysteine, and fiber; by contrast, ethanol, fructose, and dietary emulsifiers increase permeability. Effects of prebiotics on barrier function are modest; on the other hand, probiotics exert direct and indirect antagonism of pathogens, and there are documented effects of diverse probiotic species, especially combination agents, on barrier function in vitro, in vivo in animal studies, and in human randomized controlled trials conducted in response to stress or disease. Clinical observations of benefits with combination probiotics in inflammatory diseases have simultaneously not appraised effects on intestinal permeability. In summary, probiotics and synbiotics enhance intestinal barrier function in response to stressor or disease states. Future studies should address the changes in barrier function and microbiota concomitant with assessment of clinical outcomes.
Collapse
Affiliation(s)
- Michael Camilleri
- Division of Gastroenterology and Hepatology, Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
176
|
Armstrong H, Mander I, Zhang Z, Armstrong D, Wine E. Not All Fibers Are Born Equal; Variable Response to Dietary Fiber Subtypes in IBD. Front Pediatr 2021; 8:620189. [PMID: 33520902 PMCID: PMC7844368 DOI: 10.3389/fped.2020.620189] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Diet provides a safe and attractive alternative to available treatment options in a variety of diseases; however, research has only just begun to elucidate the role of diet in chronic diseases, such as the inflammatory bowel diseases (IBD). The chronic and highly debilitating IBDs, Crohn disease and ulcerative colitis, are hallmarked by intestinal inflammation, immune dysregulation, and dysbiosis; and evidence supports a role for genetics, microbiota, and the environment, including diet, in disease pathogenesis. This is true especially in children with IBD, where diet-based treatments have shown excellent results. One interesting group of dietary factors that readily links microbiota to gut health is dietary fibers. Fibers are not digested by human cells, but rather fermented by the gut microbes within the bowel. Evidence has been mounting over the last decade in support of the importance of dietary fibers in the maintenance of gut health and in IBD; however, more recent studies highlight the complexity of this interaction and importance of understanding the role of each individual dietary fiber subtype, especially during disease. There are roughly ten subtypes of dietary fibers described to date, categorized as soluble or insoluble, with varying chemical structures, and large differences in their fermentation profiles. Many studies to date have described the benefits of the byproducts of fermentation in healthy individuals and the potential health benefits in select disease models. However, there remains a void in our understanding of how each of these individual fibers affect human health in dysbiotic settings where appropriate fermentation may not be achieved. This review highlights the possibilities for better defining the role of individual dietary fibers for use in regulating inflammation in IBD.
Collapse
Affiliation(s)
- Heather Armstrong
- Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Inderdeep Mander
- Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, AB, Canada
| | - Zhengxiao Zhang
- Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - David Armstrong
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Eytan Wine
- Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
177
|
Fischer C, Kleinschmidt T. Synthesis of galactooligosaccharides by Cryptococcus laurentii and Aspergillus oryzae using different kinds of acid whey. Int Dairy J 2021. [DOI: 10.1016/j.idairyj.2020.104867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
178
|
Tang G, Zhang L. Update on Strategies of Probiotics for the Prevention and Treatment of Colorectal Cancer. Nutr Cancer 2020; 74:27-38. [PMID: 33356609 DOI: 10.1080/01635581.2020.1865420] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, with the further research on probiotics, probiotics may become an indispensable part in the prevention and treatment of colorectal cancer (CRC) in the future. As one of the most common cancer, the incidence of CRC is still rising in developing countries. Nowadays, there are lacking in prevention methods with low side effect. Surgery and chemotherapy, as the main treatment of CRC, bring many complications and affect the quality of life of patients. Probiotics has provided new ideas to solve these problems. Probiotics have anti-inflammatory, immune-enhancing, tumor-suppressing and other beneficial effects. Probiotics may provide some safe and effective prevention strategies for CRC. In addition, probiotics can also reduce the complications of surgery and chemotherapy, and improve the effectiveness of chemotherapy. Target administration with probiotics or probiotics cooperated with TRAIL to treat CRC. This article aims to review the mechanisms of probiotics for the prevention and treatment of CRC, as well as specific ways to use probiotics, in order to provide more new strategies for the prevention and treatment of CRC in the future, and reduce the incidence of and improve the quality of life of patients.
Collapse
Affiliation(s)
- Gang Tang
- Department of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Linyu Zhang
- Department of Clinical Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
179
|
Protection of Galacto-Oligosaccharide against E. coli O157 Colonization through Enhancing Gut Barrier Function and Modulating Gut Microbiota. Foods 2020; 9:foods9111710. [PMID: 33233359 PMCID: PMC7700679 DOI: 10.3390/foods9111710] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Galacto-oligosaccharide (GOS) has been added to infant formula as prebiotics and can bring many benefits to human health. This study proved the effect of GOS in prevention and alleviation against E. coli O157 invasion and colonization and the mechanism behind this was explored in a mice model. The results showed that the expression of Muc2 and Occlaudin were both significantly down-regulated (p < 0.05) by E. coli O157 infection, while GOS alleviated this phenomenon, which means that GOS can reduce the colonization of E. coli O157 by enhancing the gut barrier function. Through the determination of inflammatory cytokines, we found that GOS can relieve inflammation caused by pathogens. At the same time, GOS can promote the growth of probiotics such as Akkermansia, Ruminococcaceae and Bacteroides, thus modulating microorganism environments and improving short chain fatty acid (SCFA) levels in the intestine. This study provides an explanation for the mechanism behind the protection of GOS against pathogen infection.
