101
|
Kim JE, Tun HM, Bennett DC, Leung FC, Cheng KM. Microbial diversity and metabolic function in duodenum, jejunum and ileum of emu (Dromaius novaehollandiae). Sci Rep 2023; 13:4488. [PMID: 36934111 PMCID: PMC10024708 DOI: 10.1038/s41598-023-31684-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 03/15/2023] [Indexed: 03/20/2023] Open
Abstract
Emus (Dromaius novaehollandiae), a large flightless omnivorous ratite, are farmed for their fat and meat. Emu fat can be rendered into oil for therapeutic and cosmetic use. They are capable of gaining a significant portion of its daily energy requirement from the digestion of plant fibre. Despite of its large body size and low metabolic rate, emus have a relatively simple gastroinstetinal (GI) tract with a short mean digesta retention time. However, little is known about the GI microbial diversity of emus. The objective of this study was to characterize the intraluminal intestinal bacterial community in the different segments of small intestine (duodenum, jejunum, and ileum) using pyrotag sequencing and compare that with the ceca. Gut content samples were collected from each of four adult emus (2 males, 2 females; 5-6 years old) that were free ranged but supplemented with a barley-alfalfa-canola based diet. We amplified the V3-V5 region of 16S rRNA gene to identify the bacterial community using Roche 454 Junior system. After quality trimming, a total of 165,585 sequence reads were obtained from different segments of the small intestine (SI). A total of 701 operational taxonomic units (OTUs) were identified in the different segments of small intestine. Firmicutes (14-99%) and Proteobacteria (0.5-76%) were the most predominant bacterial phyla in the small intestine. Based on species richness estimation (Chao1 index), the average number of estimated OTUs in the small intestinal compartments were 148 in Duodenum, 167 in Jejunum, and 85 in Ileum, respectively. Low number of core OTUs identified in each compartment of small intestine across individual birds (Duodenum: 13 OTUs, Jejunum: 2 OTUs, Ileum: 14 OTUs) indicated unique bacterial community in each bird. Moreover, only 2 OTUs (Escherichia and Sinobacteraceae) were identified as core bacteria along the whole small intestine. PICRUSt analysis has indicated that the detoxification of plant material and environmental chemicals seem to be performed by SI microbiota, especially those in the jejunum. The emu cecal microbiome has more genes than SI segments involving in protective or immune response to enteric pathogens. Microbial digestion and fermentation is mostly in the jejunum and ceca. This is the first study to characterize the microbiota of different compartments of the emu intestines via gut samples and not fecal samples. Results from this study allow us to further investigate the influence of the seasonal and physiological changes of intestinal microbiota on the nutrition of emus and indirectly influence the fatty acid composition of emu fat.
Collapse
Affiliation(s)
- Ji Eun Kim
- Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Hein M Tun
- School of Public Health, Li Ka Shing, Faculty of Medicine, HKU-Pasteur Research Pole, University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
- JC School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Darin C Bennett
- Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Frederick C Leung
- School of Biological Sciences, Faculty of Science, University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Kimberly M Cheng
- Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada.
| |
Collapse
|
102
|
Pansai N, Detarun P, Chinnaworn A, Sangsupawanich P, Wichienchot S. Effects of dragon fruit oligosaccharides on immunity, gut microbiome, and their metabolites in healthy adults – a randomized double-blind placebo controlled study. Food Res Int 2023; 167:112657. [PMID: 37087207 DOI: 10.1016/j.foodres.2023.112657] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/26/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
Healthy food has wide popularity and relates positively to health. Our previous studies have shown that dragon fruit oligosaccharides (DFO) have prebiotic activities, balancing the gut microbiota in a simulated human colon system, and are safe and stimulate the immune system in rats. The effects of DFO on immune stimulation gut microbe modulation and the correlation of gut microbiota and nutrients were investigated in a human trial. This clinical study was a randomized, double-blinded, placebo-controlled trial. The participants were 107 healthy adults, divided into 3 groups that received DFO in drinking waterdoses of 4 and 8 g/day, compared to the placebo group for 4 consecutive weeks. DFO consumption at 4 g/day increased IgA level (11.31 mg/dL or 10.95% from baseline) and 8 g/day outstandingly promoted the growth of Bifidobacterium spp. (8.41%) and Faecalibacterium (1.99%) and decreased harmful bacteria, especially, Escherichia coli (8.44%). The relationship between gut microbes and nutrient intake was explored and significant (p < 0.05) correlations between specific microbial groups and intakes of specific macro- and micronutrients were observed. The potential dose of DFO for healthy adults was established as 4 g/day for improving IgA level and 8 g/day for promoting beneficial gut microbiota.
Collapse
|
103
|
Advances and challenges in interaction between heteroglycans and Bifidobacterium: Utilization strategies, intestinal health and future perspectives. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
104
|
Impact of Molasses on Ruminal Volatile Fatty Acid Production and Microbiota Composition In Vitro. Animals (Basel) 2023; 13:ani13040728. [PMID: 36830515 PMCID: PMC9952058 DOI: 10.3390/ani13040728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The aim of this study was to assess if molasses could modify VFA production and the rumen microbial community in vitro. Three beet (treatment Beet) and three cane (treatment Cane) molasses preparations were randomly selected from a variety of samples collected worldwide and incubated in vitro with rumen fluid along with a control sample (treatment CTR, in which no molasses was used). Flasks for VFA analysis were sampled at 0, 1, 2, 3, 4, 6, 8, and 24 h of each incubation. For microbiota analysis, samples from each fermentation flask after 12 and 24 h were subjected to microbial DNA extraction and V3-V4 16S rRNA gene sequencing on an Illumina MiSeq platform. Total net VFA production was higher in the beet and cane preparations than in the control (CTR) group at 24 h (33 mmol/L, 34 mmol/L, and 24.8 mmol/L, respectively), and the composition of VFAs was affected by the inclusion of molasses: acetic acid increased in the CTR group (73.5 mol%), while propionic acid increased in the beet and cane molasses (19.6 mol% and 18.6 mol%, respectively), and butyric acid increased, especially in the cane group (23.2 mol%). Molasses even influenced the composition of the rumen microbiota, and particularly the relative abundance of the most dominant family in the rumen, Prevotellaceae, which decreased compared to CTR (37.13%, 28.88%, and 49.6%, respectively). In contrast, Streptococcaceae (19.62% and 28.10% in molasses compared to 6.23% in CTR), Veillonellaceae (6.48% and 8.67% in molasses compared to 4.54% in CTR), and Fibrobacteraceae (0.90% and 0.88% in molasses compared to 0.62% in CTR) increased in the beet and cane groups compared to the CTR group. Another important finding is the lower proportion of Methanobacteriaceae following the addition of molasses compared to CTR (0.26%, 0.28%, and 0.43%, respectively). This study showed the impact of molasses in influencing VFA production and composition as a result of a modified rumen microbial composition.
Collapse
|
105
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
106
|
Lai Y, Chen Y, Zheng J, Liu Z, Nong D, Liang J, Li Y, Huang Z. Gut microbiota of white-headed black langurs ( Trachypithecus leucocephalus) in responses to habitat fragmentation. Front Microbiol 2023; 14:1126257. [PMID: 36860490 PMCID: PMC9968942 DOI: 10.3389/fmicb.2023.1126257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/20/2023] [Indexed: 02/15/2023] Open
Abstract
The white-headed black langur (Trachypithecus leucocephalus) is exclusively distributed in the karst forests and is critically endangered owing to habitat fragmentation. Gut microbiota can provide physiological data for a comprehensive study of the langur's response to human disturbance in the limestone forest; to date, data on spatial variations in the langurs' gut microbiota are limited. In this study, we examined intersite variations in the gut microbiota of white-headed black langurs in the Guangxi Chongzuo White-headed Langur National Nature Reserve, China. Our results showed that langurs in the Bapen area with a better habitat had higher gut microbiota diversity. In the Bapen group, the Bacteroidetes (13.65% ± 9.73% vs. 4.75% ± 4.70%) and its representative family, Prevotellaceae, were significantly enriched. In the Banli group, higher relative abundance of Firmicutes (86.30% ± 8.60% vs. 78.85% ± 10.35%) than the Bapen group was observed. Oscillospiraceae (16.93% ± 5.39% vs. 16.13% ± 3.16%), Christensenellaceae (15.80% ± 4.59% vs. 11.61% ± 3.60%), and norank_o__Clostridia_UCG-014 (17.43% ± 6.64% vs. 9.78% ± 3.83%) were increased in comparison with the Bapen group. These intersite variations in microbiota diversity and composition could be accounted for by differences in food resources caused by fragmentation. Furthermore, compared with the Banli group, the community assembly of gut microbiota in the Bapen group was influenced by more deterministic factors and had a higher migration rate, but the difference between the two groups was not significant. This might be attributed to the serious fragmentation of the habitats for both groups. Our findings highlight the importance of gut microbiota response for the integrity of wildlife habitats and the need in using physiological indicators to study the mechanisms by which wildlife responds to human disturbances or ecological variations.
Collapse
Affiliation(s)
- Ying Lai
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- College of Life Sciences, Guangxi Normal University, Guilin, China
| | - Yanqiong Chen
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- College of Life Sciences, Guangxi Normal University, Guilin, China
| | - Jingjin Zheng
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- College of Life Sciences, Guangxi Normal University, Guilin, China
| | - Zheng Liu
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- College of Life Sciences, Guangxi Normal University, Guilin, China
| | - Dengpan Nong
- Administration Center of Guangxi Chongzuo White-headed Langur National Nature Reserve, Chongzuo, China
| | - Jipeng Liang
- Administration Center of Guangxi Chongzuo White-headed Langur National Nature Reserve, Chongzuo, China
| | - Youbang Li
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- College of Life Sciences, Guangxi Normal University, Guilin, China
| | - Zhonghao Huang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin, China
- Guangxi Key Laboratory of Rare and Endangered Animal Ecology, Guangxi Normal University, Guilin, China
- College of Life Sciences, Guangxi Normal University, Guilin, China
| |
Collapse
|
107
|
Dietary fiber and SCFAs in the regulation of mucosal immunity. J Allergy Clin Immunol 2023; 151:361-370. [PMID: 36543697 DOI: 10.1016/j.jaci.2022.11.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022]
Abstract
Gut bacterial metabolites such as short-chain fatty acids (SCFAs) have important effects on immune cells and the gut. SCFAs derive from the fermentation of dietary fiber by gut commensal bacteria. Insufficient fiber intake thus compromises SCFA production and, as a consequence, the host's physiology (particularly immune functions). We propose that many Western diseases, including those associated with impaired mucosal responses such as food allergy and asthma, may be affected by insufficient fiber intake and reduced SCFA levels in the gut and blood. Insufficient fiber intake is 1 alternative, or contributor, on top of the "hygiene hypothesis" to the rise of Western lifestyle diseases, and the 2 ideas need to be reconciled. The mechanisms by which SCFAs influence immunity and gut homeostasis are varied; they include stimulation of G protein-coupled receptors (GPCRs), such as GPR43 or GPR41; inhibition of histone deacetylases (and hence, gene transcription changes); and induction of intracellular metabolic changes. SCFAs modulate at many different levels to alter mucosal homeostasis, including changes to gut epithelial integrity, increases in regulatory T-cell numbers and function, and decreased expression of numerous inflammatory cytokines. There is scope for preventing and/or treating diseases by using diets that alter SCFA levels.
