1
|
Yoon KN, Choi YH, Keum GB, Yeom SJ, Kim SS, Kim ES, Park HJ, Kim JE, Park JH, Song BS, Eun JB, Park SH, Lee JH, Lee JH, Kim HB, Kim JK. Lactiplantibacillus argentoratensis AGMB00912 alleviates diarrhea and promotes the growth performance of piglets during the weaning transition. BMC Microbiol 2024; 24:404. [PMID: 39390387 PMCID: PMC11465746 DOI: 10.1186/s12866-024-03536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Preventing post-weaning diarrhea (PWD) in weaned piglets is a crucial challenge in the swine production industry. The stress of weaning, dietary shifts from maternal milk to solid feed, and environmental changes lead to decreased microbial diversity, increased pathogen abundance, and compromised intestinal integrity. We have previously identified Lactiplantibacillus argentoratensis AGMB00912 (LA) in healthy porcine feces, which demonstrated antimicrobial activity against pathogens and enhanced short-chain fatty acid production. This research aimed to evaluate the efficacy of LA strain supplementation as a strategy to inhibit PWD and enhance overall growth performance in weaned piglets. RESULTS LA supplementation in weaned piglets significantly increased body weight gain, average daily gain, and average daily feed intake. It also alleviated diarrhea symptoms (diarrhea score and incidence). Notably, LA was found to enrich beneficial microbial populations (Lactobacillus, Anaerobutyricum, Roseburia, Lachnospiraceae, and Blautia) while reducing the abundance of harmful bacteria (Helicobacter and Campylobacter). This not only reduces the direct impact of pathogens but also improves the overall gut microbiota structure, thus enhancing the resilience of weaned piglets. LA treatment also promotes the growth of the small intestinal epithelial structure, strengthens gut barrier integrity, and increases short-chain fatty acid levels in the gut. CONCLUSIONS The study findings demonstrate the promising potential of LA in preventing PWD. Supplementation with the LA strain offers a promising feed additive for improving intestinal health and growth in piglets during the weaning transition, with the potential to significantly reduce the incidence and severity of PWD.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, 56212, Republic of Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yo-Han Choi
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Gi Beom Keum
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, 56212, Republic of Korea
| | - Sang-Su Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, 56212, Republic of Korea
| | - Eun Sol Kim
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hyun Ju Park
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Jo Eun Kim
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Jong-Heum Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, 56212, Republic of Korea
| | - Beom-Seok Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, 56212, Republic of Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, 56212, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, 56212, Republic of Korea
| | - Ju-Hoon Lee
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, 56212, Republic of Korea.
| |
Collapse
|
2
|
Koch F, Albrecht D, Albrecht E, Hansen C, Kuhla B. Novel Perspective on Molecular and Cellular Adaptations of the Mammary Gland-Regulating Milk Constituents and Immunity of Heat-Stressed Dairy Cows. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20286-20298. [PMID: 39226405 PMCID: PMC11421017 DOI: 10.1021/acs.jafc.4c03879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Climate change with increasing ambient temperatures negatively influences the biology of dairy cows and their milk production in the mammary gland (MG). This study aimed to elucidate the MG proteome, differences in milk composition, and ruminal short-chain fatty acid concentrations of dairy cows experiencing 7 days of heat stress [HS, 28 °C, temperature humidity index (THI) = 76], pair-feeding (PF), or ad libitum feeding (CON) at thermoneutrality (16 °C, THI = 60). Ruminal acetate, acetate/propionate ratio, and milk urea concentrations were greater, whereas milk protein and lactose were lower in HS than in control cows. Proteome analysis revealed an induced bacterial invasion of epithelial cells, leukocyte transendothelial migration, reduction of the pyruvate and carbon metabolism, and platelet activation in the MG of HS compared to CON or PF cows. These results highlight adaptive metabolic and immune responses to mitigate the negative effects of ambient heat in the MG.
Collapse
Affiliation(s)
- Franziska Koch
- Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Dirk Albrecht
- Department for Microbial Physiology and Molecular Biology, University of Greifswald, Greifswald 17489, Germany
| | - Elke Albrecht
- Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Christiane Hansen
- Mecklenburg-Vorpommern Research Centre for Agriculture and Fisheries, Institute of Livestock Farming, Dummerstorf 18196, Germany
| | - Björn Kuhla
- Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| |
Collapse
|
3
|
Yoon KN, Yang J, Yeom SJ, Kim SS, Park JH, Song BS, Eun JB, Park SH, Lee JH, Kim HB, Lee JH, Kim JK. Lactiplantibacillus argentoratensis AGMB00912 protects weaning mice from ETEC infection and enhances gut health. Front Microbiol 2024; 15:1440134. [PMID: 39318427 PMCID: PMC11420142 DOI: 10.3389/fmicb.2024.1440134] [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: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Maintaining a healthy intestinal environment, optimal epithelial barrier integrity, and balanced gut microbiota composition are essential for the growth performance of weaning pigs. We identified Lactiplantibacillus argentoratensis AGMB00912 (LA) in healthy porcine feces as having antimicrobial activity against pathogens and enhanced short-chain fatty acid (SCFA) production. Herein, we assess the protective role of LA using a weaning mouse model with enterotoxigenic Escherichia coli (ETEC) infection. LA treatment improves feed intake and weight gain and alleviates colon shortening. Furthermore, LA inhibits intestinal damage, increases the small intestine villus height compared with the ETEC group, and enhances SCFA production. Using the Kyoto Encyclopedia of Genes and Genomes and other bioinformatic tools, including InterProScan and COGNIZER, we validated the presence of SCFA-producing pathways of LA and Lactiplantibacillus after whole genome sequencing. LA mitigates ETEC-induced shifts in the gut microbiota, decreasing the proportion of Escherichia and Enterococcus and increasing SCFA-producing bacteria, including Kineothrix, Lachnoclostridium, Roseuburia, Lacrimispora, Jutongia, and Blautia. Metabolic functional prediction analysis revealed enhanced functions linked to carbohydrate, amino acid, and vitamin biosynthesis, along with decreased functions associated with infectious bacterial diseases compared to the ETEC group. LA mitigates the adverse effects of ETEC infection in weaning mice, enhances growth performance and intestinal integrity, rebalances gut microbiota, and promotes beneficial metabolic functions. These findings validate the functionality of LA in a small animal model, supporting its potential application in improving the health and growth performance of weaning pigs.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Jihye Yang
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Sang-Su Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Heum Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Beom-Seok Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Ju-Hoon Lee
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| |
Collapse
|
4
|
Wang J, Shi L, Zhang X, Hu R, Yue Z, Zou H, Peng Q, Jiang Y, Wang Z. Metabolomics and proteomics insights into subacute ruminal acidosis etiology and inhibition of proliferation of yak rumen epithelial cells in vitro. BMC Genomics 2024; 25:394. [PMID: 38649832 PMCID: PMC11036571 DOI: 10.1186/s12864-024-10242-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Untargeted metabolomics and proteomics were employed to investigate the intracellular response of yak rumen epithelial cells (YRECs) to conditions mimicking subacute rumen acidosis (SARA) etiology, including exposure to short-chain fatty acids (SCFA), low pH5.5 (Acid), and lipopolysaccharide (LPS) exposure for 24 h. RESULTS These treatments significantly altered the cellular morphology of YRECs. Metabolomic analysis identified significant perturbations with SCFA, Acid and LPS treatment affecting 259, 245 and 196 metabolites (VIP > 1, P < 0.05, and fold change (FC) ≥ 1.5 or FC ≤ 0.667). Proteomic analysis revealed that treatment with SCFA, Acid, and LPS resulted in differential expression of 1251, 1396, and 242 proteins, respectively (FC ≥ 1.2 or ≤ 0.83, P < 0.05, FDR < 1%). Treatment with SCFA induced elevated levels of metabolites involved in purine metabolism, glutathione metabolism, and arginine biosynthesis, and dysregulated proteins associated with actin cytoskeleton organization and ribosome pathways. Furthermore, SCFA reduced the number, morphology, and functionality of mitochondria, leading to oxidative damage and inhibition of cell survival. Gene expression analysis revealed a decrease the genes expression of the cytoskeleton and cell cycle, while the genes expression associated with inflammation and autophagy increased (P < 0.05). Acid exposure altered metabolites related to purine metabolism, and affected proteins associated with complement and coagulation cascades and RNA degradation. Acid also leads to mitochondrial dysfunction, alterations in mitochondrial integrity, and reduced ATP generation. It also causes actin filaments to change from filamentous to punctate, affecting cellular cytoskeletal function, and increases inflammation-related molecules, indicating the promotion of inflammatory responses and cellular damage (P < 0.05). LPS treatment induced differential expression of proteins involved in the TNF signaling pathway and cytokine-cytokine receptor interaction, accompanied by alterations in metabolites associated with arachidonic acid metabolism and MAPK signaling (P < 0.05). The inflammatory response and activation of signaling pathways induced by LPS treatment were also confirmed through protein interaction network analysis. The integrated analysis reveals co-enrichment of proteins and metabolites in cellular signaling and metabolic pathways. CONCLUSIONS In summary, this study contributes to a comprehensive understanding of the detrimental effects of SARA-associated factors on YRECs, elucidating their molecular mechanisms and providing potential therapeutic targets for mitigating SARA.