Collapse
|
180
|
Study on the additive protective effect of PGLYRP3 and Bifidobacterium adolescentis Reuter 1963 on severity of DSS-induced colitis in Pglyrp3 knockout (Pglyrp3 -/-) and wild-type (WT) mice. Immunobiology 2020; 226:152028. [PMID: 33242664 DOI: 10.1016/j.imbio.2020.152028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/05/2020] [Accepted: 11/02/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Pglyrp3 is a bactericidal innate immunity protein known to sustain the habitual gut microbiome and protect against experimental colitis. Intestinal inflammation and metaflammation are commonly associated with a marked reduction of commensal bifidobacteria. Whether Pglyrp3 and bifidobacteria interact synergistically or additively to alleviate metaflammation is unknown. We investigated the extent to which Pglyrp3 and bifidobacteria regulate metaflammation and gut bacterial dysbiosis in DSS-induced mouse models of intestinal inflammation. MATERIAL & METHODS 8-10 weeks old male mice were used. In both WT and Pglyrp3 -/- experiments, the mice were randomly divided into three groups of 16 mice per group: (1) a control group receiving sterile tap water, (2) an experimental group receiving sterile tap water supplemented with only 5% DSS, and (3) an experimental group receiving sterile tap water supplemented with 5% DSS and 1 × 109 CFU/ml of Bifidobacterium adolescentis (B.a.) for 7 days. Wild-type (WT) littermates of the respective gene (i.e. Pglyrp3) were used as controls throughout the study. Clinical signs of general health and inflammation were monitored daily. Faecal pellet samples were analysed by qRT-PCR for microbial composition. Histology of relevant organs was carried out on day 8. Metabolic parameters and liver inflammation were determined in serum samples. RESULTS Intestinal inflammation in mice of group 2 were significantly increased compared to those of control group 1. There was a significant difference in mean scores for inflammation severity between DSS-treated WT and DSS-treated Pglyrp3 -/- mice. Buildup of key serum metabolic markers (cholesterol, triglyceride and glucose) was set off by colonic inflammation. qRT-PCR quantification showed that DSS significantly decreased the Clostridium coccoides and Bifidobacterium cell counts while increasing those of Bacteroides group in both WT and Pglyrp3 -/- mice. These manifestations of DSS-induced dysbiosis were significantly attenuated by feeding B.a. Both the local and systemic ill-being of the mice alleviated when they received B.a. DISCUSSION This study shows that Pglyrp3 facilitates recognition of bifidobacterial cell wall-derived peptidoglycan, thus leading additively to a reduction of metaflammation through an increase in the number of bifidobacteria, which were able to mitigate intestinal immunopathology in the context of Pglyrp3 blockade.
Collapse
|
181
|
Ma C, Wasti S, Huang S, Zhang Z, Mishra R, Jiang S, You Z, Wu Y, Chang H, Wang Y, Huo D, Li C, Sun Z, Sun Z, Zhang J. The gut microbiome stability is altered by probiotic ingestion and improved by the continuous supplementation of galactooligosaccharide. Gut Microbes 2020; 12:1785252. [PMID: 32663059 PMCID: PMC7524268 DOI: 10.1080/19490976.2020.1785252] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The stable gut microbiome plays a key role in sustaining host health, while the instability of gut microbiome also has been found to be a risk factor of various metabolic diseases. At the ecological and evolutionary scales, the inevitable competition between the ingested probiotic and indigenous gut microbiome can lead to an increase in the instability. It remains largely unclear if and how exogenous prebiotic can improve the overall gut microbiome stability in probiotic consumption. In this study, we used Lactobacillus plantarum HNU082 (Lp082) as a model probiotic to examine the impact of the continuous or pulsed supplementation of galactooligosaccharide (GOS) on the gut microbiome stability in mice using shotgun metagenomic sequencing. Only continuous GOS supplement promoted the growth of probiotic and decreased its single-nucleotide polymorphisms (SNPs) mutation under competitive conditions. Besides, persistent GOS supplementation increased the overall stability, reshaped the probiotic competitive interactions with Bacteroides species in the indigenous microbiome, which was also evident by over-abundance of carbohydrate-active enzymes (CAZymes) accordingly. Also, we identified a total of 793 SNPs arisen in probiotic administration in the indigenous microbiome. Over 90% of them derived from Bacteroides species, which involved genes encoding transposase, CAZymes, and membrane proteins. However, neither GOS supplementation here de-escalated the overall adaptive mutations within the indigenous microbes during probiotic intake. Collectively, our study demonstrated the beneficial effect of continuous prebiotic supplementation on the ecological and genetic stability of gut microbiomes.
Collapse
Affiliation(s)
- Chenchen Ma
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Sanjeev Wasti
- Department of Human Nutrition, Food and Animal Science, University of Hawaii, Honolulu, HI, USA
| | - Shi Huang
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Zeng Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Rajeev Mishra
- Department of Human Nutrition, Food and Animal Science, University of Hawaii, Honolulu, HI, USA
| | - Shuaiming Jiang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Zhengkai You
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Yixuan Wu
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Haibo Chang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Yuanyuan Wang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Dongxue Huo
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Congfa Li
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education P. R. C., Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs China, Inner Mongolia Agricultural University, Hohhot, China,Zhihong Sun Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education P. R. C., Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs China, Inner Mongolia Agricultural University, Hohhot, Inner mongolia Autonomous Region, 010018, China
| | - Zheng Sun
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China,Zheng Sun Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
| | - Jiachao Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China,CONTACT Jiachao Zhang College of Food Science and Engineering, Hainan University, Haikou, Hainan570228, China
| |
Collapse
|
182
|
Binienda A, Twardowska A, Makaro A, Salaga M. Dietary Carbohydrates and Lipids in the Pathogenesis of Leaky Gut Syndrome: An Overview. Int J Mol Sci 2020; 21:ijms21218368. [PMID: 33171587 PMCID: PMC7664638 DOI: 10.3390/ijms21218368] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
This review summarizes the recent knowledge on the effects of dietary carbohydrates and lipids on the pathophysiology of leaky gut syndrome (LGS). Alterations in intestinal barrier permeability may lead to serious gastrointestinal (GI) disorders. LGS is caused by intestinal hyperpermeability due to changes in the expression levels and functioning of tight junctions. The influence of dietary habits on intestinal physiology is clearly visible in incidence rates of intestinal diseases in industrial and developing countries. Diseases which are linked to intestinal hyperpermeability tend to localize to Westernized countries, where a diet rich in fats and refined carbohydrates predominates. Several studies suggest that fructose is one of the key carbohydrates involved in the regulation of the intestinal permeability and its overuse may cause harmful effects, such as tight junction protein dysfunction. On the other hand, short chain fatty acids (mainly butyrate) at appropriate concentrations may lead to the reduction of intestinal permeability, which is beneficial in LGS. However, long chain fatty acids, including n-3 and n-6 polyunsaturated fatty acids have unclear properties. Some of those behave as components untightening and tightening the intestinal membrane.