Collapse
|
108
|
Alves-Santos AM, Sampaio KB, Lima MDS, Coelho ASG, Souza ELD, Naves MMV. Chemical composition and prebiotic activity of baru (Dipteryx alata Vog.) pulp on probiotic strains and human colonic microbiota. Food Res Int 2023; 164:112366. [PMID: 36737953 DOI: 10.1016/j.foodres.2022.112366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/08/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022]
Abstract
Little knowledge is available in literature regarding the chemical composition and health-promoting effects of baru (Dipteryx alata Vog.) pulp, a by-product usually discarded by the agro-industry during the processing of baru fruit. This study evaluated the chemical composition of baru pulp and investigated its prebiotic activity on distinct probiotic strains and human colonic microbiota with in vitro assays. Baru pulp had high contents of insoluble dietary fibers and phenolic compounds (mainly hesperidin). Baru pulp stimulated the growth and metabolism of the probiotics Bifidobacterium animalis subsp. lactis BB-12, Lactobacillus acidophilus LA-05, and Lacticaseibacillus casei L-26. In addition, digested baru pulp induced significant benefits on the human colonic microbiota, increasing the relative abundance of Lactobacillus-Enterococcus, Bifidobacterium, and Bacteroides-Prevotella, as well as the production of lactate, acetate, propionate, and butyrate. The results show that baru pulp has potential prebiotic properties to be explored in the formulation of new health-promoting foods.
Collapse
Affiliation(s)
- Aline Medeiros Alves-Santos
- Laboratory of Experimental Nutrition, School of Nutrition, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Karoliny Brito Sampaio
- Laboratory of Food Microbiology, Department of Nutrition, Federal University of Paraíba, João Pessoa, Brazil
| | - Marcos Dos Santos Lima
- Department of Food Technology, Federal Institute of Sertão Pernambucano, Petrolina, Brazil
| | | | - Evandro Leite de Souza
- Laboratory of Food Microbiology, Department of Nutrition, Federal University of Paraíba, João Pessoa, Brazil
| | | |
Collapse
|
109
|
Zhang G, Zhang J, Yan S, Hao M, Fei C, Ji D, Mao C, Tong H, Lu T, Su L. Study on the plasma metabolomics of Schisandra chinensis polysaccharide against ulcerative colitis and its correlation with gut microbes metabolism. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2023. [DOI: 10.1016/j.cjac.2023.100244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
110
|
Effect of Tea Saponins on Rumen Microbiota and Rumen Function in Qinchuan Beef Cattle. Microorganisms 2023; 11:microorganisms11020374. [PMID: 36838339 PMCID: PMC9963011 DOI: 10.3390/microorganisms11020374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Antibiotics can promote livestock growth but have side effects, so the search for safe and effective alternatives to antibiotics is urgent. This study aimed to evaluate the effect of supplementing cattle feed with tea saponins on ruminal bacteria and fungi. Sixteen Qinchuan beef cattle with a live body weight of 250 ± 10 kg were divided into four groups (four animals in each group) using a completely randomized experimental design. Four different levels of tea saponins were provided to the Qinchuan cattle as treatments, including 0 g/cattle per day control, CON), 10 g/cattle per day (low-level, LT), 20 g/cattle per day (medium-level, MT) and 30 g/cattle per day (high-level, HT). The pre-feeding period was 10 days and the official period was 80 days in this experiment. After 90 days of feeding, the rumen fluid from sixteen Qinchuan beef cattle was collected using an oral stomach tube for evaluating changes in ruminal microbiota and rumen fermentation parameters. Results indicate that the total VFAs and proportions of propionate in the LT group was significantly higher than that in the CON and HT groups (p < 0.05). For ruminal bacteria, results indicate that the Chao1 index of the MT group was significantly lower than the CON and HT groups (p < 0.05). The phyla Bacteroidetes and Firmicutes were found to be the most abundant in all treatment groups, with the LT group having significantly increased relative abundances of Proteobacteria, Actinobacteria and Ascomycota at the phylum level (p < 0.05). The relative abundance of Bacteroides was found to be relatively lower in the LT, MT and HT treatment groups compared with the CON treatment group at the genus level (p < 0.05). For ruminal fungi, the LT treatment group was found to have higher relative abundances of Saccharomyces and Aspergillus, and lower relative abundances of Succiniclasticum and Bacteroides at the at the phylum level (p < 0.05). Compared with the CON treatment group, a significant increase in the relative abundance of Saccharomyces and Aspergillus were observed in the LT treatment group at the genus level (p < 0.05). PICRUSt analyses identified pathways associated with Xenobiotic biodegradation and metabolism and glycolysisIII to be significantly enriched in the LT and HT treatment groups (p < 0.05). These findings could provide insights on how tea saponins may influence ruminal bacteria and fungi, providing a theoretical basis for replacing antibiotics with tea saponins for promoting growth in cattle.
Collapse
|
111
|
Pandey H, Tang DWT, Wong SH, Lal D. Gut Microbiota in Colorectal Cancer: Biological Role and Therapeutic Opportunities. Cancers (Basel) 2023; 15:cancers15030866. [PMID: 36765824 PMCID: PMC9913759 DOI: 10.3390/cancers15030866] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths worldwide. While CRC is thought to be an interplay between genetic and environmental factors, several lines of evidence suggest the involvement of gut microbiota in promoting inflammation and tumor progression. Gut microbiota refer to the ~40 trillion microorganisms that inhabit the human gut. Advances in next-generation sequencing technologies and metagenomics have provided new insights into the gut microbial ecology and have helped in linking gut microbiota to CRC. Many studies carried out in humans and animal models have emphasized the role of certain gut bacteria, such as Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, and colibactin-producing Escherichia coli, in the onset and progression of CRC. Metagenomic studies have opened up new avenues for the application of gut microbiota in the diagnosis, prevention, and treatment of CRC. This review article summarizes the role of gut microbiota in CRC development and its use as a biomarker to predict the disease and its potential therapeutic applications.
Collapse
Affiliation(s)
- Himani Pandey
- Redcliffe Labs, Electronic City, Noida 201301, India
| | - Daryl W. T. Tang
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Sunny H. Wong
- Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Correspondence: (S.H.W.); (D.L.)
| | - Devi Lal
- Department of Zoology, Ramjas College, University of Delhi, Delhi 110007, India
- Correspondence: (S.H.W.); (D.L.)
| |
Collapse
|
112
|
Ma B, Gavzy SJ, Saxena V, Song Y, Piao W, Lwin HW, Lakhan R, Iyyathurai J, Li L, France M, Paluskievicz C, Shirkey MW, Hittle L, Munawwar A, Mongodin EF, Bromberg JS. Strain-specific alterations in gut microbiome and host immune responses elicited by tolerogenic Bifidobacterium pseudolongum. Sci Rep 2023; 13:1023. [PMID: 36658194 PMCID: PMC9852428 DOI: 10.1038/s41598-023-27706-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
The beneficial effects attributed to Bifidobacterium are largely attributed to their immunomodulatory capabilities, which are likely to be species- and even strain-specific. However, their strain-specificity in direct and indirect immune modulation remain largely uncharacterized. We have shown that B. pseudolongum UMB-MBP-01, a murine isolate strain, is capable of suppressing inflammation and reducing fibrosis in vivo. To ascertain the mechanism driving this activity and to determine if it is specific to UMB-MBP-01, we compared it to a porcine tropic strain B. pseudolongum ATCC25526 using a combination of cell culture and in vivo experimentation and comparative genomics approaches. Despite many shared features, we demonstrate that these two strains possess distinct genetic repertoires in carbohydrate assimilation, differential activation signatures and cytokine responses signatures in innate immune cells, and differential effects on lymph node morphology with unique local and systemic leukocyte distribution. Importantly, the administration of each B. pseudolongum strain resulted in major divergence in the structure, composition, and function of gut microbiota. This was accompanied by markedly different changes in intestinal transcriptional activities, suggesting strain-specific modulation of the endogenous gut microbiota as a key to immune modulatory host responses. Our study demonstrated a single probiotic strain can influence local, regional, and systemic immunity through both innate and adaptive pathways in a strain-specific manner. It highlights the importance to investigate both the endogenous gut microbiome and the intestinal responses in response to probiotic supplementation, which underpins the mechanisms through which the probiotic strains drive the strain-specific effect to impact health outcomes.
Collapse
Affiliation(s)
- Bing Ma
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Samuel J Gavzy
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yang Song
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wenji Piao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hnin Wai Lwin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ram Lakhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jegan Iyyathurai
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michael France
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Christina Paluskievicz
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Marina W Shirkey
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lauren Hittle
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Emmanuel F Mongodin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Lung Diseases, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jonathan S Bromberg
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
113
|
Mazhar S, Khokhlova E, Colom J, Simon A, Deaton J, Rea K. In vitro and in silico assessment of probiotic and functional properties of Bacillus subtilis DE111 ®. Front Microbiol 2023; 13:1101144. [PMID: 36713219 PMCID: PMC9880548 DOI: 10.3389/fmicb.2022.1101144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023] Open
Abstract
Bacillus subtilis DE111® is a safe, well-tolerated commercially available spore-forming probiotic that has been clinically shown to support a healthy gut microbiome, and to promote digestive and immune health in both adults and children. Recently it was shown that this spore-forming probiotic was capable of germinating in the gastrointestinal tract as early as 3 h after ingestion. However, a better understanding of the mechanisms involved in the efficacy of DE111® is required. Therefore, the present investigation was undertaken to elucidate the functional properties of DE111® through employing a combination of in vitro functional assays and genome analysis. DE111® genome mining revealed the presence of several genes encoding acid and stress tolerance mechanisms in addition to adhesion proteins required to survive and colonize harsh gastrointestinal environment including multi subunit ATPases, arginine deiminase (ADI) pathway genes (argBDR), stress (GroES/GroEL and DnaK/DnaJ) and extracellular polymeric substances (EPS) biosynthesis genes (pgsBCA). DE111® harbors several genes encoding enzymes involved in the metabolism of dietary molecules (protease, lipases, and carbohyrolases), antioxidant activity and genes associated with the synthesis of several B-vitamins (thiamine, riboflavin, pyridoxin, biotin, and folate), vitamin K2 (menaquinone) and seven amino acids including five essential amino acids (threonine, tryptophan, methionine, leucine, and lysine). Furthermore, a combined in silico analysis of bacteriocin producing genes with in vitro analysis highlighted a broad antagonistic activity of DE111® toward numerous urinary tract, intestinal, and skin pathogens. Enzymatic activities included proteases, peptidases, esterase's, and carbohydrate metabolism coupled with metabolomic analysis of DE111® fermented ultra-high temperature milk, revealed a high release of amino acids and beneficial short chain fatty acids (SCFAs). Together, this study demonstrates the genetic and phenotypic ability of DE111® for surviving harsh gastric transit and conferring health benefits to the host, in particular its efficacy in the metabolism of dietary molecules, and its potential to generate beneficial SCFAs, casein-derived bioactive peptides, as well as its high antioxidant and antimicrobial potential. Thus, supporting the use of DE111® as a nutrient supplement and its pottential use in the preparation of functional foods.