Collapse
Affiliation(s)
- JunMei Wang
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liyuan Shi
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaohong Zhang
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Rui Hu
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ziqi Yue
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Huawei Zou
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Quanhui Peng
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yahui Jiang
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhisheng Wang
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
5
|
Liu J, Gu QH, Cui Z, Zhao MH, Jia XY. Short-chain fatty acids ameliorate experimental anti-glomerular basement membrane disease. Clin Immunol 2024; 259:109903. [PMID: 38218211 DOI: 10.1016/j.clim.2024.109903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
BACKGROUND Short-chain fatty acids (SCFAs), as the link between gut microbiota and the immune system, had been reported to be protective in many autoimmune diseases by the modulation of T cell differentiation. The pathogenic role of autoreactive Th1 and Th17 cells and the protective role of Treg cells in the pathogenesis of anti-GBM disease have been fully demonstrated. Thus, the present study aimed to investigate the therapeutic effects of SCFAs in a rat model of anti-GBM disease. MATERIALS AND METHODS Experimental anti-GBM disease was constructed by immunizing Wistar Kyoto rats with a nephrogenic T cell epitope α3127-148, and intervened by sodium acetate, sodium propionate, or sodium butyrate, 150 mM in the drinking water from day 0 to 42. Kidney injury was accessed by the biochemical analyzer, immunofluorescence, and immunohistochemistry. Antibody response was detected by ELISA. T cell clustering and proliferation were detected by flow cytometry. Human kidney 2 (HK2) cells were stimulated in vitro and cytokines were assessed by quantitative real-time PCR. RESULTS Treatment with sodium acetate, sodium propionate, or sodium butyrate ameliorated the severity of kidney impairment in rats with anti-GBM glomerulonephritis. In the sodium butyrate-treated rats, the urinary protein, serum creatinine, and blood urea nitrogen levels were significantly lower; the percentage of crescent formation in glomeruli was significantly reduced; and the kidneys showed reduced IgG deposition, complement activation, T cell, and macrophage infiltration as well as the level of circulating antibodies against anti-α3(IV)NC1. The treatment of sodium butyrate reduced the α3127-148-specific T cell activation and increased the Treg cells differentiation and the intestinal beneficial bacteria flora. It also alleviated the damage of HK2 cells treated with inflammatory factors and complement. CONCLUSION Treatment with SCFAs, especially butyrate, alleviated anti-GBM nephritis in rat model, indicating its potential therapeutic effects in clinical usage.
Collapse
Affiliation(s)
- Jing Liu
- Renal Division, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Qiu-Hua Gu
- Renal Division, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China; Nephrology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhao Cui
- Renal Division, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Yu Jia
- Renal Division, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Qiu B, Shen Z, Yang D, Qin X, Ren W, Wang Q. Gut microbiota and common gastrointestinal diseases: a bidirectional two-sample Mendelian randomized study. Front Microbiol 2023; 14:1273269. [PMID: 38045030 PMCID: PMC10691374 DOI: 10.3389/fmicb.2023.1273269] [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: 08/05/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Background Several recent studies have shown an association between gut microbiota and gastrointestinal diseases. However, the causal relationship between gut microbiota and gastrointestinal disorders is unclear. Methods We assessed causal relationships between gut microbiota and eight common gastrointestinal diseases using Mendelian randomization (MR) analyses. IVW results were considered primary results. Cochrane's Q and MR-Egger tests were used to test for heterogeneity and pleiotropy. Leave-one-out was used to test the stability of the MR results, and Bonferroni correction was used to test the strength of the causal relationship between exposure and outcome. Results MR analyses of 196 gut microbiota and eight common gastrointestinal disease phenotypes showed 62 flora and common gastrointestinal diseases with potential causal relationships. Among these potential causal relationships, after the Bonferroni-corrected test, significant causal relationships remained between Genus Oxalobacter and CD (OR = 1.29, 95% CI: 1.13-1.48, p = 2.5 × 10-4, q = 4.20 × 10-4), and between Family Clostridiaceae1 and IBS (OR = 0.9967, 95% CI: 0.9944-0.9991, p = 1.3 × 10-3, q = 1.56 × 10-3). Cochrane's Q-test showed no significant heterogeneity among the various single nucleotide polymorphisms (SNPs). In addition, no significant level of pleiotropy was found according to the MR-Egger. Conclusion This study provides new insights into the mechanisms of gut microbiota-mediated gastrointestinal disorders and some guidance for targeting specific gut microbiota for treating gastrointestinal disorders.
Collapse
Affiliation(s)
- Binxu Qiu
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Zixiong Shen
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dongliang Yang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xinxin Qin
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Wenyong Ren
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Quan Wang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Rodrigues Pessoa E, Roger Vasconcelos F, de Oliveira Paula-Marinho S, de Menezes Daloso D, Damasceno Guerreiro D, Matias Martins JA, Gomes-Filho E, Alencar Moura A. Metabolomic profile of seminal plasma from Guzerá bulls (Bos indicus) with contrasting sperm freezability phenotypes. Reprod Domest Anim 2023; 58:1379-1392. [PMID: 37592767 DOI: 10.1111/rda.14453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/20/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023]
Abstract
The present study evaluated the seminal plasma metabolome of Bos indicus Guzerá bulls with good (n = 4) and poor (n = 5) sperm freezability. Animals were raised in natural pasture of a 'Caatinga' ecosystem, in the semi-arid region of Brazil. Seminal plasma samples were subjected to gas chromatography coupled to mass spectrometry and data, analysed using bioinformatics tools (Cytoscape with the MetScape plug-in). Sixty-two metabolites were identified in the bovine seminal plasma. Fatty acids and conjugates and organic compounds were the predominant seminal fluid metabolites, followed by carboxylic acids and derivatives, amino acids, benzenes and steroids and derivatives, carbohydrates and carbohydrate conjugates and prenol lipids. Multivariate analysis indicated a distinct separation of seminal plasma metabolomes from bulls with contrasting sperm freezability. Abundances of propanoic acid, d-ribose and glycine were greater in the seminal plasma of bulls with good sperm freezability. Heptadecanoic acid and undecanoic acid were the predominant in bulls of poor sperm freezability. Propanoic acid is an energy source for spermatozoa and may act as an antimicrobial component in semen. Glycine acts against oxidizing and denaturing reactions. d-ribose is also an energy source and reduces apoptosis and oxidative stress. Undecanoic acid may protect sperm against fungal damage. This study provides fundamental information approximately the seminal plasma metabolome of tropically adapted bulls and its association with sperm freezability. However, further studies with larger groups of animals are needed to validate those metabolites as markers of sperm freezability. This strategy could support the selection of sires with superior sperm cryoresistance.
Collapse
Affiliation(s)
| | | | | | - Danilo de Menezes Daloso
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Jorge André Matias Martins
- Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
- Center for Agricultural Sciences and Biodiversity, Federal University of Cariri, Crato, Brazil
| | - Enéas Gomes-Filho
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | | |
Collapse
|
8
|
Enichen E, Adams RB, Demmig-Adams B. Physical Activity as an Adjunct Treatment for People Living with HIV? Am J Lifestyle Med 2023; 17:502-517. [PMID: 37426740 PMCID: PMC10328202 DOI: 10.1177/15598276221078222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
This review evaluates physical activity as a candidate for an adjunct treatment, in conjunction with antiretroviral therapy (ART), for people living with HIV (PLWH). Evidence is summarized that chronic, non-resolving inflammation (a principal feature of immune system dysfunction) and a dysfunctional state of the gut environment are key factors in HIV infection that persist despite treatment with ART. In addition, evidence is summarized that regular physical activity may restore normal function of both the immune system and the gut environment and may thereby ameliorate symptoms and non-resolving inflammation-associated comorbidities that burden PLWH. Physicians who care for PLWH could thus consider incorporating physical activity into treatment plans to complement ART. It is also discussed that different types of physical activity can have different effects on the gut environment and immune function, and that future research should establish more specific criteria for the design of exercise regimens tailored to PLWH.
Collapse
Affiliation(s)
- Elizabeth Enichen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| | - Robert B. Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| |
Collapse
|
9
|
Liu T, Sun Z, Yang Z, Qiao X. Microbiota-derived short-chain fatty acids and modulation of host-derived peptides formation: Focused on host defense peptides. Biomed Pharmacother 2023; 162:114586. [PMID: 36989711 DOI: 10.1016/j.biopha.2023.114586] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The byproducts of bacterial fermentation known as short-chain fatty acids (SCFAs) are chemically comprised of a carboxylic acid component and a short hydrocarbon chain. Recent investigations have demonstrated that SCFAs can affect intestinal immunity by inducing endogenous host defense peptides (HDPs) and their beneficial effects on barrier integrity, gut health, energy supply, and inflammation. HDPs, which include defensins, cathelicidins, and C-type lectins, perform a significant function in innate immunity in gastrointestinal mucosal membranes. SCFAs have been demonstrated to stimulate HDP synthesis by intestinal epithelial cells via interactions with G protein-coupled receptor 43 (GPR43), activation of the Jun N-terminal kinase (JNK) and Mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways, and the cell growth pathways. Furthermore, SCFA butyrate has been demonstrated to enhance the number of HDPs released from macrophages. SCFAs promote monocyte-to-macrophage development and stimulate HDP synthesis in macrophages by inhibiting histone deacetylase (HDAC). Understanding the etiology of many common disorders might be facilitated by studies into the function of microbial metabolites, such as SCFAs, in the molecular regulatory processes of immune responses (e.g., HDP production). This review will focus on the current knowledge of the role and mechanism of microbiota-derived SCFAs in influencing the synthesis of host-derived peptides, particularly HDPs.