Collapse
Affiliation(s)
| | | | | | - Maciej Salaga
- Correspondence: ; Tel.: +48-42-272-57-07; Fax: +48-42-272-56-94
| |
Collapse
|
183
|
Sharma L, Riva A. Intestinal Barrier Function in Health and Disease-Any role of SARS-CoV-2? Microorganisms 2020; 8:E1744. [PMID: 33172188 PMCID: PMC7694956 DOI: 10.3390/microorganisms8111744] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Alterations in the structure and function of the intestinal barrier play a role in the pathogenesis of a multitude of diseases. During the recent and ongoing coronavirus disease (COVID-19) pandemic, it has become clear that the gastrointestinal system and the gut barrier may be affected by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, and disruption of barrier functions or intestinal microbial dysbiosis may have an impact on the progression and severity of this new disease. In this review, we aim to provide an overview of current evidence on the involvement of gut alterations in human disease including COVID-19, with a prospective outlook on supportive therapeutic strategies that may be investigated to rescue intestinal barrier functions and possibly facilitate clinical improvement in these patients.
Collapse
Affiliation(s)
- Lakshya Sharma
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK;
| | - Antonio Riva
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK;
- Foundation for Liver Research, Institute of Hepatology, London SE5 9NT, UK
| |
Collapse
|
184
|
Pujari R, Banerjee G. Impact of prebiotics on immune response: from the bench to the clinic. Immunol Cell Biol 2020; 99:255-273. [PMID: 32996638 DOI: 10.1111/imcb.12409] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/31/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
Several preclinical and clinical studies have shown the immunomodulatory role exerted by prebiotics in regulating the immune response. In this review, we describe the mechanistic and clinical studies that decipher the cell signaling pathways implicated in the process. Prebiotic fibers are conventionally known to serve as substrate for probiotic commensal bacteria that release of short-chain fatty acids in the intestinal tract along with several other metabolites. Subsequently, they then act on the local as well as the systemic immune cells and the gut-associated epithelial cells, primarily through G-protein-coupled receptor-mediated pathways. However, other pathways including histone deacetylase inhibition and inflammasome pathway have also been implicated in regulating the immunomodulatory effect. The prebiotics can also induce a microbiota-independent effect by directly acting on the gut-associated epithelial and innate immune cells through the Toll-like receptors. The cumulative effect results in the maintenance of the epithelial barrier integrity and modulation of innate immunity through secretion of pro- and anti-inflammatory cytokines, switches in macrophage polarization and function, neutrophil recruitment and migration, dendritic cell and regulatory T-cell differentiation. Extending these in vitro and ex vivo observations, some prebiotics have been well investigated, with successful human and animal trials demonstrating the association between gut microbes and immunity biomarkers leading to improvement in health endpoints across populations. This review discusses scientific insights into the association between prebiotics, innate immunity and gut microbiome from in vitro to human oral intervention.
Collapse
Affiliation(s)
- Radha Pujari
- Innovation Centre, Tata Chemicals Ltd, Pune, Maharashtra, India
| | - Gautam Banerjee
- Innovation Centre, Tata Chemicals Ltd, Pune, Maharashtra, India
| |
Collapse
|
185
|
Khoshbin K, Camilleri M. Effects of dietary components on intestinal permeability in health and disease. Am J Physiol Gastrointest Liver Physiol 2020; 319:G589-G608. [PMID: 32902315 PMCID: PMC8087346 DOI: 10.1152/ajpgi.00245.2020] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Altered intestinal permeability plays a role in many pathological conditions. Intestinal permeability is a component of the intestinal barrier. This barrier is a dynamic interface between the body and the food and pathogens that enter the gastrointestinal tract. Therefore, dietary components can directly affect this interface, and many metabolites produced by the host enzymes or the gut microbiota can act as signaling molecules or exert direct effects on this barrier. Our aim was to examine the effects of diet components on the intestinal barrier in health and disease states. Herein, we conducted an in-depth PubMed search based on specific key words (diet, permeability, barrier, health, disease, and disorder), as well as cross references from those articles. The normal intestinal barrier consists of multiple components in the lumen, epithelial cell layer and the lamina propria. Diverse methods are available to measure intestinal permeability. We focus predominantly on human in vivo studies, and the literature is reviewed to identify dietary factors that decrease (e.g., emulsifiers, surfactants, and alcohol) or increase (e.g., fiber, short-chain fatty acids, glutamine, and vitamin D) barrier integrity. Effects of these dietary items in disease states, such as metabolic syndrome, liver disease, or colitis are documented as examples of barrier dysfunction in the multifactorial diseases. Effects of diet on intestinal barrier function are associated with precise mechanisms in some instances; further research of those mechanisms has potential to clarify the role of dietary interventions in treating diverse pathologic states.