Collapse
Affiliation(s)
- Shahneela Mazhar
- Deerland Ireland R&D Ltd., ADM, Food Science Building, University College Cork, Cork, Ireland
| | - Ekaterina Khokhlova
- Deerland Ireland R&D Ltd., ADM, Food Science Building, University College Cork, Cork, Ireland
| | - Joan Colom
- Deerland Ireland R&D Ltd., ADM, Food Science Building, University College Cork, Cork, Ireland
| | - Annie Simon
- Deerland Ireland R&D Ltd., ADM, Food Science Building, University College Cork, Cork, Ireland
| | - John Deaton
- Deerland Probiotics and Enzymes, ADM, Kennesaw, GA, United States
| | - Kieran Rea
- Deerland Ireland R&D Ltd., ADM, Food Science Building, University College Cork, Cork, Ireland
| |
Collapse
|
114
|
Senftleber NK, Ramne S, Moltke I, Jørgensen ME, Albrechtsen A, Hansen T, Andersen MK. Genetic Loss of Sucrase-Isomaltase Function: Mechanisms, Implications, and Future Perspectives. Appl Clin Genet 2023; 16:31-39. [PMID: 36994449 PMCID: PMC10041990 DOI: 10.2147/tacg.s401712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
Genetic variants causing loss of sucrase-isomaltase (SI) function result in malabsorption of sucrose and starch components and the condition congenital sucrase-isomaltase deficiency (CSID). The identified genetic variants causing CSID are very rare in all surveyed populations around the globe, except the Arctic-specific c.273_274delAG loss-of-function (LoF) variant, which is common in the Greenlandic Inuit and other Arctic populations. In these populations, it is, therefore, possible to study people with loss of SI function in an unbiased way to elucidate the physiological function of SI, and investigate both short-term and long-term health effects of reduced small intestinal digestion of sucrose and starch. Importantly, a recent study of the LoF variant in Greenlanders reported that adult homozygous carriers have a markedly healthier metabolic profile. These findings indicate that SI inhibition could potentially improve metabolic health also in individuals not carrying the LoF variant, which is of great interest considering the massive number of individuals with obesity and type 2 diabetes worldwide. Therefore, the objectives of this review, are 1) to describe the biological role of SI, 2) to describe the metabolic impact of the Arctic SI LoF variant, 3) to reflect on potential mechanisms linking reduced SI function to metabolic health, and 4) to discuss what knowledge is necessary to properly evaluate whether SI inhibition is a potential therapeutic target for improving cardiometabolic health.
Collapse
Affiliation(s)
- Ninna Karsbæk Senftleber
- Clinical Research, Copenhagen University Hospital – Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Stina Ramne
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Moltke
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marit Eika Jørgensen
- Clinical Research, Copenhagen University Hospital – Steno Diabetes Center Copenhagen, Herlev, Denmark
- Centre for Public Health in Greenland, National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Steno Diabetes Center Greenland, Nuuk, Greenland
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Correspondence: Mette K Andersen, University of Copenhagen, Blegdamsvej 3B, Mærsk Tårnet, 8. sal, 2200 København N., Copenhagen, Denmark, Tel +45 35325282, Email
| |
Collapse
|
115
|
Mercer EM, Arrieta MC. Probiotics to improve the gut microbiome in premature infants: are we there yet? Gut Microbes 2023; 15:2201160. [PMID: 37122152 PMCID: PMC10153018 DOI: 10.1080/19490976.2023.2201160] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
Gut microbiome maturation in infants born prematurely is uniquely influenced by the physiological, clinical, and environmental factors surrounding preterm birth and early life, leading to altered patterns of microbial succession relative to term infants during the first months of life. These differences in microbiome composition are implicated in acute clinical conditions that disproportionately affect preterm infants, including necrotizing enterocolitis (NEC) and late-onset sepsis (LOS). Probiotic supplementation initiated early in life is an effective prophylactic measure for preventing NEC, LOS, and other clinical concerns relevant to preterm infants. In parallel, reported benefits of probiotics on the preterm gut microbiome, metabolome, and immune function are beginning to emerge. This review summarizes the current literature on the influence of probiotics on the gut microbiome of preterm infants, outlines potential mechanisms by which these effects are exerted, and highlights important clinical considerations for determining the best practices for probiotic use in premature infants.
Collapse
Affiliation(s)
- Emily M. Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
116
|
Castro-Ramos JJ, Solís-Oba A, Solís-Oba M, Calderón-Vázquez CL, Higuera-Rubio JM, Castro-Rivera R. Effect of the initial pH on the anaerobic digestion process of dairy cattle manure. AMB Express 2022; 12:162. [PMID: 36576594 PMCID: PMC9797631 DOI: 10.1186/s13568-022-01486-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/01/2022] [Indexed: 12/29/2022] Open
Abstract
Anaerobic digestion (AD) has recently been studied to obtain products of greater interest than biogas, such as volatile fatty acids (VFAs) and phytoregulators. The effect of the initial pH of cow manure and the fermentation time of the AD on the microbial composition, VFAs, indole-3-acetic acid (IAA) and gibberellic acid (GA3) production was evaluated. The cow manure (7% solids) was adjusted to initial pH values of 5.5, 6.5, 7.5, and 8.5, and the AD products were analyzed every four days until day 20. The initial pH and the fermentation time had an important effect on the production of metabolites. During AD, only the hydrolytic and acidogenic stages were identified, and the bacteria found were from the phyla Firmicutes, Bacteroidetes, Actinobacteria, and Spirochaetes. The most abundant genera produced in the four AD were Caproiciproducens, Clostridium sensu stricto 1, Romboutsia, Paeniclostridium, Turicibacter, Peptostreptococcaceae, Ruminococcaceae and Fonticella. The highest amount of VFAs was obtained at pH 8.5, and the production of the acids was butyric > acetic > propionic. The maximum production of GA3 and IAA was at an initial pH of 6.5 on day 20 and a pH of 5.5 on day 4, respectively. There was a strong correlation (> 0.8) between the most abundant microorganisms and the production of VFAs and GA3. The anaerobic digestion of cow manure is a good alternative for the production of VFAs, GA3 and IAA.
Collapse
Affiliation(s)
- Job Jonathan Castro-Ramos
- grid.418275.d0000 0001 2165 8782Instituto Politécnico Nacional, Centro de Investigación en Biotecnologia Aplicada, 90700 Tepetitla de Lardizábal, Tlaxcala Mexico
| | - Aida Solís-Oba
- grid.7220.70000 0001 2157 0393Universidad Autónoma Metropolitana, Unidad Xochimilco, Ciudad de Mexico, Mexico
| | - Myrna Solís-Oba
- grid.418275.d0000 0001 2165 8782Instituto Politécnico Nacional, Centro de Investigación en Biotecnologia Aplicada, 90700 Tepetitla de Lardizábal, Tlaxcala Mexico
| | - Carlos Ligne Calderón-Vázquez
- grid.418275.d0000 0001 2165 8782Instituto Politécnico Nacional, CIIDIR Unidad Sinaloa, 81100 Guasave, Sinaloa Mexico
| | - Jesús Mireya Higuera-Rubio
- grid.418275.d0000 0001 2165 8782Instituto Politécnico Nacional, CIIDIR Unidad Sinaloa, 81100 Guasave, Sinaloa Mexico
| | - Rigoberto Castro-Rivera
- grid.418275.d0000 0001 2165 8782Instituto Politécnico Nacional, Centro de Investigación en Biotecnologia Aplicada, 90700 Tepetitla de Lardizábal, Tlaxcala Mexico
| |
Collapse
|
117
|
Abstract
Bifidobacteria naturally inhabit diverse environments, including the gastrointestinal tracts of humans and animals. Members of the genus are of considerable scientific interest due to their beneficial effects on health and, hence, their potential to be used as probiotics. By definition, probiotic cells need to be viable despite being exposed to several stressors in the course of their production, storage, and administration. Examples of common stressors encountered by probiotic bifidobacteria include oxygen, acid, and bile salts. As bifidobacteria are highly heterogenous in terms of their tolerance to these stressors, poor stability and/or robustness can hamper the industrial-scale production and commercialization of many strains. Therefore, interest in the stress physiology of bifidobacteria has intensified in recent decades, and many studies have been established to obtain insights into the molecular mechanisms underlying their stability and robustness. By complementing traditional methodologies, omics technologies have opened new avenues for enhancing the understanding of the defense mechanisms of bifidobacteria against stress. In this review, we summarize and evaluate the current knowledge on the multilayered responses of bifidobacteria to stressors, including the most recent insights and hypotheses. We address the prevailing stressors that may affect the cell viability during production and use as probiotics. Besides phenotypic effects, molecular mechanisms that have been found to underlie the stress response are described. We further discuss strategies that can be applied to improve the stability of probiotic bifidobacteria and highlight knowledge gaps that should be addressed in future studies.
Collapse
Affiliation(s)
- Marie Schöpping
- Systems Biology, Discovery, Chr. Hansen A/S, Hørsholm, Denmark
- Division of Industrial Biotechnology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Ahmad A. Zeidan
- Systems Biology, Discovery, Chr. Hansen A/S, Hørsholm, Denmark
| | - Carl Johan Franzén
- Division of Industrial Biotechnology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
118
|
Diet and Proteinuria: State of Art. Int J Mol Sci 2022; 24:ijms24010044. [PMID: 36613485 PMCID: PMC9819984 DOI: 10.3390/ijms24010044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Proteinuria is a broad term used to describe the pathological presence of proteins, including albumin, globulin, Bence-Jones protein, and mucoprotein in the urine. When persistent, proteinuria is a marker of kidney damage and represents a reliable predictor of the risk of progression of renal failure. Medical nutrition therapy is imperative for patients with proteinuria because it may slow the progression of renal disease. The aim of this review is to explore different nutritional approaches in the management of proteinuria and their influence on pathophysiological processes. As such, protein restriction is the main dietary intervention. Indeed, other management approaches are frequently used to reduce it regarding micro and macronutrients, but also the dietary style. Among these, the nutritional approach represents one of the most used and controversial interventions and the studies rarely take the form of randomized and controlled trials. With this work we aspire to analyze current clinical knowledge of how nutrition could influence proteinuria, potentially representing a useful tool in the management of proteinuric nephropathy.
Collapse
|
119
|
Ayuda-Durán B, Sánchez-Hernández E, González-Manzano S, Santos-Buelga C, González-Paramás AM. The effects of polyphenols against oxidative stress in Caenorhabditis elegans are determined by coexisting bacteria. Front Nutr 2022; 9:989427. [PMID: 36532522 PMCID: PMC9752899 DOI: 10.3389/fnut.2022.989427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/10/2022] [Indexed: 08/18/2023] Open
Abstract
Introduction Increasing evidence supports the role of gut microbiota in many aspects of human health, including immune, metabolic and neurobehavioral traits. Several studies have focused on how different components of the diet, such as polyphenols, can modulate the composition and function of the gut microbiota leading to health benefits. Methods The effects on the resistance against thermally induced oxidative stress of C. elegans grown in the presence of flavonoids (quercetin or epicatechin) and fed different probiotic strains, namely Lactobacillus plantarum CLC17, Bifidobacterium longum NCIMB 8809 and Enterococcus faecium CECT 410, were explored. Results Feeding C. elegans with the assayed bacteria in the absence of flavonoids did not significantly affect body size and fertility of the worms neither improve their resistance against oxidative stress compared to E. coli controls. However, increased resistance to stress was found when C. elegans was cultivated in the presence of both L. plantarum and flavonoids, but not with B. longum or E. faecium. An exploratory study revealed the presence of glycosylated and sulfated metabolites together with the aglycone in worms treated with quercetin and fed any of the different assayed LAB strains. However, in the assays with epicatechin a differential metabolite, tentatively identified as 5-(4'-hydroxyphenyl)-γ-valerolactone 3'-O-glucoside, was detected in the worms fed L. plantarum but not with the other bacteria. Conclusion The obtained results indicated that the interactions bacteria/polyphenol play a key role in the effects produced in C. elegans regarding resistance against oxidative stress, although those effects cannot be only explained by the ability of bacteria to metabolize polyphenols, but other mechanisms should also be involved.