Collapse
|
10
|
Mazidi M, Katsiki N, Banach M. Higher Plasma Levels of Valerate Produced by Gut Microbiota May Have a Beneficial Impact on Renal Function. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2023:1-7. [PMID: 36786830 DOI: 10.1080/07315724.2019.1664955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
OBJECTIVE Observational studies have evaluated the relationships among plasma short chain fatty acids (SCFA) produced by gut microbiota, renal function, and risk of chronic kidney disease (CKD). In the present study, Mendelian Randomization (MR) analysis was applied to obtain unconfounded estimates of the casual association of genetically determined plasma valerate (an SCFA) with kidney function and risk of CKD. METHOD MR was performed by using summary-level data from the largest genome-wide association studies (GWAS) conducted on plasma valerate, CKD, and estimated glomerular filtration rate (eGFR; separately in diabetic and nondiabetic individuals). Inverse variance weighted method (IVW), weighted median-based method, MR-Egger, as well as MR-Pleiotropy RESidual Sum and Outlier (PRESSO) were applied. Sensitivity analysis was conducted using the leave-one-out method. RESULTS No significant association was observed between plasma valerate and CKD (IVW: β = 0.234, p = 0.744). In contrast, plasma valerate was positively associated with eGFR in the total population (IVW: β = 0.049, p = 0.022) and among nondiabetic individuals (IVW: β = 0.058, p = 0.009), but not in the diabetic population (IVW: β = -0.052, p = 0.603). None of the estimated associations was subjected to significant level of heterogeneity. Furthermore, MR-PRESSO analysis did not show any chance of outlier for all estimates. The pleiotropy test, with very a negligible intercept and insignificant p value, also indicated no chance of pleiotropy for all of our estimations (all p > 0.539). The results of the MR-Robust Adjusted Profile Score were identical with the IVW estimates, highlighting again no possibility of pleiotropy. Results of the leave-one-out method demonstrated that the links were not driven by single-nucleotide polymorphisms. CONCLUSIONS Individuals with higher plasma valerate levels had better renal function, defined by eGFR. This finding was observed in the total population and in nondiabetic subjects, but not in those with diabetes. Further research is needed to elucidate the links among plasma valerate, kidney function, and CKD.
Collapse
Affiliation(s)
- Mohsen Mazidi
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Niki Katsiki
- First Department of Internal Medicine, Center for Diabetes, Metabolism and Endocrinology, AHEPA University Hospital, Thessaloniki, Greece
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Poland.,Polish Mother's Memorial Hospital Research Institute, Lodz, Poland.,Cardiovascular Research Centre, University of Zielona Gora, Poland
| |
Collapse
|
11
|
Yoon JH, Hwang J, Son SU, Choi J, You SW, Park H, Cha SY, Maeng S. How Can Insulin Resistance Cause Alzheimer's Disease? Int J Mol Sci 2023; 24:3506. [PMID: 36834911 PMCID: PMC9966425 DOI: 10.3390/ijms24043506] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with cognitive decline. Despite worldwide efforts to find a cure, no proper treatment has been developed yet, and the only effective countermeasure is to prevent the disease progression by early diagnosis. The reason why new drug candidates fail to show therapeutic effects in clinical studies may be due to misunderstanding the cause of AD. Regarding the cause of AD, the most widely known is the amyloid cascade hypothesis, in which the deposition of amyloid beta and hyperphosphorylated tau is the cause. However, many new hypotheses were suggested. Among them, based on preclinical and clinical evidence supporting a connection between AD and diabetes, insulin resistance has been pointed out as an important factor in the development of AD. Therefore, by reviewing the pathophysiological background of brain metabolic insufficiency and insulin insufficiency leading to AD pathology, we will discuss how can insulin resistance cause AD.
Collapse
Affiliation(s)
- Ji Hye Yoon
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - JooHyun Hwang
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Sung Un Son
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Junhyuk Choi
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Seung-Won You
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Hyunwoo Park
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
- Health Park Co., Ltd., Seoul 02447, Republic of Korea
| | - Seung-Yun Cha
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Sungho Maeng
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| |
Collapse
|
12
|
Immobilization of Lactobacillus plantarum NCIMB 8826 ameliorates Citrobacter rodentium induced lesions and enhances the gut inflammatory response in C57BL/6 weanling mice. FOOD PRODUCTION, PROCESSING AND NUTRITION 2022. [DOI: 10.1186/s43014-022-00111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
AbstractInfectious diarrhea is a major cause of infant mortality in most developing countries. In this research, we evaluated the potential of immobilized Lactobacillus plantarum NCIMB 8826 on weanimix infant cereal and its effectiveness in reducing the severity of Citrobacter rodentium-induced diarrhea in weanling mice. Thirty-six C57BL/6 weanling mice were placed into four groups (n = 9 each; negative, positive, prevention and cure). Mice received either L. plantarum (109 CFU/g) immobilized on weanimix infant cereal 3 days before C. rodentium (109 CFU/ ml) infection (Prevention) or 3 days after C. rodentium infection (Cure). A positive control group was infected with C. rodentium only, while a negative control group received neither L. plantarum nor C. rodentium. Positive control mice showed colonic mucosal and submucosal inflammation, erosion, and mucosal epithelia hyperplasia with the C. rodentium infection. Mice in the prevention and cure groups had less severe histologic alterations in the colon. Some beneficial effect of L. plantarum was observed in cecal short-chain fatty acid concentrations, which stimulates water and electrolytes absorption to reduce diarrhea. Our findings demonstrated that L. plantarum NCIMB 8826 could be immobilized on weanimix infant cereal to help reduce diarrhea during weaning.
Graphical Abstract
Collapse
|
13
|
Lin CY, Jha AR, Oba PM, Yotis SM, Shmalberg J, Honaker RW, Swanson KS. Longitudinal fecal microbiome and metabolite data demonstrate rapid shifts and subsequent stabilization after an abrupt dietary change in healthy adult dogs. Anim Microbiome 2022; 4:46. [PMID: 35915514 PMCID: PMC9341101 DOI: 10.1186/s42523-022-00194-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Diet has a large influence on gut microbiota diversity and function. Although previous studies have investigated the effect of dietary interventions on the gut microbiome, longitudinal changes in the gut microbiome, microbial functions, and metabolite profiles post dietary interventions have been underexplored. How long these outcomes require to reach a steady-state, how they relate to one another, and their impact on host physiological changes are largely unknown. To address these unknowns, we collected longitudinal fecal samples following an abrupt dietary change in healthy adult beagles (n = 12, age: 5.16 ± 0.87 year, BW: 13.37 ± 0.68 kg) using a crossover design. All dogs were fed a kibble diet (control) from d1-14, and then fed that same diet supplemented with fiber (HFD) or a protein-rich canned diet (CD) from d15-27. Fresh fecal samples were collected on d13, 16, 20, 24, and 27 for metabolite and microbiome assessment. Fecal microbial diversity and composition, metabolite profiles, and microbial functions dramatically diverged and stabilized within a few days (2 d for metabolites; 6 d for microbiota) after dietary interventions. Fecal acetate, propionate, and total short-chain fatty acids increased after change to HFD, while fecal isobutyrate, isovalerate, total branched-chain fatty acids, phenol, and indole increased after dogs consumed CD. Relative abundance of ~ 100 bacterial species mainly belonging to the Firmicutes, Proteobacteria, and Actinobacteria phyla increased in HFD. These shifts in gut microbiome diversity and composition were accompanied by functional changes. Transition to HFD led to increases in the relative abundance of KEGG orthology (KO) terms related to starch and sucrose metabolism, fatty acid biosynthesis, and amino sugar and nucleotide sugar metabolism, while transition to CD resulted in increased relative abundance of KO terms pertaining to inositol phosphate metabolism and sulfur metabolism. Significant associations among fecal microbial taxa, KO terms, and metabolites were observed, allowing for high-accuracy prediction of diet group by random forest analysis.
Conclusions
Longitudinal sampling and a multi-modal approach to characterizing the gastrointestinal environment allowed us to demonstrate how drastically and quickly dietary changes impact the fecal microbiome and metabolite profiles of dogs following an abrupt dietary change and identify key microbe-metabolite relationships that allowed for treatment prediction.