Collapse
Affiliation(s)
- Katayoun Khoshbin
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Mayo Clinic, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
186
|
Kim MG, Jo K, Chang YB, Suh HJ, Hong KB. Changes in the Gut Microbiome after Galacto-Oligosaccharide Administration in Loperamide-Induced Constipation. J Pers Med 2020; 10:jpm10040161. [PMID: 33050434 PMCID: PMC7711924 DOI: 10.3390/jpm10040161] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 01/02/2023] Open
Abstract
Unbalanced dietary habits and the consumption of high protein and instant foods cause an increase in constipation. Here, we evaluated the effects of galacto-oligosaccharide (GOS) on a rat model of loperamide-induced constipation by measuring various biological markers and cecal microbiota. The fecal water content and intestinal transit ratio significantly increased in the GOS-administered (GL and GH) groups than in the control group (p < 0.05, p < 0.01, and p < 0.001, respectively). The length of intestinal mucosa (p < 0.05 and p < 0.01, respectively) and area of crypt cells were (p < 0.01, both) significantly increased in the GOS-administered groups compared to the control group. The distribution of interstitial cells of Cajal, which is related to the intestinal movement, showed a significant increase in GOS-administered groups than in the control group (p < 0.01, both). The relative abundance of lactic acid bacteria (LAB), especially Lactobacillus and Lactococcus, significantly increased in the GL group than in the control group. Furthermore, there was a positive correlation between short chain fatty acids (SCFAs) and the gut microbiota in the GL groups. These results demonstrated that GOS administration effectively alleviates constipation by increasing LAB proliferation in the intestinal microbiota and SCFA production.
Collapse
Affiliation(s)
| | | | | | | | - Ki-Bae Hong
- Correspondence: ; Tel.: +82-2-940-2853; Fax: +82-2-921-7207
| |
Collapse
|
187
|
Han D, Li Z, Liu T, Yang N, Li Y, He J, Qian M, Kuang Z, Zhang W, Ni C, Guo X. Prebiotics Regulation of Intestinal Microbiota Attenuates Cognitive Dysfunction Induced by Surgery Stimulation in APP/PS1 Mice. Aging Dis 2020; 11:1029-1045. [PMID: 33014520 PMCID: PMC7505279 DOI: 10.14336/ad.2020.0106] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence indicates that the intestinal microbiota could interact with the central nervous system and modulate multiple pathophysiological changes, including the integrity of intestinal barrier and blood-brain barrier, as well as neuroinflammatory response. In the present study, we investigated the potential role of intestinal microbiota in the pathophysiological process of postoperative cognitive dysfunction. Six-month-old APP/PS1 mice were subjected to partial hepatectomy to establish surgery model and exhibited cognitive dysfunction. The expressions of inflammatory mediators increased and tight junction proteins (ZO-1 and Occludin) levels decreased in the intestine and hippocampus. The 16S ribosomal RNA gene sequencing showed altered β diversity and intestinal microbiota richness after surgery, including genus Rodentibacter, Bacteroides, Ruminococcaceae_UCG_014 and Faecalibaculum, as well as family Eggerthellaceae and Muribaculaceae. Furthermore, prebiotics (Xylooligosaccharides, XOS) intervention effectively attenuated surgery-induced cognitive dysfunction and intestinal microbiota alteration, reduced inflammatory responses, and improved the integrity of tight junction barrier in the intestine and hippocampus. In summary, the present study indicates that intestinal microbiota alteration, the related intestinal barrier and blood-brain barrier damage, and inflammatory responses participate the pathophysiological process of postoperative cognitive dysfunction. Prebiotics intervention could be a potential preventative approach.
Collapse
Affiliation(s)
- Dengyang Han
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhengqian Li
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Taotao Liu
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ning Yang
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yue Li
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jindan He
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Min Qian
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhongshen Kuang
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Wen Zhang
- 2National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Cheng Ni
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiangyang Guo
- 1Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
188
|
Šuligoj T, Vigsnæs LK, den Abbeele PV, Apostolou A, Karalis K, Savva GM, McConnell B, Juge N. Effects of Human Milk Oligosaccharides on the Adult Gut Microbiota and Barrier Function. Nutrients 2020; 12:E2808. [PMID: 32933181 PMCID: PMC7551690 DOI: 10.3390/nu12092808] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Human milk oligosaccharides (HMOs) shape the gut microbiota in infants by selectively stimulating the growth of bifidobacteria. Here, we investigated the impact of HMOs on adult gut microbiota and gut barrier function using the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), Caco2 cell lines, and human intestinal gut organoid-on-chips. We showed that fermentation of 2'-O-fucosyllactose (2'FL), lacto-N-neotetraose (LNnT), and combinations thereof (MIX) led to an increase of bifidobacteria, accompanied by an increase of short chain fatty acid (SCFA), in particular butyrate with 2'FL. A significant reduction in paracellular permeability of FITC-dextran probe was observed using Caco2 cell monolayers with fermented 2'FL and MIX, which was accompanied by an increase in claudin-8 gene expression as shown by qPCR, and a reduction in IL-6 as determined by multiplex ELISA. Using gut-on-chips generated from human organoids derived from proximal, transverse, and distal colon biopsies (Colon Intestine Chips), we showed that claudin-5 was significantly upregulated across all three gut-on-chips following treatment with fermented 2'FL under microfluidic conditions. Taken together, these data show that, in addition to their bifidogenic activity, HMOs have the capacity to modulate immune function and the gut barrier, supporting the potential of HMOs to provide health benefits in adults.
Collapse
Affiliation(s)
- Tanja Šuligoj
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich NR4 7UQ, UK;
| | | | | | - Athanasia Apostolou
- Emulate Inc., 27 Drydock Ave, Boston, MA 02210, USA; (A.A.); (K.K.)