Collapse
Affiliation(s)
- Begoña Ayuda-Durán
- Grupo de Investigación en Polifenoles (GIP-USAL), Universidad de Salamanca, Salamanca, Spain
| | - Eva Sánchez-Hernández
- Department of Agricultural and Forestry Engineering, ETSIIAA, University of Valladolid, Palencia, Spain
| | - Susana González-Manzano
- Grupo de Investigación en Polifenoles (GIP-USAL), Universidad de Salamanca, Salamanca, Spain
| | - Celestino Santos-Buelga
- Grupo de Investigación en Polifenoles (GIP-USAL), Universidad de Salamanca, Salamanca, Spain
| | - Ana M. González-Paramás
- Grupo de Investigación en Polifenoles (GIP-USAL), Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
120
|
Mary PR, Kapoor M. Co-culture fermentations suggest cross-feeding among Bacteroides ovatus DSMZ 1896, Lactiplantibacillus plantarum WCFS1 and Bifidobacterium adolescentis DSMZ 20083 for utilizing dietary galactomannans. Food Res Int 2022; 162:111942. [DOI: 10.1016/j.foodres.2022.111942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/27/2022] [Accepted: 09/12/2022] [Indexed: 02/08/2023]
|
121
|
Wu Q, Chen H, Zhang F, Wang W, Xiong F, Liu Y, Lv L, Li W, Bo Y, Yang H. Cysteamine Supplementation In Vitro Remarkably Promoted Rumen Fermentation Efficiency towards Propionate Production via Prevotella Enrichment and Enhancing Antioxidant Capacity. Antioxidants (Basel) 2022; 11:antiox11112233. [PMID: 36421419 PMCID: PMC9686782 DOI: 10.3390/antiox11112233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Cysteamine (CS) is a vital antioxidant product and nutritional regulator that improves the productive performance of animals. A 2 × 4 factorial in vitro experiment was performed to determine the effect of the CS supplementation levels of 0, 20, 40, and 60 mg/g, based on substrate weight, on the ruminal fermentation, antioxidant capacity, and microorganisms of a high-forage substrate (HF, forage:corn meal = 7:3) in the Statistical Analysis System Institute. After 48 h of incubation, the in vitro dry matter disappearance and gas production in the LF group were higher when compared with a low-forage substrate (LF, forge hay:corn meal = 3:7), which was analyzed via the use of the MIXED procedure of the HF group, and these increased linearly with the increasing CS supplementation (p < 0.01). With regard to rumen fermentation, the pH and acetate were lower in the LF group compared to the HF group (p < 0.01). However, the ammonia N, microbial crude protein, total volatile fatty acids (VFA), and propionate in the LF group were greater than those in the HF group (p < 0.05). With the CS supplementation increasing, the pH, ammonia N, acetate, and A:P decreased linearly, while the microbial crude protein, total VFA, and propionate increased linearly (p < 0.01). Greater antioxidant capacity was observed in the LF group, and the increasing CS supplementation linearly increased the superoxide dismutase, catalase, glutathione peroxidase, total antioxidant capacity, glutathione, and glutathione reductase, while it decreased the malondialdehyde (p < 0.05). No difference occurred in the ruminal bacteria alpha diversity with the increasing CS supplementation, but it was higher in the LF group than in the HF group (p < 0.01). Based on the rumen bacterial community, a higher proportion of Bacteroidota, instead of Firmicutes, was in the LF group than in the HF group. Furthermore, increasing the CS supplementation linearly increased the relative abundance of Prevotella, norank_f_F082, and Prevotellaceae_UCG-001 under the two substrates (p < 0.05). Prevotella, norank_f_F082, and Prevotellaceae_UCG-001 were positively correlated with gas production, rumen fermentation, and antioxidant capacity in a Spearman correlation analysis (r > 0.31, p < 0.05). Overall, a CS supplementation of not less than 20 mg/g based on substrate weight enhanced the rumen fermentation and rumen antioxidant capacity of the fermentation system, and it guided the rumen fermentation towards glucogenic propionate by enriching the Prevotella in Bacteroidetes.
Collapse
Affiliation(s)
- Qichao Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Hewei Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Fan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Weikang Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Fengliang Xiong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Yingyi Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Liangkang Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Wenjuan Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
| | - Yukun Bo
- Animal Husbandry Technology Promotion Institution of Zhangjiakou, Zhangjiakou 075000, China
| | - Hongjian Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agri-Cultural University, Beijing 100193, China
- Correspondence:
| |
Collapse
|
122
|
Sharma N, Behl T, Singh S, Kaur P, Zahoor I, Mohan S, Rachamalla M, Dailah HG, Almoshari Y, Salawi A, Alshamrani M, Aleya L. Targeting Nanotechnology and Nutraceuticals in Obesity: An Updated Approach. Curr Pharm Des 2022; 28:3269-3288. [PMID: 36200206 DOI: 10.2174/1381612828666221003105619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/22/2022] [Indexed: 01/28/2023]
Abstract
HYPOTHESIS This review article represents a brief layout of the risk factors and pathophysiology responsible for obesity, customary treatment strategies, and nanotechnology-based nutraceutical for the therapeutics of obesity. EXPERIMENTS An exhaustive search of the literature was done for this purpose, using Google Scholar, PubMed, and ScienceDirect databases. A literature study was conducted using publications published in peer-reviewed journals between 2000 and 2022. FINDINGS This was revealed that risk factors responsible for obesity were genetic abnormalities and environmental and socio-economic factors. Several research articles published between 2000 and 2022 were based on phytoconstituents-based nanoformulation for obesity therapeutics and, therefore, have been systematically compiled in this review. Various nutraceuticals like Garcinia cambogia, quercetin, resveratrol, capsaicin, Capsicum, Curcuma longa, Camella Sinensis, Zingiber officinalis, Citrus aurantium, Aegle marmelos, Coffea canephora, Asparagus officinalis, Gardenia jasminoides, Catha edulis, Clusia nemroisa, Rosmarinus officinalis, Cirsium setidens, Betula platyphylla, Tripterygium wilfordi possessing anti-obesity actions are discussed in this review along with their patents, clinical trials as well as their nanoformulation available. CONCLUSION This review illustrates that nanotechnology has a great propensity to impart a promising role in delivering phytochemicals and nutraceuticals in managing obesity conditions and other related disorders.
Collapse
Affiliation(s)
- Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana- Ambala, Haryana 133207, India
| | - Tapan Behl
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana- Ambala, Haryana 133207, India
| | - Parneet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Ishrat Zahoor
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Syam Mohan
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.,Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Hamed Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, Saudi Arabia
| | - Yosif Almoshari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Meshal Alshamrani
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Lotfi Aleya
- Chrono-environment Laboratory, Bourgogne Franche-Comté University, Besançon, France
| |
Collapse
|
123
|
Wang H, Huang X, Tan H, Chen X, Chen C, Nie S. Interaction between dietary fiber and bifidobacteria in promoting intestinal health. Food Chem 2022; 393:133407. [PMID: 35696956 DOI: 10.1016/j.foodchem.2022.133407] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 01/10/2023]
Abstract
Bifidobacteria are considered as probiotics due to their role in promoting intestinal health, including regulating intestinal flora, controlling glycolipid metabolism, anti-colitis effects. Dietary fiber is considered as prebiotic favoring gut health. It also can be used as carbon source to support the growth and colonization of probiotics like bifidobacteria. However, because of genetic diversity, different bifidobacterial species differ in their ability to utilize dietary fiber. Meanwhile, dietary fiber with different structural properties has different effects on the bifidobacteria proliferation. The interaction between dietary fiber and bifidobacteria will consequently lead to a synergistic or antagonistic function in promoting intestinal health, therefore affecting the application of combined use of dietary fiber and bifidobacteria. In this case, we summarize the biological function of bifidobacteria, and their interaction with different dietary fiber in promoting gut health, and finally provide several strategies about their combined use.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaojun Huang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaomin Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Chunhua Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| |
Collapse
|
124
|
Drey E, Kok CR, Hutkins R. Role of Bifidobacterium pseudocatenulatum in Degradation and Consumption of Xylan-Derived Carbohydrates. Appl Environ Microbiol 2022; 88:e0129922. [PMID: 36200766 PMCID: PMC9599329 DOI: 10.1128/aem.01299-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Xylans, a family of xylose-based polysaccharides, are dietary fibers resistant to digestion. They therefore reach the large intestine intact; there, they are utilized by members of the gut microbiota. They are initially broken down by primary degraders that utilize extracellular xylanases to cleave xylan into smaller oligomers. The resulting xylooligosaccharides (XOS) can either be further metabolized directly by primary degraders or cross-feed secondary consumers, including Bifidobacterium. While several Bifidobacterium species have metabolic systems for XOS, most grow poorly on longer-chain XOS and xylan substrates. In this study, we isolated strains of Bifidobacterium pseudocatenulatum and observed that some, including B. pseudocatenulatum ED02, displayed growth on XOS with a high degree of polymerization (DP) and straight-chain xylan, suggesting a primary degrader phenotype that is rare in Bifidobacterium. In silico analyses revealed that only the genomes of these xylan-fermenting (xylan+) strains contained an extracellular GH10 endo-β-1.4 xylanase, a key enzyme for primary degradation of xylan. The presence of an extracellular xylanase was confirmed by the appearance of xylan hydrolysis products in cell-free supernatants. Extracellular xylanolytic activity was only detected in xylan+ strains, as indicated by the production of XOS fragments with a DP of 2 to 6, identified by thin-layer chromatography (TLC) and high-performance liquid chromatography (HPLC). Additionally, in vitro fecal fermentations revealed that strains with a xylan+ phenotype can persist with xylan supplementation. These results indicate that xylan+ B. pseudocatenulatum strains may have a competitive advantage in the complex environment of the gastrointestinal tract, due to their ability to act as primary degraders of xylan through extracellular enzymatic degradation. IMPORTANCE The beneficial health effects of dietary fiber are now well established. Moreover, low fiber consumption is associated with increased risks of metabolic and systemic diseases. This so-called "fiber gap" also has a profound impact on the composition of the gut microbiome, leading to a disrupted or dysbiotic microbiota. Therefore, understanding the mechanisms by which keystone bacterial species in the gut utilize xylans and other dietary fibers may provide a basis for developing strategies to restore gut microbiome function. The results described here provide biochemical and genetic evidence for primary xylan utilization by human-derived Bifidobacterium pseudocatenulatum and show also that cooperative utilization of xylans occurs among other members of this species.
Collapse
Affiliation(s)
- Elizabeth Drey
- Department of Food Science and Technology, Food Innovation Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
| | - Car Reen Kok
- Nebraska Food for Health Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
- Complex Biosystems, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
| | - Robert Hutkins
- Department of Food Science and Technology, Food Innovation Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
125
|
Mancin L, Amatori S, Caprio M, Sattin E, Bertoldi L, Cenci L, Sisti D, Bianco A, Paoli A. Effect of 30 days of ketogenic Mediterranean diet with phytoextracts on athletes' gut microbiome composition. Front Nutr 2022; 9:979651. [PMID: 36386948 PMCID: PMC9642348 DOI: 10.3389/fnut.2022.979651] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/21/2022] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Recent research suggest that gut microbiome may play a fundamental role in athlete's health and performance. Interestingly, nutrition can affect athletic performance by influencing the gut microbiome composition. Among different dietary patterns, ketogenic diet represents an efficient nutritional approach to get adequate body composition in athletes, however, some concerns have been raised about its potential detrimental effect on gut microbiome. To the best of our knowledge, only one study investigated the effect of ketogenic diet on the gut microbiome in athletes (elite race walkers), whilst no studies are available in a model of mixed endurance/power sport such as soccer. This study aimed to investigate the influence of a ketogenic Mediterranean diet with phytoextracts (KEMEPHY) diet on gut microbiome composition in a cohort of semi-professional soccer players. METHODS 16 male soccer players were randomly assigned to KEMEPHY diet (KDP n = 8) or western diet (WD n = 8). Body composition, performance measurements and gut microbiome composition were measured before and after 30 days of intervention by 16S rRNA amplicon sequencing. Alpha-diversity measures and PERMANOVA was used to investigate pre-post differences in the relative abundance of all taxonomic levels (from phylum to genus) and Spearman's correlations was used to investigate associations between microbial composition and macronutrient intake. Linear discriminant analysis was also performed at the different taxonomic levels on the post-intervention data. RESULTS No differences were found between pre and post- dietary intervention for microbial community diversity: no significant effects of time (p = 0.056, ES = 0.486 and p = 0.129, ES = 0.388, respectively for OTUs number and Shannon's ENS), group (p = 0.317, ES = 0.180 and p = 0.809, ES = 0.047) or time × group (p = 0.999, ES = 0.01 and p = 0.230, ES = 0.315). Post-hoc paired Wilcoxon test showed a significant time × group effect for Actinobacteriota (p = 0.021, ES = 0.578), which increased in the WD group (median pre: 1.7%; median post: 2.3%) and decreased in the KEMEPHY group (median pre: 4.3%; median post: 1.7%). At genus level, the linear discriminant analysis in the post intervention differentiated the two groups for Bifidobacterium genus (pertaining to the Actinobacteria phylum), Butyricicoccus and Acidaminococcus genera, all more abundant in the WD group, and for Clostridia UCG-014 (order, family, and genus), Butyricimonas, Odoribacterter genera (pertaining to the Marinifilaceae family), and Ruminococcus genus, all more abundant in the KEMEPHY group. CONCLUSIONS Our results demonstrate that 30 days of KEMEPHY intervention, in contrast with previous research on ketogenic diet and gut microbiome, do not modify the overall composition of gut microbiome in a cohort of athletes. KEMEPHY dietary pattern may represent an alternative and safety tool for maintaining and/or regulating the composition of gut microbiome in athletes practicing regular exercise. Due to the fact that not all ketogenic diets are equal, we hypothesized that each version of ketogenic diet, with different kind of nutrients or macronutrients partitioning, may differently affect the human gut microbiome.