Collapse
|
14
|
Xu J, Moore BN, Pluznick JL. Short-Chain Fatty Acid Receptors and Blood Pressure Regulation: Council on Hypertension Mid-Career Award for Research Excellence 2021. Hypertension 2022; 79:2127-2137. [PMID: 35912645 PMCID: PMC9458621 DOI: 10.1161/hypertensionaha.122.18558] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gut microbiome influences host physiology and pathophysiology through several pathways, one of which is microbial production of chemical metabolites which interact with host signaling pathways. Short-chain fatty acids (SCFAs) are a class of gut microbial metabolites known to activate multiple signaling pathways in the host. Growing evidence indicates that the gut microbiome is linked to blood pressure, that SCFAs modulate blood pressure regulation, and that delivery of exogenous SCFAs lowers blood pressure. Given that hypertension is a key risk factor for cardiovascular disease, the examination of novel contributors to blood pressure regulation has the potential to lead to novel approaches or treatments. Thus, this review will discuss SCFAs with a focus on their host G protein-coupled receptors including GPR41 (G protein-coupled receptor 41), GPR43, and GPR109A, as well as OLFR78 (olfactory receptor 78) and OLFR558. This includes a discussion of the ligand profiles, G protein coupling, and tissue distribution of each receptor. We will also review phenotypes relevant to blood pressure regulation which have been reported to date for Gpr41, Gpr43, Gpr109a, and Olfr78 knockout mice. In addition, we will consider how SCFA signaling influences physiology at baseline, and, how SCFA signaling may contribute to blood pressure regulation in settings of hypertension. In sum, this review will integrate current knowledge regarding how SCFAs and their receptors regulate blood pressure.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Brittni N. Moore
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jennifer L. Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
15
|
Barriga SA, Peñ SI, Caballero SC, Gracia MI, del Villar JL, Escobar-Ch JJ, Rivera M, Macias L, Garcia H, Vargas-Est D. Effect of Tributyrin Supplementation on Glucose Levels, Liver and Kidney Integrity in an Experimental Model of Diabetes Mellitus. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.1387.1399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
16
|
Abdalkareem Jasim S, Jade Catalan Opulencia M, Alexis Ramírez-Coronel A, Kamal Abdelbasset W, Hasan Abed M, Markov A, Raheem Lateef Al-Awsi G, Azamatovich Shamsiev J, Thaeer Hammid A, Nader Shalaby M, Karampoor S, Mirzaei R. The emerging role of microbiota-derived short-chain fatty acids in immunometabolism. Int Immunopharmacol 2022; 110:108983. [PMID: 35750016 DOI: 10.1016/j.intimp.2022.108983] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 02/07/2023]
Abstract
The accumulating evidence revealed that microbiota plays a significant function in training, function, and the induction of host immunity. Once this interaction (immune system-microbiota) works correctly, it enables the production of protective responses against pathogens and keeps the regulatory pathways essential for maintaining tolerance to innocent antigens. This concept of immunity and metabolic activity redefines the realm of immunometabolism, paving the way for innovative therapeutic interventions to modulate immune cells through immune metabolic alterations. A body of evidence suggests that microbiota-derived metabolites, including short-chain fatty acids (SCFAs) such as butyrate, acetate, and propionate, play a key role in immune balance. SCFAs act on many cell types to regulate various vital biological processes, including host metabolism, intestinal function, and the immune system. Such SCFAs generated by gut bacteria also impact immunity, cellular function, and immune cell fate. This is a new concept of immune metabolism, and better knowledge about how lifestyle affects intestinal immunometabolism is crucial for preventing and treating disease. In this review article, we explicitly focus on the function of SCFAs in the metabolism of immune cells, especially macrophages, neutrophils, dendritic cells (DCs), B cells, T (Th) helper cells, and cytotoxic T cells (CTLs).
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-anbar-Ramadi, Iraq.
| | | | - Andrés Alexis Ramírez-Coronel
- Laboratory of Psychometrics, Comparative Psychology and Ethology (LABPPCE), Universidad Católica de Cuenca, Ecuador and Universidad CES, Medellín, Colombia, Cuenca, Ecuador.
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia; Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt.
| | - Murtadha Hasan Abed
- Department of Medical Laboratory, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq.
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation; Tyumen Industrial University, Tyumen, Russian Federation.
| | | | - Jamshid Azamatovich Shamsiev
- Department of Pediatric Surgery, Anesthesiology and Intensive Care, Samarkand State Medical Institute, Samarkand, Uzbekistan; Research scholar, Department of Scientific Affairs, Tashkent State Dental Institute, Makhtumkuli Street 103, Tashkent, 100047, Uzbekistan.
| | - Ali Thaeer Hammid
- Computer Engineering Techniques Department, Faculty of Information Technology, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq.
| | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Egypt.
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
17
|
Spivak I, Fluhr L, Elinav E. Local and systemic effects of microbiome‐derived metabolites. EMBO Rep 2022; 23:e55664. [PMID: 36031866 PMCID: PMC9535759 DOI: 10.15252/embr.202255664] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 12/12/2022] Open
Abstract
Commensal microbes form distinct ecosystems within their mammalian hosts, collectively termed microbiomes. These indigenous microbial communities broadly expand the genomic and functional repertoire of their host and contribute to the formation of a “meta‐organism.” Importantly, microbiomes exert numerous biochemical reactions synthesizing or modifying multiple bioactive small molecules termed metabolites, which impact their host's physiology in a variety of contexts. Identifying and understanding molecular mechanisms of metabolite–host interactions, and how their disrupted signaling can contribute to diseases, may enable their therapeutic application, a modality termed “postbiotic” therapy. In this review, we highlight key examples of effects of bioactive microbe‐associated metabolites on local, systemic, and immune environments, and discuss how these may impact mammalian physiology and associated disorders. We outline the challenges and perspectives in understanding the potential activity and function of this plethora of microbially associated small molecules as well as possibilities to harness them toward the promotion of personalized precision therapeutic interventions.
Collapse
Affiliation(s)
- Igor Spivak
- Systems Immunology Department Weizmann Institute of Science Rehovot Israel
- Medical Clinic III University Hospital Aachen Aachen Germany
| | - Leviel Fluhr
- Systems Immunology Department Weizmann Institute of Science Rehovot Israel
| | - Eran Elinav
- Systems Immunology Department Weizmann Institute of Science Rehovot Israel
- Microbiome & Cancer Division, DKFZ Heidelberg Germany
| |
Collapse
|
18
|
Gao Y, Ma L, Su J. Host and microbial-derived metabolites for Clostridioides difficile infection: Contributions, mechanisms and potential applications. Microbiol Res 2022; 263:127113. [PMID: 35841835 DOI: 10.1016/j.micres.2022.127113] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 12/23/2022]
Abstract
Clostridioides difficile infection (CDI), which mostly occurs in hospitalized patients, is the most common and costly health care-associated disease. However, the biology of C. difficile remains incompletely understood. Current therapeutics are still challenged by the frequent recurrence of CDI. Advances in metabolomics facilitate our understanding of the etiology of CDI, which is not merely an alteration in the structure of the gut microbial community but also a dysbiosis metabolic setting promoting the germination, expansion and virulence of C. difficile. Therefore, we summarized the gut microbial and metabolic profiles for CDI under different conditions, such as those of postantibiotic treatment and postfecal microbiota transplantation. The current understanding of the role of host and gut microbial-derived metabolites as well as other nutrients in preventing or alleviating the disease symptoms of CDI will also be provided in this review. We hope that a specific nutrient-centric dietary strategy or the administration of certain nutrients to the colon could serve as an alternate line of investigation for the prophylaxis and mitigation of CDI in the future. Nevertheless, rigorously designed basic studies and randomized controlled trials need to be conducted to assess the functional mechanisms and effects of such therapeutics.
Collapse
Affiliation(s)
- Yan Gao
- Department of Clinical Laboratory Diagnostics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Liyan Ma
- Department of Clinical Laboratory Diagnostics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianrong Su
- Department of Clinical Laboratory Diagnostics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
19
|
Watanabe DHM, Doelman J, Steele MA, Guan LL, Seymour DJ, Metcalf JA, Penner GB. Effect of Feeding Calcium Gluconate Embedded in a Hydrogenated Fat Matrix on Feed Intake, Gastrointestinal Fermentation and Morphology, Intestinal Brush Boarder Enzyme Activity and Blood Metabolites in Growing Lambs. J Anim Sci 2022; 100:6598089. [PMID: 35652468 DOI: 10.1093/jas/skac205] [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: 02/18/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Gluconate salts have been identified as a butyrate precursor when fed to non-ruminant species and may increase the butyrate concentration in the large intestine supporting gastrointestinal health and development. The objective of this study was to evaluate the dose response of hydrogenated fat-embedded calcium gluconate (HFCG) on performance and gastrointestinal tract (GIT) development in growing lambs. Thirty-two wether lambs were used in a randomized complete block design and assigned to 1 of 4 treatments differing in the inclusion of HFCG: 0.0% (CON), 0.075% (LOW), 0.30% (MED), and 0.60% of the diet (HIGH). Lambs were allocated into individual pens and fed ad libitum with feed delivered twice daily. Feed intake was recorded daily, and body weight (BW) was assessed at the beginning and the end of the 29-d period. Blood was sampled on d 21, prior to feeding and 6 h post-feeding to evaluate changes in β-hydroxybutyrate, glucose, and insulin concentrations. Total fecal collection was conducted during d 25 to 28 to assess apparent total tract digestibility. On d 29, lambs were slaughtered, and the entire GIT was separated by region to enable sampling of tissue and digesta. Data were analyzed to assess linear, quadratic, and cubic effects of HFCG dose. Final BW, average daily gain, and dry matter intake decreased linearly (P ≤ 0.02) with increasing HFCG. Increasing inclusion of HFCG linearly decreased (P = 0.01) the thickness of the stratum corneum in ruminal papillae but did not affect other strata (P ≥ 0.34). Omasal digesta weight linearly decreased (P = 0.01) as the concentration of HFCG increased and abomasal digesta weight was cubically affected (P = 0.03) the increasing dose of HFCG. Short-chain fatty acid concentration in the cecum was cubically affected (P < 0.01) with increasing dose of HFCG where low dose had the greatest concentration. Moreover, increasing the dietary supply of HFCG linearly increased the proportion of acetate (P = 0.04) in the cecum and linearly decreased the proportion of propionate in the digesta of both the cecum (P < 0.01) and colon (P = 0.01). Colon crypt depth was quadratically (P = 0.03) affected with the increasing dose of HFCG, where lambs fed MED had greatest crypt depth. We conclude that feeding HFCG to growing lambs did not increase butyrate concentration in the large intestine and consequently does not increase the absorptive surface area of the whole tract, the size of the GIT, or the functionality of the intestine.