- Graduate Program, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Katia Karalis
- Emulate Inc., 27 Drydock Ave, Boston, MA 02210, USA; (A.A.); (K.K.)
| | - George M. Savva
- Quadram Institute Bioscience, Core Sciences Resources, Norwich Research Park, Norwich NR5 7UQ, UK;
| | - Bruce McConnell
- Glycom A/S, Kogle Allé 4, DK-2970 Hørsholm, Denmark; (L.K.V.); (B.M.)
| | - Nathalie Juge
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich NR4 7UQ, UK;
| |
Collapse
|
189
|
Aponte M, Murru N, Shoukat M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front Microbiol 2020; 11:562048. [PMID: 33042069 PMCID: PMC7516994 DOI: 10.3389/fmicb.2020.562048] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Probiotics are considered as the twenty-first century panpharmacon due to their competent remedial power to cure from gastrointestinal dysbiosis, systematic metabolic diseases, and genetic impairments up to complicated neurodegenerative disorders. They paved the way for an innovative managing of various severe diseases through palatable food products. The probiotics' role as a "bio-therapy" increased their significance in food and medicine due to many competitive advantages over traditional treatment therapies. Their prophylactic and therapeutic potential has been assessed through hundreds of preclinical and clinical studies. In addition, the food industry employs probiotics as functional and nutraceutical ingredients to enhance the added value of food product in terms of increased health benefits. However, regardless of promising health-boosting effects, the probiotics' efficacy still needs an in-depth understanding of systematic mechanisms and factors supporting the healthy actions.
Collapse
Affiliation(s)
- Maria Aponte
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Nicoletta Murru
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Mahtab Shoukat
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| |
Collapse
|
190
|
Vallianou N, Stratigou T, Christodoulatos GS, Tsigalou C, Dalamaga M. Probiotics, Prebiotics, Synbiotics, Postbiotics, and Obesity: Current Evidence, Controversies, and Perspectives. Curr Obes Rep 2020; 9:179-192. [PMID: 32472285 DOI: 10.1007/s13679-020-00379-w] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW In this review, we summarize current evidence on gut microbiome and obesity; we discuss the role of probiotics, prebiotics, synbiotics, and postbiotics in obesity prevention and management; and we highlight and analyze main limitations, challenges, and controversies of their use. RECENT FINDINGS Overall, the majority of animal studies and meta-analyses of human studies examining the use of probiotics and synbiotics in obesity has shown their beneficial effects on weight reduction and other metabolic parameters via their involvement in gut microbiota modulation. Bifidobacterium and Lactobacillus strains are still the most widely used probiotics in functional foods and dietary supplements, but next generation probiotics, such as Faecalibacterium prausnitzii, Akkermansia muciniphila, or Clostridia strains, have demonstrated promising results. On the contrary, meta-analyses of human studies on the use of prebiotics in obesity have yielded contradictory results. In animal studies, postbiotics, mainly short-chain fatty acids, may increase energy expenditure through induction of thermogenesis in brown adipose tissue as well as browning of the white adipose tissue. The main limitations of studies on biotics in obesity include the paucity of human studies; heterogeneity among the studied subgroups regarding age, gender, and lifestyle; and use of different agents with potential therapeutic effects in different formulations, doses, ratio and different pharmacodynamics/pharmacokinetics. In terms of safety, the supplementation with prebiotics, probiotics, and synbiotics has not been associated with serious adverse effects among immune-competent individuals, with the exception of the use of probiotics and synbiotics in immunocompromised patients. Further large-scale Randomized Controlled Trials (RCTs) in humans are required to evaluate the beneficial properties of probiotics, prebiotics, synbiotics, and postbiotics; their ideal dose; the duration of supplementation; and the durability of their beneficial effects as well as their safety profile in the prevention and management of obesity.
Collapse
Affiliation(s)
- Natalia Vallianou
- Department of Endocrinology, 'Evangelismos' General Hospital of Athens, 45-47 Ypsilantou street, 10676, Athens, Greece.
| | - Theodora Stratigou
- Department of Endocrinology, 'Evangelismos' General Hospital of Athens, 45-47 Ypsilantou street, 10676, Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Christina Tsigalou
- Laboratory of Microbiology, Medical School, Democritus University of Thrace, 6th Km Alexandroupolis-Makri, Alexandroupolis, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| |
Collapse
|
191
|
Ashaolu TJ. Immune boosting functional foods and their mechanisms: A critical evaluation of probiotics and prebiotics. Biomed Pharmacother 2020; 130:110625. [PMID: 32795926 DOI: 10.1016/j.biopha.2020.110625] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Comprehensive studies conducted on the link between the gut microbiome and immunity in recent decades have correspondingly led to ever increasing interests in functional foods, especially probiotics and prebiotics. Probiotics and prebiotics play crucial roles in managing the intestinal microbiota in order to improve host health, even though their influence on other body sites are being investigated. Different colonic bacteria metabolize dietary prebiotics to produce beneficial metabolites, especially short chain fatty acids (SCFAs) that improve luminal contents and intestinal performance, while positively affecting overall host physiology. Thus, this review provides a general perspective of the immune system, the gut immune system and its microbiota. The review also evaluates functional foods with critical but comprehensive perspectives into probiotics and prebiotics, their immune boosting and mechanisms of action. It is recommended that further mechanistic and translational studies are conducted to promote health, social life and also empower poverty-stricken communities.
Collapse
Affiliation(s)
- Tolulope Joshua Ashaolu
- Smart Agriculture Research and Application Team, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
192
|
Perillo F, Amoroso C, Strati F, Giuffrè MR, Díaz-Basabe A, Lattanzi G, Facciotti F. Gut Microbiota Manipulation as a Tool for Colorectal Cancer Management: Recent Advances in Its Use for Therapeutic Purposes. Int J Mol Sci 2020; 21:E5389. [PMID: 32751239 PMCID: PMC7432108 DOI: 10.3390/ijms21155389] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease influenced by both environmental and genetic factors. A large body of literature has demonstrated the role of gut microbes in promoting inflammatory responses, creating a suitable microenvironment for the development of skewed interactions between the host and the gut microbiota and cancer initiation. Even if surgery is the primary therapeutic strategy, patients with advanced disease or cancer recurrence after surgery remain difficult to cure. Therefore, the gut microbiota has been proposed as a novel therapeutic target in light of recent promising data in which it seems to modulate the response to cancer immunotherapy. The use of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics, and fecal microbiota transplantation, is therefore considered to support current therapies in CRC management. In this review, we will discuss the importance of host-microbe interactions in CRC and how promoting homeostatic immune responses through microbe-targeted therapies may be useful in preventing/treating CRC development.