Collapse
Affiliation(s)
- Laura Mancin
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Human Inspired Technology Research Center, University of Padua, Padua, Italy
| | - Stefano Amatori
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Massimiliano Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | | | | | - Lorenzo Cenci
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Human Inspired Technology Research Center, University of Padua, Padua, Italy
| | - Davide Sisti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Antonino Bianco
- Sport and Exercise Sciences Research Unit, University of Palermo, Palermo, Italy
| | - Antonio Paoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Human Inspired Technology Research Center, University of Padua, Padua, Italy
- Research Center for High Performance Sport, UCAM, Catholic University of Murcia, Murcia, Spain
| |
Collapse
|
126
|
González-Vázquez R, Zúñiga-León E, Torres-Maravilla E, Leyte-Lugo M, Mendoza-Pérez F, Hernández-Delgado NC, Pérez-Pastén-Borja R, Azaola-Espinosa A, Mayorga-Reyes L. Genomic and Biochemical Characterization of Bifidobacterium pseudocatenulatum JCLA3 Isolated from Human Intestine. Microorganisms 2022; 10:2100. [PMID: 36363691 PMCID: PMC9695335 DOI: 10.3390/microorganisms10112100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 07/30/2023] Open
Abstract
Bifidobacteria have been investigated due to their mutualistic microbe-host interaction with humans throughout their life. This work aims to make a biochemical and genomic characterization of Bifidobacterium pseudocatenulatum JCLA3. By multilocus analysis, the species of B. pseudocatenulatum JCLA3 was established as pseudocatenulatum. It contains one circular genome of 2,369,863 bp with G + C content of 56.6%, no plasmids, 1937 CDSs, 54 tRNAs, 16 rRNAs, 1 tmRNA, 1 CRISPR region, and 401 operons predicted, including a CRISPR-Cas operon; it encodes an extensive number of enzymes, which allows it to utilize different carbohydrates. The ack gene was found as part of an operon formed by xfp and pta genes. Two genes of ldh were found at different positions. Chromosomally encoded resistance to ampicillin and cephalothin, non-hemolytic activity, and moderate inhibition of Escherichia coli ATCC 25922 and Staphylococcus aureus ATCC 6538 were demonstrated by B. pseudocatenulatum JCLA3; it can survive 100% in simulated saliva, can tolerate primary and secondary glyco- or tauro-conjugated bile salts but not in a mix of bile; the strain did not survive at pH 1.5-5. The cbh gene coding to choloylglycine hydrolase was identified in its genome, which could be related to the ability to deconjugate secondary bile salts. Intact cells showed twice as much antioxidant activity than debris. B. pseudocatenulatum JCLA3 showed 49% of adhesion to Caco-2 cells. The genome and biochemical analysis help to elucidate further possible biotechnological applications of B. pseudocatenulatum JCLA3.
Collapse
Affiliation(s)
- Raquel González-Vázquez
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, CONACYT-Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Eduardo Zúñiga-León
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Edgar Torres-Maravilla
- INRAE, AgroPArisTEch, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en Josas, France
| | - Martha Leyte-Lugo
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, CONACYT-Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Felipe Mendoza-Pérez
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Natalia C. Hernández-Delgado
- Laboratorio de Toxicología Molecular y Celular, Escuela Nacional de Ciencias Biológicas-Campus Zacatenco, Instituto Politécnico Nacional, Ciudad de Mexico 07738, Mexico
| | - Ricardo Pérez-Pastén-Borja
- Laboratorio de Toxicología Molecular y Celular, Escuela Nacional de Ciencias Biológicas-Campus Zacatenco, Instituto Politécnico Nacional, Ciudad de Mexico 07738, Mexico
| | - Alejandro Azaola-Espinosa
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Lino Mayorga-Reyes
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| |
Collapse
|
127
|
Dysbiotic microbiota contributes to the extent of acute myocardial infarction in rats. Sci Rep 2022; 12:16517. [PMID: 36192578 PMCID: PMC9530207 DOI: 10.1038/s41598-022-20826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022] Open
Abstract
Increasing evidence suggests that the intestinal microbiota composition could play a role in specific pathologies such as hypertension, obesity and diabetes. This study aims to demonstrate that the intestinal microbiota modulated by a diet creating dysbiosis increased the size of the myocardial infarction and that probiotics could attenuate this effect. To do this, microbiota transplants from rats fed a dysbiotic or non-dysbiotic diet in the presence or absence of probiotics were performed for 10 days on rats whose microbiota had been previously suppressed by antibiotic therapy. Then, the anterior coronary artery of the transplanted rats was occluded for 30 min. Infarct size was measured after 24 h of reperfusion, while signaling pathways were evaluated after 15 min of reperfusion. Intestinal resistance, plasma concentration of LPS (lipopolysaccharides), activation of NF-κB and Akt and composition of the microbiota were also measured. Our results demonstrate a larger infarct size in animals transplanted with the dysbiotic microbiota without probiotics compared to the other groups, including those that received the dysbiotic microbiota with probiotics. This increase in infarct size correlates with a higher firmicutes/bacteroidetes ratio, NF-kB phosphorylation and plasma LPS concentration, and a decrease in intestinal barrier resistance and Akt. These results indicate that dysbiotic microbiota promotes an increase in infarct size, an effect that probiotics can attenuate.
Collapse
|
128
|
Parhi P, Song KP, Choo WS. Growth and survival of Bifidobacterium breve and Bifidobacterium longum in various sugar systems with fructooligosaccharide supplementation. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2022; 59:3775-3786. [PMID: 36193365 PMCID: PMC9525548 DOI: 10.1007/s13197-022-05361-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Revised: 12/16/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
This study aims to investigate the effect of fructooligosaccharide (FOS) (0.5, 1, 2, 3, and 4%) supplementation on the growth and survival of Bifidobacterium breve and Bifidobacterium longum in glucose, fructose, lactose, and sucrose (2, 3, and 4%) systems with 24-h growth and 10-day survival assays at 37 °C. FOS supplementation showed a higher growth-promoting effect on B. longum than B. breve in various sugar systems. The highest percentage of increase in growth index, 78.5%, was observed with 4% sucrose supplemented with 0.5% FOS in B. longum. In comparison, the highest percentage increase in growth index, 5.6 and 6.6%, was observed in the presence of 2% glucose and 4% lactose supplemented with 0.5% FOS in B. breve. In survival assay, FOS supplementation (0.5–4%) in a 2% lactose system showed the highest positive effect on the cell viability of B. longum on day-10. As for B. breve, FOS supplementation (1 and 2%) in the 2% sucrose system showed the highest positive effect on the cell viability, followed by FOS supplementation (0.5, 3, and 4%) in 2% sucrose and FOS supplementation (3 and 4%) in 2% lactose on day-10. This study demonstrated that the efficacy of FOS supplementation was depended on its concentration, sugar system and its concentration, and Bifidobacterium strain.
Collapse
Affiliation(s)
- Priyanka Parhi
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor Malaysia
| | - Keang Peng Song
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor Malaysia
| |
Collapse
|
129
|
Arzamasov AA, Osterman AL. Milk glycan metabolism by intestinal bifidobacteria: insights from comparative genomics. Crit Rev Biochem Mol Biol 2022; 57:562-584. [PMID: 36866565 PMCID: PMC10192226 DOI: 10.1080/10409238.2023.2182272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Bifidobacteria are early colonizers of the human neonatal gut and provide multiple health benefits to the infant, including inhibiting the growth of enteropathogens and modulating the immune system. Certain Bifidobacterium species prevail in the gut of breastfed infants due to the ability of these microorganisms to selectively forage glycans present in human milk, specifically human milk oligosaccharides (HMOs) and N-linked glycans. Therefore, these carbohydrates serve as promising prebiotic dietary supplements to stimulate the growth of bifidobacteria in the guts of children suffering from impaired gut microbiota development. However, the rational formulation of milk glycan-based prebiotics requires a detailed understanding of how bifidobacteria metabolize these carbohydrates. Accumulating biochemical and genomic data suggest that HMO and N-glycan assimilation abilities vary remarkably within the Bifidobacterium genus, both at the species and strain levels. This review focuses on the delineation and genome-based comparative analysis of differences in respective biochemical pathways, transport systems, and associated transcriptional regulatory networks, providing a foundation for genomics-based projection of milk glycan utilization capabilities across a rapidly growing number of sequenced bifidobacterial genomes and metagenomic datasets. This analysis also highlights remaining knowledge gaps and suggests directions for future studies to optimize the formulation of milk-glycan-based prebiotics that target bifidobacteria.
Collapse
Affiliation(s)
- Aleksandr A Arzamasov
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
130
|
Comparative genomics reveals the evolution of antimicrobial resistance in Bacteroides nordii. Microb Pathog 2022; 173:105811. [PMID: 36183960 DOI: 10.1016/j.micpath.2022.105811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022]
Abstract
Bacteroides nordii, is an understudied member of the pathogenic B. fragilis group which comprises several multidrug-resistant (MDR) strains. Thus, it is of great interest to study the genome biology of Bacteroides nordii. However, no detailed study is available that characterized B. nordii at the genetic level and explored its role as a potential pathogen. We isolated an MDR strain viz., B. nordii PGMM4098 from the pus sample and subjected it to whole genome sequencing using Illumina technology. The draft genome was de-novo assembled and annotated, followed by comprehensive comparative genomics analyses using the publicly available genome dataset of B. nordii. The pan-genome analysis revealed the open nature of B. nordii, indicating the continuous accumulation of novel genes in non-core components leading to the emergence of new strains of this species. The thirteen antimicrobial resistance (AMR) genes identified in the genomes of all B. nordii strains were part of the non-core component of the pan-genome. Of these, four AMR genes, nimE, aadS, mef(En2), and ermB/F/G were found to be acquired via the process of horizontal gene transfer (HGT) from anaerobic Bacteroidetes. Importantly, the nimE gene conferring metronidazole resistance was found to be present only in B. nordii PGMM4098, which harbors five other AMR genes encoded in its genome. Of these, nimE (metronidazole resistance), ermB/F/G (macrolide-lincosamide-streptogramin B resistance), and cfxA2/A3 (class A β-lactam resistance) genes were further validated using targeted polymerase chain reaction assay. Notably, these three genes were also found to be under the operation of positive selective pressure suggesting the diversification of these genes, which might lead to the emergence of new MDR strains of B. nordii in the near future. Our study reported and characterized the genome of the first MDR strain of B. nordii and revealed the AMR evolution in this species using a comprehensive comparative genomics approach.