Collapse
Affiliation(s)
- Daniel H M Watanabe
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John Doelman
- Trouw Nutrition R&D, PO Box 299, 3800 AG, Amersfoort, the Netherlands
| | - Michael A Steele
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Le L Guan
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Dave J Seymour
- Trouw Nutrition R&D, PO Box 299, 3800 AG, Amersfoort, the Netherlands
| | - John A Metcalf
- Trouw Nutrition North America, 7504 McLean Rd E., Puslinch, ON
| | - Gregory B Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
20
|
The microbiota-gut-kidney axis mediates host osmoregulation in a small desert mammal. NPJ Biofilms Microbiomes 2022; 8:16. [PMID: 35379849 PMCID: PMC8980004 DOI: 10.1038/s41522-022-00280-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/21/2022] [Indexed: 12/17/2022] Open
Abstract
Regulating sodium and water balances is crucial for survival of small, desert mammals. Studies demonstrate that the gut microbiota and their metabolites are involved in host energy homeostasis, but little is known on the interactions among salt loading, gut microbiota, and osmoregulation. The aim of this study was to fill this gap. Mongolian gerbils (Meriones unguiculatus) were offered drinking water (Con) and either water containing moderate (4%, MS) or high NaCl (8%, HS) ad libitum. Intake of HS reduced α diversity of the microbial community and, at the genus level, reduced the relative abundances of Rikenella and Christensenella but increased Atopobium. To confirm the function of gut microbiota in host osmoregulation, we transplanted caecal microbiota in HS gerbils. To cope with salt loading, the gerbils concentrated urine, resulting in negative energy balance and systemic inflammation. The HS gerbils increased hypothalamic arginine vasopressin and intestinal and renal aquaporin 2 to support water retention, and reduced intestinal and renal epithelial sodium channel α to promote sodium excretion. However, HS gerbils with caecal microbiota transplant (CMT) from Con donors maintained energy balance and osmoregulation, and had a much reduced systemic inflammation. Further, CMT from Con donors to HS recipients reshaped the gut microbiota, particularly by reducing Parabacteroides distasonis and Prevotella copri, and increasing Lactobacillus reuteri abundances, with a resulting increase in bacterial metabolites such as butyrate. These findings highlight a vital role of the microbiota-gut-kidney axis in mediating salt-related osmoregulation, allowing small mammals to adapt to high salt loads in a desert habitat.
Collapse
|
21
|
Waclawiková B, Codutti A, Alim K, El Aidy S. Gut microbiota-motility interregulation: insights from in vivo, ex vivo and in silico studies. Gut Microbes 2022; 14:1997296. [PMID: 34978524 PMCID: PMC8741295 DOI: 10.1080/19490976.2021.1997296] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/30/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
The human gastrointestinal tract is home to trillions of microbes. Gut microbial communities have a significant regulatory role in the intestinal physiology, such as gut motility. Microbial effect on gut motility is often evoked by bioactive molecules from various sources, including microbial break down of carbohydrates, fibers or proteins. In turn, gut motility regulates the colonization within the microbial ecosystem. However, the underlying mechanisms of such regulation remain obscure. Deciphering the inter-regulatory mechanisms of the microbiota and bowel function is crucial for the prevention and treatment of gut dysmotility, a comorbidity associated with many diseases. In this review, we present an overview of the current knowledge on the impact of gut microbiota and its products on bowel motility. We discuss the currently available techniques employed to assess the changes in the intestinal motility. Further, we highlight the open challenges, and incorporate biophysical elements of microbes-motility interplay, in an attempt to lay the foundation for describing long-term impacts of microbial metabolite-induced changes in gut motility.
Collapse
Affiliation(s)
- Barbora Waclawiková
- Host-Microbe Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| | - Agnese Codutti
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Karen Alim
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Physics Department and Center for Protein Assemblies (CPA), Technische Universität München, Garching, Germany
| | - Sahar El Aidy
- Host-Microbe Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Theurer ME, Fox JT, McCarty TM, McCollum RM, Jones TM, Simpson J, Martin T. Evaluation of the reticulorumen pH throughout the feeding period for beef feedlot steers maintained in a commercial feedlot and its association with liver abscesses. J Am Vet Med Assoc 2021; 259:899-908. [PMID: 34609179 DOI: 10.2460/javma.259.8.899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the reticulorumen pH of beef feedlot steers throughout the feeding period and to assess the association between the respective durations that the reticulorumen pH was ≤ 5.6 (subacute ruminal acidosis) and ≤ 5.2 (acute ruminal acidosis) and liver abscess severity. ANIMALS 59 feedlot steers (mean body weight, 349.5 kg). PROCEDURES On day 0, each steer was orally administered an electronic bolus that monitored the reticulorumen pH every 10 minutes for 150 days. Steers were transitioned from a starter to intermediate ration on day 8 (transition 1) and from the intermediate to finish ration on day 19 (transition 2). The ration carbohydrate and megacalorie contents increased with each transition. During each transition, the lower megacalorie ration was fed at the 8:00 am feeding and the higher megacalorie ration was fed at the 2:00 pm feeding for 3 days before the higher megacalorie ration was fed extensively. Steers were sent to slaughter after 182 days; each carcass was assessed for liver abscesses. RESULTS The diurnal reticulorumen pH pattern was characterized by a peak at 7:00 am and nadir at 8:00 pm. The mean percentages of time that the reticulorumen pH was ≤ 5.6 and ≤ 5.2 were more than 10-fold greater during transition 1, compared with during transition 2, and were significantly greater for steers with extensive liver abscesses than for steers without extensive liver abscesses. CONCLUSIONS AND CLINICAL RELEVANCE Efforts to minimize the duration that the reticulorumen pH is ≤ 5.6 might mitigate liver abscess formation in feedlot cattle.
Collapse
|
23
|
Guzmán JL, Martín-García I, Pérez-Écija A, García-Brenes MD, Zarazaga LÁ, Delgado-Pertíñez M. Supplementing the Diet of Dairy Goats with Dried Orange Pulp throughout Lactation: I. Effect on Milk Performance, Nutrient Utilisation, Blood Parameters and Production Economics. Animals (Basel) 2021; 11:ani11092601. [PMID: 34573567 PMCID: PMC8467038 DOI: 10.3390/ani11092601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Dried orange pulp can conveniently replace cereals in ruminant diets; however, no published reports have considered a similar substitution for the goat diet throughout lactation. Therefore, in this study, we evaluated the effects of cereal replacement (40% and 80%) by dried orange pulp in the diet of Payoya breed goats during the entire lactation period (180 days) on milk yield and composition, blood metabolites and production economics. Also evaluated, in mid-lactation, were the nutrient utilisation and ruminal fermentation of the dried orange pulp diets. Milk production and gross composition did not change in goats fed diets that included dried orange pulp. A decrease in nitrogen availability and retention was observed, while an increase detected in the digestibility of cellulose likely boosted rumen microbial protein synthesis. No pathological effects of dried orange pulp supplementation were detected in blood metabolites. The partial substitution of cereals for dried orange pulp reduced costs and raised economic profits. In conclusion, the partial replacement of cereals by dried orange pulp is a profitable and healthy nutritional strategy in dairy goats that does not compromise their productivity and is suitable for the entire lactation period. Abstract Dried orange pulp (DOP) can be incorporated into ruminant diets, but no reports have considered this strategy during the entire lactation period in goats. Two experiments were performed using lactating Payoya goats. In experiment 1, to study the effect, over 180 days, of DOP on milk yield and composition, blood metabolites and economic values, 44 primiparous goats were allocated into three groups: control diet (concentrate plus lucerne) and DOP40 and DOP80 diets, in which DOP replaced 40% and 80%, respectively, of the cereals. Nutrient digestibility and rumen fermentation were also studied (experiment 2). The DOP diets did not affect milk yield and composition. DOP triggered lower intake and digestibility of ether extract and crude protein. Ruminal fermentation was unaffected by DOP, except for a decrease in butyrate for DOP80. The energy balance was unaltered by diet while the balance and retention of nitrogen decreased. Regarding plasma biochemistry, DOP supplementation caused changes that could indicate an improvement in hepatic function and reduced muscular damage and oxidative muscular stress. Moreover, DOP80 provided a profit increase of EUR 3.27/goat. In conclusion, the partial replacement of cereals by DOP is a profitable and healthy nutritional strategy in dairy goats and is suitable for the entire lactation period without compromising productivity.