Collapse
Affiliation(s)
- Federica Perillo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Chiara Amoroso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Francesco Strati
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Maria Rita Giuffrè
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Angélica Díaz-Basabe
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20135 Milan, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20135 Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| |
Collapse
|
193
|
Nishikawa H, Enomoto H, Nishiguchi S, Iijima H. Liver Cirrhosis and Sarcopenia from the Viewpoint of Dysbiosis. Int J Mol Sci 2020; 21:5254. [PMID: 32722100 PMCID: PMC7432211 DOI: 10.3390/ijms21155254] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Sarcopenia in patients with liver cirrhosis (LC) has been attracting much attention these days because of the close linkage to adverse outcomes. LC can be related to secondary sarcopenia due to protein metabolic disorders and energy metabolic disorders. LC is associated with profound alterations in gut microbiota and injuries at the different levels of defensive mechanisms of the intestinal barrier. Dysbiosis refers to a state in which the diversity of gut microbiota is decreased by decreasing the bacterial species and the number of bacteria that compose the gut microbiota. The severe disturbance of intestinal barrier in LC can result in dysbiosis, several bacterial infections, LC-related complications, and sarcopenia. Here in this review, we will summarize the current knowledge of the relationship between sarcopenia and dysbiosis in patients with LC.
Collapse
Affiliation(s)
- Hiroki Nishikawa
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hyogo College of Medicine, Nishinomiya 6638136, Japan; (H.E.); (H.I.)
- Center for Clinical Research and Education, Hyogo College of Medicine, Nishinomiya 6638136, Japan
| | - Hirayuki Enomoto
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hyogo College of Medicine, Nishinomiya 6638136, Japan; (H.E.); (H.I.)
| | | | - Hiroko Iijima
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hyogo College of Medicine, Nishinomiya 6638136, Japan; (H.E.); (H.I.)
| |
Collapse
|
194
|
Machado AS, Oliveira JR, Lelis DDF, de Paula AMB, Guimarães ALS, Andrade JMO, Brandi IV, Santos SHS. Oral Probiotic Bifidobacterium Longum Supplementation Improves Metabolic Parameters and Alters the Expression of the Renin-Angiotensin System in Obese Mice Liver. Biol Res Nurs 2020; 23:100-108. [PMID: 32700545 DOI: 10.1177/1099800420942942] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Obesity and non-alcoholic fatty liver disease (NAFLD) have been increasing at an alarming rate worldwide. Bifidobacterium longum (BL), a common member of the human gut microbiota, has important health benefits through several mechanisms. OBJECTIVES We evaluated the BL supplementation effects on body metabolism and renin-angiotensin components hepatic expression in mice fed a high-fat diet. METHODS Thirty-two male mice were divided into four groups: standard diet + placebo (ST), standard diet + Bifidobacterium longum (ST + BL), high-fat diet + placebo (HFD) and high-fat diet + Bifidobacterium longum (HFD + BL). Following the obesity induction period, the ST + BL and HFD + BL groups were supplemented with Bifidobacterium longum for 4 weeks. Then, body, biochemical, histological and molecular parameters were evaluated. RESULTS HFD + BL mice had a significant decrease in adipose tissue mass and blood glucose levels, as well as a significant reduction in blood glucose during an intraperitoneal glucose tolerance test. The treatment also resulted in reduced levels of total cholesterol and hepatic fat accumulation. Moreover, we observed an increase in angiotensin converting enzyme 2 (ACE2) and Mas receptor (MASR) expression levels in BL-treated obese mice. CONCLUSIONS These data demonstrate that BL may have the potential to prevent obesity and NAFLD by modulating the mRNA expression of renin-angiotensin system components.
Collapse
Affiliation(s)
- Amanda S Machado
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Janaína R Oliveira
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Deborah de F Lelis
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Alfredo M B de Paula
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - André L S Guimarães
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - João M O Andrade
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Igor V Brandi
- Institute of Agricultural Sciences, Food Engineering, 28114Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Sérgio H S Santos
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil.,Institute of Agricultural Sciences, Food Engineering, 28114Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
195
|
Saresella M, Marventano I, Barone M, La Rosa F, Piancone F, Mendozzi L, d'Arma A, Rossi V, Pugnetti L, Roda G, Casagni E, Cas MD, Paroni R, Brigidi P, Turroni S, Clerici M. Alterations in Circulating Fatty Acid Are Associated With Gut Microbiota Dysbiosis and Inflammation in Multiple Sclerosis. Front Immunol 2020; 11:1390. [PMID: 32733460 PMCID: PMC7358580 DOI: 10.3389/fimmu.2020.01390] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Butyric acid (BA) is a short-chain fatty acid (SCFA) with anti-inflammatory properties, which promotes intestinal barrier function. Medium-chain fatty acids (MCFA), including caproic acid (CA), promote TH1 and TH17 differentiation, thus supporting inflammation. Aim: Since most SCFAs are absorbed in the cecum and colon, the measurement of BA in peripheral blood could provide information on the health status of the intestinal ecosystem. Additionally, given the different immunomodulatory properties of BA and CA the evaluation of their serum concentration, as well as their ratio could be as a simple and rapid biomarker of disease activity and/or treatment efficacy in MS. Methods: We evaluated serum BA and CA concentrations, immune parameters, intestinal barrier integrity and the gut microbiota composition in patients with multiple sclerosis (MS) comparing result to those obtained in healthy controls. Results: In MS, the concentration of BA was reduced and that of CA was increased. Concurrently, the microbiota was depleted of BA producers while it was enriched in mucin-degrading, pro-inflammatory components. The reduced serum concentration of BA seen in MS patients correlated with alterations of the barrier permeability, as evidenced by the higher plasma concentrations of lipopolysaccharide and intestinal fatty acid-binding protein, and inflammation. Specifically, CA was positively associated with CD4+/IFNγ+ T lymphocytes, and the BA/CA ratio correlated positively with CD4+/CD25high/Foxp3+ and negatively with CD4+/IFNγ+ T lymphocytes. Conclusion: The gut microbiota dysbiosis found in MS is possibly associated with alterations of the SCFA/MCFA ratio and of the intestinal barrier; this could explain the chronic inflammation that characterizes this disease. SCFA and MCFA quantification could be a simple biomarker to evaluate the efficacy of therapeutic and rehabilitation procedures in MS.