Collapse
|
131
|
Yue B, Zong G, Tao R, Wei Z, Lu Y. Crosstalk between traditional Chinese medicine-derived polysaccharides and the gut microbiota: A new perspective to understand traditional Chinese medicine. Phytother Res 2022; 36:4125-4138. [PMID: 36100366 DOI: 10.1002/ptr.7607] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/04/2022] [Accepted: 08/20/2022] [Indexed: 11/09/2022]
Abstract
Polysaccharide is a kind of macromolecule polymer composed of monosaccharides connected by glycosidic bonds. Traditional Chinese medicine (TCM), composed of various bioactive ingredients, is usually rich in polysaccharides. In recent years, extensive research on TCM polysaccharides has demonstrated their pharmacological effects. Polysaccharides can hardly be catabolized by enzymes encoded by the human genome but can be degraded to absorbable metabolites by bacteria inhabiting the colon. Hence, the gut microbiota plays a vital role in degrading TCM polysaccharides into short-chain fatty acids (SCFAs) which exert physiological functions locally and systemically. Besides, TCM polysaccharides can also modulate the composition and activities of the gut microbiota by promoting the growth of beneficial bacteria and inhibiting the colonization of pathogenic bacteria, ultimately restoring gut homeostasis and improving human health. In this review, we discuss the extraction and pharmacological effects of TCM polysaccharides, various functions of the gut microbiota, and the interactions between TCM polysaccharides and the gut microbiota, illuminating the mechanisms of TCM polysaccharides modulating host physiology via the gut microbiota. To firmly establish the clinical efficacy of TCM polysaccharides, further high-quality studies especially clinical trials are needed. Generally, discussion on the interplay between TCM polysaccharides and the gut microbiota is expected to elucidate their application prospects and inspire new thoughts in the development of TCM.
Collapse
Affiliation(s)
- Bingjie Yue
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gangfan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruizhi Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
132
|
Pang K, Yang Y, Chai S, Li Y, Wang X, Sun L, Cui Z, Wang S, Liu S. Dynamics Changes of the Fecal Bacterial Community Fed Diets with Different Concentrate-to-Forage Ratios in Qinghai Yaks. Animals (Basel) 2022; 12:ani12182334. [PMID: 36139194 PMCID: PMC9495249 DOI: 10.3390/ani12182334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 12/02/2022] Open
Abstract
(1) Background: This study aimed to investigate the effects of different dietary concentrate to roughage ratios on growth performance and fecal microbiota composition of yaks by 16S rRNA gene sequencing. (2) Methods: In the present study, three diets with different dietary forage-to-concentrate ratios (50:50, 65:35, and 80:20) were fed to 36 housed male yaks. (3) Results: The result shows that Final BW, TWG, and ADG were higher in the C65 group than in the C50 and C80 groups, but the difference was not significant (p > 0.05). DMI in the C65 group was significantly higher than in the other two groups (p < 0.05). The DMI/ADG of the C65 group was lower than that of the other two groups, but the difference was insignificant (p > 0.05). At the phylum level, Firmicutes were the most abundant in the C65 group, and the relative abundance of Bacteroidetes was lower in the C65 group than in the other two groups. At the genus level, the relative abundances of Ruminococcaceae_UCG_005, Romboutsia, and Christensenellaceae_R-7 were higher in the C56 group than in the C50 and C80 groups. The relative abundance of Lachnospiraceae_NK3A20 and Rikenellaceaewas_RC9_gut is lower in the C65 group, but the difference was insignificant (p > 0.05). At KEGG level 2, the relative abundance of lipid metabolism and energy metabolism were lowest in the C50 group, and both showed higher relative abundance in the C65 group. (4) Conclusions: In conclusion, the structure of fecal microbiota was affected by different concentrate-to-forage ratios. We found that feeding diets with a concentrate-to-forage ratio of 65:35 improved yaks’ growth and energy metabolism.
Collapse
Affiliation(s)
- Kaiyue Pang
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Yingkui Yang
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Shatuo Chai
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Yan Li
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Xun Wang
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Lu Sun
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Zhanhong Cui
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
| | - Shuxiang Wang
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
- Correspondence: (S.W.); (S.L.); Tel.: +86-010-6273-1254 (S.W. & S.L.)
| | - Shujie Liu
- Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Husbandry, Veterinary Sciences in Qinghai University, Xining 810016, China
- Yak Engineering Technology Research Center of Qinghai Province, Xining 810016, China
- Correspondence: (S.W.); (S.L.); Tel.: +86-010-6273-1254 (S.W. & S.L.)
| |
Collapse
|
133
|
Jackson PPJ, Wijeyesekera A, Rastall RA. Determining the metabolic fate of human milk oligosaccharides: it may just be more complex than you think? GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 3:e9. [PMID: 39295778 PMCID: PMC11406381 DOI: 10.1017/gmb.2022.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/10/2022] [Accepted: 08/26/2022] [Indexed: 09/21/2024]
Abstract
Human milk oligosaccharides (HMOs) are a class of structurally diverse and complex unconjugated glycans present in breast milk, which act as selective substrates for several genera of select microbes and inhibit the colonisation of pathogenic bacteria. Yet, not all infants are breastfed, instead being fed with formula milks which may or may not contain HMOs. Currently, formula milks only possess two HMOs: 2'-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT), which have been suggested to be similarly effective as human breast milk in supporting age-related growth. However, the in vivo evidence regarding their ability to beneficially reduce respiratory infections along with altering the composition of an infant's microbiota is limited at best. Thus, this review will explore the concept of HMOs and their metabolic fate, and summarise previous in vitro and in vivo clinical data regarding HMOs, with specific regard to 2'FL and LNnT.
Collapse
Affiliation(s)
| | - Anisha Wijeyesekera
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | | |
Collapse
|
134
|
The Impact of Short-Chain Fatty Acids on Neonatal Regulatory T Cells. Nutrients 2022; 14:nu14183670. [PMID: 36145046 PMCID: PMC9503436 DOI: 10.3390/nu14183670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022] Open
Abstract
Over the first weeks of life, the neonatal gastrointestinal tract is rapidly colonised by a diverse range of microbial species that come to form the ‘gut microbiota’. Microbial colonisation of the neonatal gut is a well-established regulator of several physiological processes that contribute to immunological protection in postnatal life, including the development of the intestinal mucosa and adaptive immunity. However, the specific microbiota-derived signals that mediate these processes have not yet been fully characterised. Accumulating evidence suggests short-chain fatty acids (SCFAs), end-products of intestinal bacterial metabolism, as one of the key mediators of immune development in early life. Critical to neonatal health is the development of regulatory T (Treg) cells that promote and maintain immunological tolerance against self and innocuous antigens. Several studies have shown that SCFAs can induce the differentiation and expansion of Tregs but also mediate pathological effects in abnormal amounts. However, the exact mechanisms through which SCFAs regulate Treg development and pathologies in early life remain poorly defined. In this review, we summarise the current knowledge surrounding SCFAs and their potential impact on the neonatal immune system with a particular focus on Tregs, and the possible mechanisms through which SCFAs achieve their immune modulatory effect.
Collapse
|
135
|
Sharma R, Diwan B, Singh BP, Kulshrestha S. Probiotic fermentation of polyphenols: potential sources of novel functional foods. FOOD PRODUCTION, PROCESSING AND NUTRITION 2022. [DOI: 10.1186/s43014-022-00101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
AbstractFermented functional food products are among the major segments of food processing industry. Fermentation imparts several characteristic effects on foods including the enhancement of organoleptic characteristics, increased shelf-life, and production of novel health beneficial compounds. However, in addition to macronutrients present in the food, secondary metabolites such as polyphenols are also emerging as suitable fermentable substrates. Despite the traditional antimicrobial view of polyphenols, accumulating research shows that polyphenols exert differential effects on bacterial communities by suppressing the growth of pathogenic microbes while concomitantly promoting the proliferation and survival of probiotic bacteria. Conversely, probiotic bacteria not only survive among polyphenols but also induce their fermentation which often leads to improved bioavailability of polyphenols, production of novel metabolic intermediates, increased polyphenolic content, and thus enhanced functional capacity of the fermented food. In addition, selective fermentation of combinations of polyphenol-rich foods or fortification with polyphenols can result in novel functional foods. The present narrative review specifically explores the potential of polyphenols as fermentable substrates in functional foods. We discuss the emerging bidirectional relationship between polyphenols and probiotic bacteria with an aim at promoting the development of novel functional foods based on the amalgamation of probiotic bacteria and polyphenols.
Graphical abstract
Collapse
|
136
|
Zafar M, Alam S, Sabir M, Saba N, Din AU, Ahmad R, Khan MR, Muhammad A, Dayisoylu KS. Isolation, characterization, bacteriocin production and biological potential of Bifidobacteria of ruminants. Anal Biochem 2022; 658:114926. [DOI: 10.1016/j.ab.2022.114926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022]
|
137
|
Mary PR, Monica P, Kapoor M. Insights into β-manno-oligosaccharide uptake and metabolism in Bifidobacterium adolescentis DSMZ 20083 from whole-genome microarray analysis. Microbiol Res 2022; 266:127215. [DOI: 10.1016/j.micres.2022.127215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 10/14/2022]
|
138
|
Viswanath K, Hayes M, Avni D. Inflammatory bowel disease - A peek into the bacterial community shift and algae-based ‘biotic’ approach to combat the disease. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
139
|
Hankel J, Chuppava B, Wilke V, Hartung CB, Muthukumarasamy U, Strowig T, Bach Knudsen KE, Kamphues J, Visscher C. High Dietary Intake of Rye Affects Porcine Gut Microbiota in a Salmonella Typhimurium Infection Study. PLANTS 2022; 11:plants11172232. [PMID: 36079614 PMCID: PMC9460007 DOI: 10.3390/plants11172232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022]
Abstract
Bacterial fermentation of undigested carbohydrates in the hindgut has considerable potential for the stimulation or inhibition of the growth of distinct bacteria within microbiota. The aim of the present study was to evaluate whether high levels of rye affect porcine gut microbiota composition with subsequent effects on the load of Salmonella Typhimurium, an intestinal pathogen with zoonotic relevance. Therefore, forty-two 25-day-old piglets were allocated to two groups and fed a diet containing either 69% wheat or 69% rye for 35 days. One week after introducing the two different diets, the piglets were experimentally infected with Salmonella Typhimurium. The microbiota composition of cecal and fecal samples of the piglets were evaluated 28 days after infection. In the cecum, promoted growth of Bifidobacterium, several lactic acid bacteria and Faecalibacterium prausnitzii were seen in pigs fed the diet containing 69% rye. Bacterial species belonging to the genera Bifidobacterium and Catenisphaera were associated with differing bacterial counts of Salmonella Typhimurium detected in the cecal contents of all piglets in both feeding groups via cultural cultivation. The high intake of rye instead of wheat seems to promote the growth of beneficial intestinal bacteria accompanied by impaired growth conditions for the foodborne pathogen Salmonella Typhimurium.