Collapse
Affiliation(s)
- José Luis Guzmán
- Departamento de Ciencias Agroforestales, Escuela Técnica Superior de Ingeniería, Universidad de Huelva, “Campus de Excelencia Internacional Agroalimentario, ceiA3”, Campus Universitario de la Rábida, Carretera Huelva-Palos de la Frontera, s/n., 21819 Palos de la Frontera (Huelva), Spain; (J.L.G.); (L.Á.Z.)
| | | | - Alejandro Pérez-Écija
- Departamento de Medicina y Cirugía Animal, Campus Rabanales, Universidad de Córdoba, 14104 Córdoba, Spain;
| | - Manuel David García-Brenes
- Departamento de Economía Aplicada II. Escuela Técnica Superior de Ingeniería Agronómica, Universidad de Sevilla, Carretera de Utrera Km 1, 41013 Sevilla, Spain;
| | - Luis Ángel Zarazaga
- Departamento de Ciencias Agroforestales, Escuela Técnica Superior de Ingeniería, Universidad de Huelva, “Campus de Excelencia Internacional Agroalimentario, ceiA3”, Campus Universitario de la Rábida, Carretera Huelva-Palos de la Frontera, s/n., 21819 Palos de la Frontera (Huelva), Spain; (J.L.G.); (L.Á.Z.)
| | - Manuel Delgado-Pertíñez
- Departamento de Agronomía, Escuela Técnica Superior de Ingeniería Agronómica, Universidad de Sevilla, Ctra. Utrera Km 1, 41013 Sevilla, Spain
- Correspondence: ; Tel.: +34-954486449
| |
Collapse
|
24
|
Gurry T, Nguyen LTT, Yu X, Alm EJ. Functional heterogeneity in the fermentation capabilities of the healthy human gut microbiota. PLoS One 2021; 16:e0254004. [PMID: 34288919 PMCID: PMC8294568 DOI: 10.1371/journal.pone.0254004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
The human gut microbiota is known for its highly heterogeneous composition across different individuals. However, relatively little is known about functional differences in its ability to ferment complex polysaccharides. Through ex vivo measurements from healthy human donors, we show that individuals vary markedly in their microbial metabolic phenotypes (MMPs), mirroring differences in their microbiota composition, and resulting in the production of different quantities and proportions of Short Chain Fatty Acids (SCFAs) from the same inputs. We also show that aspects of these MMPs can be predicted from composition using 16S rRNA sequencing. From experiments performed using the same dietary fibers in vivo, we demonstrate that an ingested bolus of fiber is almost entirely consumed by the microbiota upon passage. We leverage our ex vivo data to construct a model of SCFA production and absorption in vivo, and argue that inter-individual differences in quantities of absorbed SCFA are directly related to differences in production. Though in vivo studies are required to confirm these data in the context of the gut, in addition to in vivo read outs of SCFAs produced in response to specific fiber spike-ins, these data suggest that optimizing SCFA production in a given individual through targeted fiber supplementation requires quantitative understanding of their MMP.
Collapse
Affiliation(s)
- Thomas Gurry
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- The Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Institute for Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Le Thanh Tu Nguyen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- The Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Xiaoqian Yu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Eric J. Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- The Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Tomasova L, Grman M, Ondrias K, Ufnal M. The impact of gut microbiota metabolites on cellular bioenergetics and cardiometabolic health. Nutr Metab (Lond) 2021; 18:72. [PMID: 34266472 PMCID: PMC8281717 DOI: 10.1186/s12986-021-00598-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
Recent research demonstrates a reciprocal relationship between gut microbiota-derived metabolites and the host in controlling the energy homeostasis in mammals. On the one hand, to thrive, gut bacteria exploit nutrients digested by the host. On the other hand, the host utilizes numerous products of gut bacteria metabolism as a substrate for ATP production in the colon. Finally, bacterial metabolites seep from the gut into the bloodstream and interfere with the host’s cellular bioenergetics machinery. Notably, there is an association between alterations in microbiota composition and the development of metabolic diseases and their cardiovascular complications. Some metabolites, like short-chain fatty acids and trimethylamine, are considered markers of cardiometabolic health. Others, like hydrogen sulfide and nitrite, demonstrate antihypertensive properties. Scientific databases were searched for pre-clinical and clinical studies to summarize current knowledge on the role of gut microbiota metabolites in the regulation of mammalian bioenergetics and discuss their potential involvement in the development of cardiometabolic disorders. Overall, the available data demonstrates that gut bacteria products affect physiological and pathological processes controlling energy and vascular homeostasis. Thus, the modulation of microbiota-derived metabolites may represent a new approach for treating obesity, hypertension and type 2 diabetes.
Collapse
Affiliation(s)
- Lenka Tomasova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05, Bratislava, Slovak Republic.
| | - Marian Grman
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Karol Ondrias
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-091, Warsaw, Poland.
| |
Collapse
|
26
|
Shepard BD. The Sniffing Kidney: Roles for Renal Olfactory Receptors in Health and Disease. KIDNEY360 2021; 2:1056-1062. [PMID: 35373087 PMCID: PMC8791376 DOI: 10.34067/kid.0000712021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
AbstractOlfactory receptors (ORs) represent the largest gene family in the human genome. Despite their name, functions exist for these receptors outside of the nose. Among the tissues known to take advantage of OR signaling is the kidney. From mouse to man, the list of renal ORs continues to expand, and they have now been linked to a variety of processes involved in the maintenance of renal homeostasis, including the modulation of blood pressure, response to acidemia, and the development of diabetes. In this review, we highlight the recent progress made on the growing appreciation for renal ORs in physiology and pathophysiology.
Collapse
|
27
|
Poll BG, Cheema MU, Pluznick JL. Gut Microbial Metabolites and Blood Pressure Regulation: Focus on SCFAs and TMAO. Physiology (Bethesda) 2021; 35:275-284. [PMID: 32490748 DOI: 10.1152/physiol.00004.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Shifts in the gut microbiome play a key role in blood pressure regulation, and changes in the production of gut microbial metabolites are likely to be a key mechanism. Known gut microbial metabolites include short-chain fatty acids, which can signal via G-protein-coupled receptors, and trimethylamine-N oxide. In this review, we provide an overview of gut microbial metabolites documented thus far to play a role in blood pressure regulation.
Collapse
Affiliation(s)
- Brian G Poll
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Muhammad Umar Cheema
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
28
|
Mirzaei R, Bouzari B, Hosseini-Fard SR, Mazaheri M, Ahmadyousefi Y, Abdi M, Jalalifar S, Karimitabar Z, Teimoori A, Keyvani H, Zamani F, Yousefimashouf R, Karampoor S. Role of microbiota-derived short-chain fatty acids in nervous system disorders. Biomed Pharmacother 2021; 139:111661. [PMID: 34243604 DOI: 10.1016/j.biopha.2021.111661] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
During the past decade, accumulating evidence from the research highlights the suggested effects of bacterial communities of the human gut microbiota and their metabolites on health and disease. In this regard, microbiota-derived metabolites and their receptors, beyond the immune system, maintain metabolism homeostasis, which is essential to maintain the host's health by balancing the utilization and intake of nutrients. It has been shown that gut bacterial dysbiosis can cause pathology and altered bacterial metabolites' formation, resulting in dysregulation of the immune system and metabolism. The short-chain fatty acids (SCFAs), such as butyrate, acetate, and succinate, are produced due to the fermentation process of bacteria in the gut. It has been noted remodeling in the gut microbiota metabolites associated with the pathophysiology of several neurological disorders, such as Alzheimer's disease, multiple sclerosis, Parkinson's disease, amyotrophic lateral sclerosis, stress, anxiety, depression, autism, vascular dementia, schizophrenia, stroke, and neuromyelitis optica spectrum disorders, among others. This review will discuss the current evidence from the most significant studies dealing with some SCFAs from gut microbial metabolism with selected neurological disorders.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Behnaz Bouzari
- Department of Pathology, Firouzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mazaheri
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Milad Abdi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Karimitabar
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Teimoori
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Keyvani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Huart J, Cirillo A, Taminiau B, Descy J, Saint-Remy A, Daube G, Krzesinski JM, Melin P, de Tullio P, Jouret F. Human Stool Metabolome Differs upon 24 h Blood Pressure Levels and Blood Pressure Dipping Status: A Prospective Longitudinal Study. Metabolites 2021; 11:metabo11050282. [PMID: 33946722 PMCID: PMC8146767 DOI: 10.3390/metabo11050282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Dysbiosis of gut microbiota (GM) has been involved in the pathophysiology of arterial hypertension (HT), via a putative role of short chain fatty acids (SCFAs). Its role in the circadian regulation of blood pressure (BP), also called “the dipping profile”, has been poorly investigated. Sixteen male volunteers and 10 female partners were subjected to 24 h ambulatory BP monitoring and were categorized in normotensive (NT) versus HT, as well as in dippers versus non-dippers. Nuclear magnetic resonance (NMR)-based metabolomics was performed on stool samples. A 5-year comparative follow-up of BP profiles and stool metabolomes was done in men. Significant correlations between stool metabolome and 24 h mean BP levels were found in both male and female cohorts and in the entire cohort (R2 = 0.72, R2 = 0.79, and R2 = 0.45, respectively). Multivariate analysis discriminated dippers versus non-dippers in both male and female cohorts and in the entire cohort (Q2 = 0.87, Q2 = 0.98, and Q2 = 0.68, respectively). Fecal amounts of acetate, propionate, and butyrate were higher in HT versus NT patients (p = 0.027; p = 0.015 and p = 0.015, respectively), as well as in non-dippers versus dippers (p = 0.027, p = 0.038, and p = 0.036, respectively) in the entire cohort. SCFA levels were significantly different in patients changing of dipping status over the 5-year follow-up. In conclusion, stool metabolome changes upon global and circadian BP profiles in both genders.