Collapse
Affiliation(s)
| | | | - Monica Barone
- Microbial Ecology of Health Unit, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | - Gabriella Roda
- Departments of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Eleonora Casagni
- Departments of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | - Rita Paroni
- Health Sciences, University of Milan, Milan, Italy
| | - Patrizia Brigidi
- Microbial Ecology of Health Unit, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Microbial Ecology of Health Unit, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Pathophysiology and Trasplantation, University of Milan, Milan, Italy
| |
Collapse
|
196
|
Structure and Function of Bovine Whey Derived Oligosaccharides Showing Synbiotic Epithelial Barrier Protective Properties. Nutrients 2020; 12:nu12072007. [PMID: 32640639 PMCID: PMC7400958 DOI: 10.3390/nu12072007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Commensal gut microbiota and probiotics have numerous effects on the host’s metabolic and protective systems, which occur primarily through the intestinal epithelial cell interface. Prebiotics, like galacto-oligosaccharides (GOS) are widely used to modulate their function and abundance. However, important structure–function relations may exist, requiring a detailed structural characterization. Here, we detailed the structural characterization of bovine whey derived oligosaccharide preparations enriched with GOS or not, dubbed GOS-enriched milk oligosaccharides (GMOS) or MOS, respectively. We explore GMOS’s and MOS’s potential to improve intestinal epithelial barrier function, assessed in a model based on barrier disruptive effects of the Clostridioides difficile toxin A. GMOS and MOS contain mainly GOS species composed of β1-6- and β1-3-linked galactoses, and 3′- and 6′-sialyllactose. Both GMOS and MOS, combined with lactobacilli, like Lactobacillus rhamnosus (LPR, NCC4007), gave synergistic epithelial barrier protection, while no such effect was observed with Bifidobacterium longum (BL NCC3001), Escherichia coli (Nissle) or fructo-oligosaccharides. Mechanistically, for barrier protection with MOS, (i) viable LPR was required, (ii) acidification of growth medium was not enough, (iii) LPR did not directly neutralize toxin A, and (iv) physical proximity of LPR with the intestinal epithelial cells was necessary. This is the first study, highlighting the importance of structure–function specificity and the necessity of the simultaneous presence of prebiotic, probiotic and host cell interactions required for a biological effect.
Collapse
|
197
|
Aoun A, Darwish F, Hamod N. The Influence of the Gut Microbiome on Obesity in Adults and the Role of Probiotics, Prebiotics, and Synbiotics for Weight Loss. Prev Nutr Food Sci 2020; 25:113-123. [PMID: 32676461 PMCID: PMC7333005 DOI: 10.3746/pnf.2020.25.2.113] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022] Open
Abstract
The link between the gut microbiome and obesity is not well defined. Understanding of the role of the gut microbiome in weight and health management may lead to future revolutionary changes for treating obesity. This review examined the relationship between obesity and the gut microbiome, and the role of probiotics, prebiotics, and synbiotics for preventing and treating obesity. We used PubMed and Google Scholar to collect appropriate articles for the review. We showed that the gut microbiome has an impact on nutrient metabolism and energy expenditure. Moreover, different modalities of obesity treatment have been shown to change the diversity and composition of the gut microbiome; this raises questions about the role these changes may play in weight loss. In addition, studies have shown that supplementation with probiotics, prebiotics, and synbiotics may alter the secretion of hormones, neurotransmitters, and inflammatory factors, thus preventing food intake triggers that lead to weight gain. Further clinical studies are needed to better understand how different species of bacteria in the gut microbiome may affect weight gain, and to determine the most appropriate doses, compositions, and regimens of probiotics, prebiotics, and synbiotics supplementation for long-term weight control.
Collapse
Affiliation(s)
- Antoine Aoun
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Zouk Mosbeh 72, Lebanon
| | - Fatima Darwish
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Zouk Mosbeh 72, Lebanon
| | - Natacha Hamod
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Zouk Mosbeh 72, Lebanon
| |
Collapse
|
198
|
Agans RT, Giles GE, Goodson MS, Karl JP, Leyh S, Mumy KL, Racicot K, Soares JW. Evaluation of Probiotics for Warfighter Health and Performance. Front Nutr 2020; 7:70. [PMID: 32582752 PMCID: PMC7296105 DOI: 10.3389/fnut.2020.00070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
The probiotic industry continues to grow in both usage and the diversity of products available. Scientific evidence supports clinical use of some probiotic strains for certain gastrointestinal indications. Although much less is known about the impact of probiotics in healthy populations, there is increasing consumer and scientific interest in using probiotics to promote physical and psychological health and performance. Military men and women are a unique healthy population that must maintain physical and psychological health in order to ensure mission success. In this narrative review, we examine the evidence regarding probiotics and candidate probiotics for physical and/or cognitive benefits in healthy adults within the context of potential applications for military personnel. The reviewed evidence suggests potential for certain strains to induce biophysiological changes that may offer physical and/or cognitive health and performance benefits in military populations. However, many knowledge gaps exist, effects on health and performance are generally not widespread among the strains examined, and beneficial findings are generally limited to single studies with small sample sizes. Multiple studies with the same strains and using similar endpoints are needed before definitive recommendations for use can be made. We conclude that, at present, there is not compelling scientific evidence to support the use of any particular probiotic(s) to promote physical or psychological performance in healthy military personnel. However, plausibility for physical and psychological health and performance benefits remains, and additional research is warranted. In particular, research in military cohorts would aid in assessing the value of probiotics for supporting physical and psychological health and performance under the unique demands required of these populations.