Collapse
Affiliation(s)
- Julia Hankel
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
- Correspondence:
| | - Bussarakam Chuppava
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Volker Wilke
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Clara Berenike Hartung
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Uthayakumar Muthukumarasamy
- Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
- Hannover Medical School, 30625 Hannover, Germany
| | - Till Strowig
- Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
- Hannover Medical School, 30625 Hannover, Germany
| | | | - Josef Kamphues
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Christian Visscher
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| |
Collapse
|
140
|
Sun Z, Yue Z, Liu E, Li X, Li C. Assessment of the bifidogenic and antibacterial activities of xylooligosaccharide. Front Nutr 2022; 9:858949. [PMID: 36091239 PMCID: PMC9453197 DOI: 10.3389/fnut.2022.858949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Xylooligosaccharide (XOS) is an attractive prebiotic mainly due to its bifidogenic effect. However, commercial XOS with different compositions is often applied in the food industry at different doses without specifications. In this study, we evaluated the bifidogenic activity of XOS at different doses with either mixtures or pure fractions with different degrees of polymerization (DP), using three strains of Bifidobacterium spp., including B. breve ATCC 15700, B. bifidum ATCC 29521, and B. animalis subsp. lactis HN019. Three growth indicators showed strain-specific bifidogenic activity of XOS, and the activity was both dose- and fraction-dependent as only certain fractions stimulated significant growth. Adding 0.25% XOS (w/v) also promoted increase in total bifidobacterial population of rat fecal samples fermented in vitro. Albeit the antibacterial activity of XOS fractions can be demonstrated, significant growth inhibition can only be achieved when 4.0% XOS mixture was added in Staphylococcus aureus ATCC 6538 pure culture. In contrast, in the presence of B. lactis HN019, 1.0% XOS showed significant antibacterial activity against S. aureus ATCC 6538 in milk. In addition, RNA sequencing suggested downregulation of genes involved in S. aureus ATCC 6538 infection, pathogenesis, and quorum sensing, by XOS. In conclusion, the report urges scientific specifications on XOS chemistry for its effective application as a novel food ingredient or functional food and provides novel insights into its bifidogenic and antibacterial activities.
Collapse
Affiliation(s)
- Zhongke Sun
- College of Biological Engineering, Henan University of Technology, Zhengzhou, China
- Institute of Food and Drug Inspection, Zhoukou Normal University, Zhoukou, China
- *Correspondence: Zhongke Sun,
| | - Zonghao Yue
- Institute of Food and Drug Inspection, Zhoukou Normal University, Zhoukou, China
| | - Erting Liu
- Henan Heagreen Bio-technology Co., Ltd., Zhoukou, China
| | - Xianfeng Li
- Henan Heagreen Bio-technology Co., Ltd., Zhoukou, China
| | - Chengwei Li
- College of Biological Engineering, Henan University of Technology, Zhengzhou, China
- Chengwei Li,
| |
Collapse
|
141
|
Xu S, Lane JA, Chen J, Zheng Y, Wang H, Fu X, Huang Q, Dhital S, Liu F, Zhang B. In Vitro Infant Fecal Fermentation Characteristics of Human Milk Oligosaccharides Were Controlled by Initial Microbiota Composition More than Chemical Structure. Mol Nutr Food Res 2022; 66:e2200098. [PMID: 35989465 DOI: 10.1002/mnfr.202200098] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/16/2022] [Indexed: 11/08/2022]
Abstract
SCOPE Human milk oligosaccharides (HMOs), multifunctional glycans naturally present in human milk, are known to contribute to the infant's microbiota and immune system development. However, the molecular specificity of HMOs on microbiota and associated fermentation is not yet fully understood, and is important for the development of infant formula optimum functionality. METHODS AND RESULTS In vitro fermentation is carried out on structurally different HMOs with infant fecal inocula dominated by Bifidobacterium longum, Bifidobacterium breve, and Bacteroides. The gas, metabolite (SCFA, lactate, and succinate) profiles, and microbiota responses differ between individual microbiota inocula patterns regardless of HMO structure. In terms of HMO pairs with same sugar composition but different glycosidic bonds, gas and metabolite profiles are similar with the B. longum- and B. breve-dominated inocula. However, large individual variations are observed with the Bacteroides-dominated inocula. The microbial communities at the end of fermentation are closely related to the initial microbiota composition. CONCLUSION The findings demonstrate that short-term in vitro fermentation outcomes largely depend on the initial gut microbiota composition more than the impact of HMO molecular specificity. These results advance the current understanding for the design of personalized infant nutritional solutions and therapies in future.
Collapse
Affiliation(s)
- Shiqi Xu
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
| | - Jonathan A Lane
- H&H Group, H&H Research, Global Research and Technology Centre, P61 K202 Co, Cork, Ireland
| | - Juchun Chen
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Yuxing Zheng
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Hongwei Wang
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Xiong Fu
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
| | - Qiang Huang
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China.,Sino-Singapore International Research Institute, Guangzhou, 510555, China
| | - Sushil Dhital
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Feitong Liu
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Bin Zhang
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China.,Sino-Singapore International Research Institute, Guangzhou, 510555, China
| |
Collapse
|
142
|
Ying C, Siao YS, Chen WJ, Chen YT, Chen SL, Chen YL, Hsu JT. Host species and habitats shape the bacterial community of gut microbiota of three non-human primates: Siamangs, white-handed gibbons, and Bornean orangutans. Front Microbiol 2022; 13:920190. [PMID: 36051771 PMCID: PMC9424820 DOI: 10.3389/fmicb.2022.920190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The gut microbiome is essential for a host to digest food, maintain health, and adapt to environments. Bacterial communities of gut microbiota are influenced by diverse factors including host physiology and the environment. Many non-human primates (NHPs), which are physiologically close to humans, are in danger of extinction. In this study, the community structure of the gut microbiota in three NHPs: siamangs (Symphalangus syndactylus, Ss), Bornean orangutans (Pongo pygmaeus, Pp), and white-handed gibbons (Hylobates lar, Hl)—housed at the largest Zoo in Taiwan were analyzed. Pp and Ss were housed in the Asian tropical rainforest area, while Hl was housed in two separate areas, the Asian tropical rainforest area and the conservation area. Bacterial community diversity of Ss, indicated by the Shannon index, was significantly higher compared with that of Hl and Pp, while the richness (Chao 1) and observed operational taxonomic units (OTUs) were similar across the three species of NHPs. Host species was the dominant factor shaping the gut microbial community structure. Beta-diversity analysis including non-metric multidimensional scaling (NMDS) and unweighted pair group method with arithmetic mean (UPGMA) suggested gut bacterial communities of Hl housed in the conservation area were closely related to each other, while the bacterial communities of Hl in the rainforest area were dispersedly positioned. Further analysis revealed significantly higher abundances of Lactobacillus fermentum, L. murinus, and an unclassified species of Lactobacillus, and a lower abundance of Escherichia-Shigella in Hl from the conservation area relative to the rainforest area. The ratio of Lactobacillus to Escherichia-Shigella was 489.35 and 0.013 in Hl inhabiting the conservation and rainforest areas, respectively. High abundances of Lactobacillus and Bifidobacterium and a high ratio of Lactobacillus to Escherichia-Shigella were also observed in one siamang with notable longevity of 53 years. Data from the study reveal that host species acted as the fundamental driving factor in modulating the community structure of gut microbiota, but that habitats also acted as key determinants within species. The presence and high abundance of probiotics, such as Bifidobacterium and Lactobacillus, provide potential indicators for future diet and habitat optimization for NHPs, especially in zoological settings.
Collapse
Affiliation(s)
- Chingwen Ying
- Department of Microbiology, Soochow University, Taipei, Taiwan
- *Correspondence: Chingwen Ying
| | - You-Shun Siao
- Department of Microbiology, Soochow University, Taipei, Taiwan
| | - Wun-Jing Chen
- Department of Microbiology, Soochow University, Taipei, Taiwan
| | | | | | - Yi-Lung Chen
- Department of Microbiology, Soochow University, Taipei, Taiwan
| | - Jih-Tay Hsu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Jih-Tay Hsu
| |
Collapse
|
143
|
Influence of Sugar Beet Pulp Supplementation on Pigs’ Health and Production Quality. Animals (Basel) 2022; 12:ani12162041. [PMID: 36009631 PMCID: PMC9404422 DOI: 10.3390/ani12162041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Fibrous feedstuffs can have a variable effect on pig growth, health and meat quality. The effect of sugar beet pulp (SBP) supplementation in the diet on pork quality has not been widely reported. This study examines the effect of an SBP-supplemented (3%) diet (TG-I group) on 300 Large White/Norwegian Landrace pigs in terms of growth performance, blood parameters, microbial profiling of faeces, carcass parameters and meat quality, including the profiles of biogenic amines (BAs), fatty acids (FAs) and volatile compounds (VCs). After 163 days of the experiment, TG-I pigs had a significantly lower average daily gain and feed conversion ratio than pigs in the control group, as well as a significantly higher percentage of carcasses in the S and KN classes and a lower percentage in the E and U classes (p ≤ 0.05). Faeces of TG-I contained significantly more bacteria that are considered probiotic. Significant differences (p ≤ 0.05) were found in most of the blood parameters, FA, VC profile and emotional responses between the two groups. Higher drip loss, protein content and redness, as well as lower cooking loss, intramuscular fat content and lightness were observed in the meat of TG-I. Most of the sensory properties, as well as overall acceptability, were rated higher for the meat of TG-I. Based on the results, a diet containing 3% of SBP could be beneficial for the improvement of pigs’ gut health and pork quality. However, further studies are needed to indicate which compounds of the SBP dietary fiber are responsible for these desirable changes.
Collapse
|
144
|
Zhong Z, Zhang Y, Li X, Li L, Zhang R, Zhang S. Differential Responses of Digesta- and Mucosa-Associated Jejunal Microbiota of Hu Sheep to Pelleted and Non-Pelleted High-Grain Diets. Animals (Basel) 2022; 12:ani12131695. [PMID: 35804593 PMCID: PMC9264909 DOI: 10.3390/ani12131695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 11/28/2022] Open
Abstract
In the present study, we utilized 16S rRNA sequencing to uncover the impacts of non-pelleted (HG) or high-grain pelleted (HP) diets on the microbial structure and potential functions of digesta- and mucosa-associated microbiota in the jejunum of Hu sheep. Here, we randomly assigned 15 healthy male Hu sheep into three groups and fed the control diets (CON), HG, and HP diets, respectively. The experiment period was 60 days. The HP diets had the same nutritional ingredients as the HG diets but in pelleted form. At the finish of the experiment, the jejunal digesta and mucosa were gathered for microbial sequencing. The results of PCoA and PERMANOVA showed that different dietary treatments had significant impact (p < 0.05) on digesta- and mucosa-associated microbiota in the jejunum of Hu sheep. For specific differences, HG diets significantly increased (p < 0.05) the abundance of some acid-producing bacteria in both jejunal digesta (Bifidobacterium, OTU151, and OTU16) and mucosa (Rikenellaceae RC9 gut group, and Bifidobacterium) of Hu sheep compared with the CON diets. Besides the similar effects of the HG diets (increased the acid-producing bacteria such as Olsenella, Pseudoramibacter, and Shuttleworthia), our results also showed that the HP diets significantly decreased (p < 0.05) the abundance of some pro-inflammatory bacteria in the jejunal digesta (Mogibacterium, and Marvinbryantia) and mucosa (Chitinophaga, and Candidatus Saccharimonas) of Hu sheep compared with the HG diets. Collectively, these findings contributed to enriching the knowledge about the effects of HG diets on the structure and function of intestinal microbiota in ruminants.