Collapse
Affiliation(s)
- Justine Huart
- Division of Nephrology, University of Liège Hospital (ULg CHU), University of Liège, B-4000 Liège, Belgium; (A.S.-R.); (J.-M.K.); (F.J.)
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, B-4000 Liège, Belgium
- Correspondence:
| | - Arianna Cirillo
- Center for Interdisciplinary Research on Medicines (CIRM), Metabolomics Group, University of Liège, B-4000 Liège, Belgium; (A.C.); (P.d.T.)
| | - Bernard Taminiau
- Fundamental and Applied Research Center for Animal & Health (FARAH), Veterinary Public Health, University of Liège, B-4000 Liège, Belgium; (B.T.); (G.D.)
- Laboratory for Food Microbiology, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | - Julie Descy
- Clinical Microbiology, University of Liège Hospital (ULg CHU), University of Liège, B-4000 Liège, Belgium; (J.D.); (P.M.)
| | - Annie Saint-Remy
- Division of Nephrology, University of Liège Hospital (ULg CHU), University of Liège, B-4000 Liège, Belgium; (A.S.-R.); (J.-M.K.); (F.J.)
| | - Georges Daube
- Fundamental and Applied Research Center for Animal & Health (FARAH), Veterinary Public Health, University of Liège, B-4000 Liège, Belgium; (B.T.); (G.D.)
- Laboratory for Food Microbiology, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | - Jean-Marie Krzesinski
- Division of Nephrology, University of Liège Hospital (ULg CHU), University of Liège, B-4000 Liège, Belgium; (A.S.-R.); (J.-M.K.); (F.J.)
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, B-4000 Liège, Belgium
| | - Pierrette Melin
- Clinical Microbiology, University of Liège Hospital (ULg CHU), University of Liège, B-4000 Liège, Belgium; (J.D.); (P.M.)
| | - Pascal de Tullio
- Center for Interdisciplinary Research on Medicines (CIRM), Metabolomics Group, University of Liège, B-4000 Liège, Belgium; (A.C.); (P.d.T.)
| | - François Jouret
- Division of Nephrology, University of Liège Hospital (ULg CHU), University of Liège, B-4000 Liège, Belgium; (A.S.-R.); (J.-M.K.); (F.J.)
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, B-4000 Liège, Belgium
| |
Collapse
|
30
|
Maas RM, Deng Y, Dersjant-Li Y, Petit J, Verdegem MCJ, Schrama JW, Kokou F. Exogenous enzymes and probiotics alter digestion kinetics, volatile fatty acid content and microbial interactions in the gut of Nile tilapia. Sci Rep 2021; 11:8221. [PMID: 33859242 PMCID: PMC8050056 DOI: 10.1038/s41598-021-87408-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Sustainable aquafeed production requires fishmeal replacement, leading to an increasing use of plant-derived ingredients. As a consequence, higher levels of antinutritional substances, such as non-starch polysaccharides and phytate, are present in aquafeeds, with negative effects on fish performance, nutrient digestibility and overall gut health. To alleviate these negative effects, providing exogenous digestive enzymes and/or probiotics can be an effective solution. In this study, we tested the effect of dietary supplementation of enzymes (phytase and xylanase) and probiotics (three strains of Bacillus amyloliquefaciens) on nutrient digestion kinetics and volatile fatty acid content along the gut, and the distal gut microbiome diversity in Nile tilapia. Chyme volatile fatty content was increased with probiotic supplementation in the proximal gut, while lactate content, measured for the first time in vivo in fish, decreased with enzymes along the gut. Enzyme supplementation enhanced crude protein, Ca and P digestibility in proximal and middle gut. Enzymes and probiotics supplementation enhanced microbial interactions as shown by network analysis, while increased the abundance of lactic acid bacteria and Bacillus species. Such results suggest that supplementation with exogenous enzymes and probiotics increases nutrient availability, while at the same time benefits gut health and contributes to a more stable microbiome environment.
Collapse
Affiliation(s)
- Roel M. Maas
- grid.4818.50000 0001 0791 5666Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Yale Deng
- grid.4818.50000 0001 0791 5666Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, The Netherlands
| | | | - Jules Petit
- grid.4818.50000 0001 0791 5666Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Marc C. J. Verdegem
- grid.4818.50000 0001 0791 5666Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Johan W. Schrama
- grid.4818.50000 0001 0791 5666Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Fotini Kokou
- grid.4818.50000 0001 0791 5666Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
31
|
Poll BG, Xu J, Jun S, Sanchez J, Zaidman NA, He X, Lester L, Berkowitz DE, Paolocci N, Gao WD, Pluznick JL. Acetate, a Short-Chain Fatty Acid, Acutely Lowers Heart Rate and Cardiac Contractility Along with Blood Pressure. J Pharmacol Exp Ther 2021; 377:39-50. [PMID: 33414131 DOI: 10.1124/jpet.120.000187] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are metabolites produced almost exclusively by the gut microbiota and are an essential mechanism by which gut microbes influence host physiology. Given that SCFAs induce vasodilation, we hypothesized that they might have additional cardiovascular effects. In this study, novel mechanisms of SCFA action were uncovered by examining the acute effects of SCFAs on cardiovascular physiology in vivo and ex vivo. Acute delivery of SCFAs in conscious radiotelemetry-implanted mice results in a simultaneous decrease in both mean arterial pressure and heart rate (HR). Inhibition of sympathetic tone by the selective β-1 adrenergic receptor antagonist atenolol blocks the acute drop in HR seen with acetate administration, yet the decrease in mean arterial pressure persists. Treatment with tyramine, an indirect sympathomimetic, also blocks the acetate-induced acute drop in HR. Langendorff preparations show that acetate lowers HR only after long-term exposure and at a smaller magnitude than seen in vivo. Pressure-volume loops after acetate injection show a decrease in load-independent measures of cardiac contractility. Isolated trabecular muscle preparations also show a reduction in force generation upon SCFA treatment, though only at supraphysiological concentrations. These experiments demonstrate a direct cardiac component of the SCFA cardiovascular response. These data show that acetate affects blood pressure and cardiac function through parallel mechanisms and establish a role for SCFAs in modulating sympathetic tone and cardiac contractility, further advancing our understanding of the role of SCFAs in blood pressure regulation. SIGNIFICANCE STATEMENT: Acetate, a short-chain fatty acid, acutely lowers heart rate (HR) as well as mean arterial pressure in vivo in radiotelemetry-implanted mice. Acetate is acting in a sympatholytic manner on HR and exerts negative inotropic effects in vivo. This work has implications for potential short-chain fatty acid therapeutics as well as gut dysbiosis-related disease states.
Collapse
Affiliation(s)
- Brian G Poll
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jiaojiao Xu
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Seungho Jun
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jason Sanchez
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Nathan A Zaidman
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Xiaojun He
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Laeben Lester
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Dan E Berkowitz
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Nazareno Paolocci
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Wei Dong Gao
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jennifer L Pluznick
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| |
Collapse
|
32
|
Cutignano A, Siano F, Romano R, Aiello A, Pizzolongo F, Berni Canani R, Paparo L, Nocerino R, Di Scala C, Addeo F, Picariello G. Short-term effects of dietary bovine milk on fatty acid composition of human milk: A preliminary multi-analytical study. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1154:122189. [PMID: 32861173 DOI: 10.1016/j.jchromb.2020.122189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
The fatty acid (FA) composition of human milk (HM) from N = 9 Italian healthy donors following a free diet exhibited FA-dependent ranges of variability, as assessed by GC-FID. The possible short-term changes in the FA profile were monitored in the milk of lactating mothers (three) collected at five time points over a 6 h period, following an oral load (200 mL) of bovine milk. An array of techniques was exploited, including UHPLC-ESI-MS/MS of intact lipids and MALDI-TOF MS before and after chemical hydrogenation or bromination, in addition to MALDI-TOF MS analysis of FA after saponification, to monitor short-chain and odd-chain FA in HM as markers of bovine milk fat. A single administration of bovine milk did not appreciably modify the lipid pattern, suggesting that the maternal diet could induce not detectable short-term changes on the lipid composition of HM. Diet-induced increase of butyric acid was also excluded by 13C NMR. The functions that HM FA exert in infant physiology appear finely regulated through maternal metabolism.