Collapse
Affiliation(s)
- Richard T Agans
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States.,Naval Medical Research Unit Dayton, Environmental Health Effects Laboratory, Dayton, OH, United States
| | - Grace E Giles
- Soldier Performance Optimization Directorate, U.S. Army Combat Capabilities Development Command - Soldier Center, Natick, MA, United States
| | - Michael S Goodson
- Air Force Research Laboratory, 711th Human Performance Wing, Wright Patterson Air Force Base, Dayton, OH, United States
| | - J Philip Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Samantha Leyh
- Air Force Research Laboratory, 711th Human Performance Wing, Wright Patterson Air Force Base, Dayton, OH, United States.,Oak Ridge Institute for Science and Education, Wright Patterson Air Force Base, Oak Ridge, TN, United States
| | - Karen L Mumy
- Naval Medical Research Unit Dayton, Environmental Health Effects Laboratory, Dayton, OH, United States
| | - Kenneth Racicot
- Soldier Performance Optimization Directorate, U.S. Army Combat Capabilities Development Command - Soldier Center, Natick, MA, United States
| | - Jason W Soares
- Soldier Performance Optimization Directorate, U.S. Army Combat Capabilities Development Command - Soldier Center, Natick, MA, United States
| |
Collapse
|
199
|
Peng M, Tabashsum Z, Anderson M, Truong A, Houser AK, Padilla J, Akmel A, Bhatti J, Rahaman SO, Biswas D. Effectiveness of probiotics, prebiotics, and prebiotic-like components in common functional foods. Compr Rev Food Sci Food Saf 2020; 19:1908-1933. [PMID: 33337097 DOI: 10.1111/1541-4337.12565] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022]
Abstract
The bioactive ingredients in commonly consumed foods include, but are not limited to, prebiotics, prebiotic-like components, probiotics, and postbiotics. The bioactive ingredients in functional foods have also been associated with beneficial effects on human health. For example, they aid in shaping of gut microflora and promotion of immunity. These functional components also contribute in preventing serious diseases such as cardiovascular malfunction and tumorigenesis. However, the specific mechanisms of these positive influences on human health are still under investigation. In this review, we aim to emphasize the major contents of probiotics, prebiotics, and prebiotic-like components commonly found in consumable functional foods, and we present an overview of direct and indirect benefits they provide on human health. The major contributors are certain families of metabolites, specifically short-chain fatty acids and polyunsaturated fatty acids produced by probiotics, and prebiotics, or prebiotic-like components such as flavonoids, polyphenols, and vitamins that are found in functional foods. These functional ingredients in foods influence the gut microbiota by stimulating the growth of beneficial microbes and the production of beneficial metabolites that, in turn, have direct benefits to the host, while also providing protection from pathogens and maintaining a balanced gut ecosystem. The complex interactions that arise among functional food ingredients, human physiology, the gut microbiota, and their respective metabolic pathways have been found to minimize several factors that contribute to the incidence of chronic disease, such as inflammation oxidative stress.
Collapse
Affiliation(s)
- Mengfei Peng
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Zajeba Tabashsum
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Mary Anderson
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Andy Truong
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Ashley K Houser
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Joselyn Padilla
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Ahlam Akmel
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Jacob Bhatti
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Sciences, University of Maryland, College Park, Maryland
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland.,Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland.,Center for Food Safety and Security Systems, University of Maryland, College Park, Maryland
| |
Collapse
|
200
|
Ahmadi S, Wang S, Nagpal R, Wang B, Jain S, Razazan A, Mishra SP, Zhu X, Wang Z, Kavanagh K, Yadav H. A human-origin probiotic cocktail ameliorates aging-related leaky gut and inflammation via modulating the microbiota/taurine/tight junction axis. JCI Insight 2020; 5:132055. [PMID: 32302292 DOI: 10.1172/jci.insight.132055] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
Inflammation is a major risk factor of morbidity and mortality in older adults. Although its precise etiology is unknown, low-grade inflammation in older adults is commonly associated with increased intestinal epithelial permeability (leaky gut) and abnormal (dysbiotic) gut microbiota. The increasing older population and lack of treatments to reduce aging-related microbiota dysbiosis, leaky gut, and inflammation culminates in a rise in aging-related comorbidities, constituting a significant public health concern. Here, we demonstrate that a human-origin probiotic cocktail containing 5 Lactobacillus and 5 Enterococcus strains isolated from healthy infant gut prevented high-fat diet-induced (HFD-induced) microbiota dysbiosis, leaky gut, inflammation, metabolic dysfunctions, and physical function decline in older mice. Probiotic-modulated gut microbiota primarily reduced leaky gut by increasing tight junctions, which in turn reduced inflammation. Mechanistically, probiotics modulated microbiota in a way to increase bile salt hydrolase activity, which in turn increased taurine abundance in the gut that stimulated tight junctions and suppressed gut leakiness. Furthermore, in Caenorhabditis elegans, taurine increased life span, reduced adiposity and leaky gut, and enhanced physical function. The results suggest that such probiotic therapies could prevent or treat aging-related leaky gut and inflammation in the elderly.
Collapse
Affiliation(s)
- Shokouh Ahmadi
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shaohua Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Bo Wang
- Department of Chemistry, North Carolina A&T State University, Greensboro, North Carolina, USA
| | - Shalini Jain
- Department of Internal Medicine-Endocrinology and Metabolism.,Mouse Metabolic Phenotyping Core
| | - Atefeh Razazan
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sidharth P Mishra
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Xuewei Zhu
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| | - Zhan Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kylie Kavanagh
- Department of Pathology-Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Sciences, University of Tasmania, Hobart, Australia
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| |
Collapse
|