Collapse
|
145
|
Xue H, Ma J, Wang Y, Lu M, Wang F, Tang X. Shen-Ling-Bai-Zhu-San (SL) and SL Derived-Polysaccharide (PL) Ameliorate the Severity of Diarrhea-Induced by High Lactose via Modification of Colonic Fermentation. Front Pharmacol 2022; 13:883355. [PMID: 35837289 PMCID: PMC9273845 DOI: 10.3389/fphar.2022.883355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 11/28/2022] Open
Abstract
In our previous study, we demonstrated that Shen-ling-bai-zhu-san (SL), a classical Chinese herbal formula, could alleviate lactose-induced diarrhea. However, little is known about the mechanism underlying SL action or the efficacy of the polysaccharide (PL) derived from SL. In this study, we investigated the effect of SL and PL on improving the dysregulated luminal and mucosal microbiota in rats with high lactose diet using 16S rRNA analysis. The concentrations of lactose, lactic acid in cecum and short-chain fatty acids (SCFAs) in cecum and portal vein were measured, meanwhile the expression of ion transporters were ascertained. Our data suggest that the SL, PL and cecal microbiota transplantation (CMT) significantly decreased fecal water content and water intake. In the luminal microbiota there was a significant increase in Akkermansia, Bifidobacterium and Blautia and a lower abundance of Lactobacillus, Escherichia-Shigella, and Dubosiella, while the mucosal microbiota showed a significant increase in Bifidobacterium, Akkermansia, Albaculum, Bilophila, and Coriobacteriaceae_UCG-002 and a lower abundance of Enterococcus, Helicobacter, Dubosiella, and Collinsella. Furthermore, the treatments enhanced lactose fermentation and SCFA production, which may be related to the modulation of the luminal microbial community. A lower ratio of phosphorylation Na/H exchanger3/Na/H exchanger3 (pNHE3/NHE3) and a higher sodium monocarboxylate1 (sMCT1) expression were found in the treatment group than in the model group, which may be related to the changes in the mucosal microbial community. Also, the treatments may restore the impacted metabolic pathways of gut microbiota. These results provide an important foundation for mechanism of SL action and developing PL-based treatment for lactose-induced diarrhea.
Collapse
Affiliation(s)
- Hong Xue
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Hong Xue, ; Xudong Tang, ; Fengyun Wang,
| | - Jinxin Ma
- Department of Integrated Traditional Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- Department of Gastrointestinal Medicine, Peking University Traditional Chinese Medicine Clinical Medican School (Xiyuan), Beijing, China
| | - Yitian Wang
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengxiong Lu
- Department of Integrated Traditional Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- Department of Gastrointestinal Medicine, Peking University Traditional Chinese Medicine Clinical Medican School (Xiyuan), Beijing, China
| | - Fengyun Wang
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Hong Xue, ; Xudong Tang, ; Fengyun Wang,
| | - Xudong Tang
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Integrated Traditional Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- Department of Gastrointestinal Medicine, Peking University Traditional Chinese Medicine Clinical Medican School (Xiyuan), Beijing, China
- *Correspondence: Hong Xue, ; Xudong Tang, ; Fengyun Wang,
| |
Collapse
|
146
|
Lopez-Tello J, Schofield Z, Kiu R, Dalby MJ, van Sinderen D, Le Gall G, Sferruzzi-Perri AN, Hall LJ. Maternal gut microbiota Bifidobacterium promotes placental morphogenesis, nutrient transport and fetal growth in mice. Cell Mol Life Sci 2022; 79:386. [PMID: 35760917 PMCID: PMC9236968 DOI: 10.1007/s00018-022-04379-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 12/22/2022]
Abstract
The gut microbiota plays a central role in regulating host metabolism. While substantial progress has been made in discerning how the microbiota influences host functions post birth and beyond, little is known about how key members of the maternal gut microbiota can influence feto-placental growth. Notably, in pregnant women, Bifidobacterium represents a key beneficial microbiota genus, with levels observed to increase across pregnancy. Here, using germ-free and specific-pathogen-free mice, we demonstrate that the bacterium Bifidobacterium breve UCC2003 modulates maternal body adaptations, placental structure and nutrient transporter capacity, with implications for fetal metabolism and growth. Maternal and placental metabolome were affected by maternal gut microbiota (i.e. acetate, formate and carnitine). Histological analysis of the placenta confirmed that Bifidobacterium modifies placental structure via changes in Igf2P0, Dlk1, Mapk1 and Mapk14 expression. Additionally, B. breve UCC2003, acting through Slc2a1 and Fatp1-4 transporters, was shown to restore fetal glycaemia and fetal growth in association with changes in the fetal hepatic transcriptome. Our work emphasizes the importance of the maternal gut microbiota on feto-placental development and sets a foundation for future research towards the use of probiotics during pregnancy.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Zoe Schofield
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Raymond Kiu
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Matthew J Dalby
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | | | - Gwénaëlle Le Gall
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Lindsay J Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK.
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
147
|
So D, Loughman A, Staudacher HM. Effects of a low FODMAP diet on the colonic microbiome in irritable bowel syndrome: a systematic review with meta-analysis. Am J Clin Nutr 2022; 116:943-952. [PMID: 35728042 PMCID: PMC9535515 DOI: 10.1093/ajcn/nqac176] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND A low fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet is increasingly used to manage symptoms in irritable bowel syndrome (IBS). Although this approach may alter the colonic microbiome, the nature of these changes has not been comprehensively synthesized. OBJECTIVES The aim of this study was to conduct a systematic review with meta-analysis of randomized controlled trials examining the impact of a low FODMAP diet on the composition and function of the microbiome in patients with IBS. METHODS A systematic search was conducted for randomized controlled trials evaluating the effects of a low FODMAP diet on the colonic microbiome in patients with IBS in MEDLINE, EMBASE, CENTRAL, and Web of Science from inception to April 2022. Outcomes included diversity of the microbiome, specific bacterial abundances, fecal SCFA concentration, and fecal pH. For fecal SCFA concentrations and pH, meta-analyses were performed via a random-effects model. RESULTS Nine trials involving 403 patients were included. There were no clear effects of the low FODMAP diet on diversity of the microbiome. A low FODMAP diet consistently led to lower abundance of Bifidobacteria, but there were no clear effects on diversity of the microbiome or abundances of other specific taxa. There were no differences in total fecal SCFA concentration between the low FODMAP diet and control diets (standardized mean difference: -0.25; 95% CI: -0.63, 0.13; P = 0.20), nor were there differences for fecal concentrations of specific SCFAs or fecal pH. CONCLUSIONS In patients with IBS, the effects of a low FODMAP diet on the colonic microbiome appear to be specific to Bifidobacteria with no consistent impacts on other microbiome metrics, including diversity, fecal SCFA concentrations, and fecal pH. Further, adequately powered trials are needed to confirm these findings.This review was registered at https://www.crd.york.ac.uk/prospero/ as CRD42020192243.
Collapse
Affiliation(s)
- Daniel So
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Amy Loughman
- Food & Mood Centre, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Victoria, Australia
| | | |
Collapse
|
148
|
Kang JW, Tang X, Walton CJ, Brown MJ, Brewer RA, Maddela RL, Zheng JJ, Agus JK, Zivkovic AM. Multi-Omic Analyses Reveal Bifidogenic Effect and Metabolomic Shifts in Healthy Human Cohort Supplemented With a Prebiotic Dietary Fiber Blend. Front Nutr 2022; 9:908534. [PMID: 35782954 PMCID: PMC9248813 DOI: 10.3389/fnut.2022.908534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
Dietary fiber, a nutrient derived mainly from whole grains, vegetables, fruits, and legumes, is known to confer a number of health benefits, yet most Americans consume less than half of the daily recommended amount. Convenience and affordability are key factors determining the ability of individuals to incorporate fiber-rich foods into their diet, and many Americans struggle to access, afford, and prepare foods rich in fiber. The objective of this clinical study was to test the changes in microbial community composition, human metabolomics, and general health markers of a convenient, easy to use prebiotic supplement in generally healthy young participants consuming a diet low in fiber. Twenty healthy adults participated in this randomized, placebo-controlled, double-blind, crossover study which was registered at clinicaltrials.gov as NCT03785860. During the study participants consumed 12 g of a prebiotic fiber supplement and 12 g of placebo daily as a powder mixed with water as part of their habitual diet in randomized order for 4 weeks, with a 4-week washout between treatment arms. Fecal microbial DNA was extracted and sequenced by shallow shotgun sequencing on an Illumina NovaSeq. Plasma metabolites were detected using liquid chromatography–mass spectrometry with untargeted analysis. The phylum Actinobacteria, genus Bifidobacterium, and several Bifidobacterium species (B. bifidum, B. adolescentis, B. breve, B. catenulatum, and B. longum) significantly increased after prebiotic supplementation when compared to the placebo. The abundance of genes associated with the utilization of the prebiotic fiber ingredients (sacA, xfp, xpk) and the production of acetate (poxB, ackA) significantly changed with prebiotic supplementation. Additionally, the abundance of genes associated with the prebiotic utilization (xfp, xpk), acetate production (ackA), and choline to betaine oxidation (gbsB) were significantly correlated with changes in the abundance of the genus Bifidobacterium in the prebiotic group. Plasma concentrations of the bacterially produced metabolite indolepropionate significantly increased. The results of this study demonstrate that an easy to consume, low dose (12 g) of a prebiotic powder taken daily increases the abundance of beneficial bifidobacteria and the production of health-promoting bacteria-derived metabolites in healthy individuals with a habitual low-fiber diet.
Collapse
Affiliation(s)
- Jea Woo Kang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | | | - Mark J. Brown
- USANA Health Sciences, Inc., Salt Lake City, UT, United States
| | | | | | - Jack Jingyuan Zheng
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Joanne K. Agus
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Angela M. Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, United States
- *Correspondence: Angela M. Zivkovic
| |
Collapse
|
149
|
The Comparative Analysis of Genomic Diversity and Genes Involved in Carbohydrate Metabolism of Eighty-Eight Bifidobacterium pseudocatenulatum Isolates from Different Niches of China. Nutrients 2022; 14:nu14112347. [PMID: 35684146 PMCID: PMC9183100 DOI: 10.3390/nu14112347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/03/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
Eighty-eight Bifidobacterium pseudocatenulatum strains, which were isolated from human, chicken and cow fecal samples from different niches of China, were compared genomically in this study to evaluate their diversity. It was found that B. pseudocatenulatum displayed a closed pan-genome, including abundant glycoside hydrolase families of the carbohydrate active enzyme (CAZy). A total of 30 kinds of glycoside hydrolases (GHs), 14 kinds of glycosyl transferases (GTs), 13 kinds of carbohydrate-binding modules (CBMs), 6 kinds of carbohydrate-esterases (CEs), and 2 kinds of auxiliary activities (AAs) gene families were identified across the genomes of the 88 B. pseudocatenulatum strains. Specifically, this showed that significant differences were also present in the number of 10 carbohydrate-active enzyme gene families (GT51, GH13_32, GH26, GH42, GH121, GH3, AA3, CBM46, CE2, and CE6) among the strains derived from the hosts of different age groups, particularly between strains from infants and those from other human age groups. Twelve different individuals of B. pseudocatenulatum from four main clusters were selected for further study to reveal the genetic diversity of carbohydrate metabolism-related genes within the same phylogenetics. The animal experiment showed that 3 weeks of oral administration and 1 week after cessation of administration of these strains did not markedly alter the serum routine inflammatory indicators in mice. Furthermore, the administration of these strains did not significantly cause adverse changes in the gut microbiota, as indicated by the α- and β-diversity indexes, relative to the control group (normal diet). Beyond that, FAHBZ9L5 significantly increased the abundance of B. pseudocatenulatum after 3 weeks and significantly increased the abundance of acetic acid and butyric acid in the host’s intestinal tract 3 and 4 weeks after the first administration, respectively, compared with the control group. Corresponding to this, comparative genomic analyses of 12 B. pseudocatenulatum suggest that FAHBZ9L5-specific genes were rich in ABC transporters and carbohydrate esterase. Combining the results of comparative genomics analyses and animal experiment, it is suggested that the strains containing certain gene clusters contribute to another competitive growth advantage of B. pseudocatenulatum, which facilitates its intestinal carbohydrate metabolism in a host.
Collapse
|
150
|
Yao H, Lu S, Williams BA, Flanagan BM, Gidley MJ, Mikkelsen D. Absolute abundance values reveal microbial shifts and co-occurrence patterns during gut microbiota fermentation of dietary fibres in vitro. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2021.107422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|