Collapse
Affiliation(s)
- Adele Cutignano
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche (CNR), Via Campi Flegrei 34, 80078 Pozzuoli (Napoli), Italy
| | - Francesco Siano
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche (CNR), Via Roma 64, 83100 Avellino, Italy
| | - Raffaele Romano
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Alessandra Aiello
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Fabiana Pizzolongo
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Roberto Berni Canani
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Lorella Paparo
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Rita Nocerino
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Carmen Di Scala
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Francesco Addeo
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Gianluca Picariello
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche (CNR), Via Roma 64, 83100 Avellino, Italy.
| |
Collapse
|
33
|
Chuang ST, Ho ST, Tu PW, Li KY, Kuo YL, Shiu JS, Wang SY, Chen MJ. The Rumen Specific Bacteriome in Dry Dairy Cows and Its Possible Relationship with Phenotypes. Animals (Basel) 2020; 10:E1791. [PMID: 33019774 PMCID: PMC7601041 DOI: 10.3390/ani10101791] [Citation(s) in RCA: 6] [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: 08/13/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Most microbiome studies of dairy cows have investigated the compositions and functions of rumen microbial communities in lactating dairy cows. The importance of the relationships among hosts, microbiota, diet composition, and milk production remains unknown in dry dairy cows. Thus, in the present study, the composition of the rumen microbiome in cows from three dairy farms was investigated to identify core bacteria contributing to various physiological roles during rumen fermentation in dry dairy cows. The results indicated that ruminal fluid in dry dairy cows from different regional farms had core rumen microbiota that could be clearly distinguished from that of cows of the other farms. Further identification of key microorganisms associated with each farm revealed that Prevotella, Methanobrevibacter, Pseudobutyrivibrio, Ruminococcus, Bacteroides, and Streptococcus were major contributors. Spearman's correlation indicated that the abundance of genera such as Prevotella and Ruminococcus in dry dairy cows could indicate milk yield in the previous lactating period. Functional pathway analysis of the rumen bacterial communities demonstrated that amino acid metabolism and carbohydrate metabolism were the major pathways. Our findings provide knowledge of the composition and predicted functions of rumen microbiota in dry dairy cows from regional farms, which underscore the importance of the relationships among hosts, microbiota, diet composition, and milk production.
Collapse
Affiliation(s)
- Shih-Te Chuang
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan;
| | - Shang-Tse Ho
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; (S.-T.H.); (P.-W.T.); (K.-Y.L.); (S.-Y.W.)
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi 600, Taiwan
| | - Po-Wen Tu
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; (S.-T.H.); (P.-W.T.); (K.-Y.L.); (S.-Y.W.)
| | - Kuan-Yi Li
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; (S.-T.H.); (P.-W.T.); (K.-Y.L.); (S.-Y.W.)
| | - Yu-Lun Kuo
- Biotools Co., Ltd., New Taipei City 221, Taiwan;
| | - Jia-Shian Shiu
- Hengchun Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Pingtung 946, Taiwan;
| | - Sheng-Yao Wang
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; (S.-T.H.); (P.-W.T.); (K.-Y.L.); (S.-Y.W.)
| | - Ming-Ju Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; (S.-T.H.); (P.-W.T.); (K.-Y.L.); (S.-Y.W.)
| |
Collapse
|
34
|
Diallo AF, Lockwood MB, Maki KA, Franks AT, Roy A, Jaime-Lara R, Joseph PV, Henderson WA, Chung SY, McGrath J, Green SJ, Fink AM. Metabolic Profiling of Blood and Urine for Exploring the Functional Role of the Microbiota in Human Health. Biol Res Nurs 2020; 22:449-457. [PMID: 32723087 DOI: 10.1177/1099800420941080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The quantification of metabolites in blood and urine allows nurses to explore new hypotheses about the microbiome. This review summarizes findings from recent studies with a focus on how the state of the science can influence future nursing research initiatives. Metabolomics can advance nursing research by identifying physiologic/pathophysiologic processes underlying patients' symptoms and can be useful for testing the effects of nursing interventions. To date, metabolomics has been used to study cardiovascular, respiratory, endocrine, autoimmune, and infectious conditions, with research focused on understanding the microbial metabolism of substrates resulting in circulating/excreted biomarkers such as trimethylamine N-oxide. This review provides specific recommendations for the collection of specimens and goals for future studies.
Collapse
Affiliation(s)
- Ana F Diallo
- Institute of Inclusion, Inquiry & Innovation (iCubed), Family and Community Health Nursing, School of Nursing, Virginia Commonwealth University, Richmond, VA, USA
| | - Mark B Lockwood
- Department of Biobehavioral Health Science, College of Nursing, University of Illinois at Chicago, Chicago, IL, USA
| | - Katherine A Maki
- Department of Biobehavioral Health Science, College of Nursing, University of Illinois at Chicago, Chicago, IL, USA
| | - Alexis T Franks
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Abhrarup Roy
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Rosario Jaime-Lara
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Paule V Joseph
- Sensory Science & Metabolism Unit, Biobehavioral Branch, Division of Intramural Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Wendy A Henderson
- Digestive Disorders Unit, Biobehavioral Branch, Division of Intramural Research, 2511National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Seon Yoon Chung
- School of Nursing, University of Maryland, Baltimore, MD, USA
| | - Jacqueline McGrath
- School of Nursing, University of Texas Health Science Center, San Antonio, TX, USA
| | - Stefan J Green
- Sequencing Core, Research Resources Center, 14681University of Illinois at Chicago, Chicago, IL, USA
| | - Anne M Fink
- Department of Biobehavioral Health Science, College of Nursing, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
35
|
Polymannuronic acid prevents dopaminergic neuronal loss via brain-gut-microbiota axis in Parkinson's disease model. Int J Biol Macromol 2020; 164:994-1005. [PMID: 32710966 DOI: 10.1016/j.ijbiomac.2020.07.180] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
The study aims to investigate the potentially neuroprotective effects and underlying mechanisms for brown seaweed polysaccharide of polymannuronic acid (PM) against Parkinson's disease (PD) pathogenesis. PD model mice were pretreated with PM via oral gavage once per day for 4 weeks and the preventative effects of PM against neuronal loss together with its modulation on brain-gut-microbiota axis were systematically explored. The results showed PM administration improved motor functions by preventing dopaminergic neuronal loss in the substantia nigra pars compacta (SNpc) and enhanced contents of striatal homovanillic acid (HVA), serotonin (5-HT), 5-hydroxyindole acetic acid (5-HIAA) and γ-aminobutyric acid (GABA) in PD mice. PM significantly alleviated inflammation in gut, brain and systemic circulation as shown by reduced levels or expressions of pro-inflammatory cytokines concurrently and inhibited mitogen-activated protein kinases (MAPK) signaling pathway in mice colon. Meanwhile, PM greatly improved integrity of intestinal barrier and blood brain barrier (BBB) as indicated by increased expressions of tight junction associated proteins in both mice colon and SNpc. Further studies indicated PM treatment resulted in changes of gut microbial compositions, together with great alterations of digestion and metabolism of dietary proteins and fats, which led to surge increase of fecal short chain fatty acids (SCFAs) in the colon of PD mice. In conclusion, pre-administration of PM could provide neuroprotective effects against PD pathogenesis by suppressing inflammation in gut, brain and systemic circulation, and by improving integrity of intestinal barrier and BBB. PM might modulate brain-gut-microbiota axis, at least in part, via gut microbiota derived SCFAs as mediators.
Collapse
|
36
|
Maki KA, Burke LA, Calik MW, Watanabe-Chailland M, Sweeney D, Romick-Rosendale LE, Green SJ, Fink AM. Sleep fragmentation increases blood pressure and is associated with alterations in the gut microbiome and fecal metabolome in rats. Physiol Genomics 2020; 52:280-292. [PMID: 32567509 PMCID: PMC7468692 DOI: 10.1152/physiolgenomics.00039.2020] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota, via the production of metabolites entering the circulation, plays a role in blood pressure regulation. Blood pressure is also affected by the characteristics of sleep. To date, no studies have examined relationships among the gut microbiota/metabolites, blood pressure, and sleep. We hypothesized that fragmented sleep is associated with elevated mean arterial pressure, an altered and dysbiotic gut microbial community, and changes in fecal metabolites. In our model system, rats were randomized to 8 h of sleep fragmentation during the rest phase (light phase) or were undisturbed (controls) for 28 consecutive days. Rats underwent sleep and blood pressure recordings, and fecal samples were analyzed during: baseline (days -4 to -1), early sleep fragmentation (days 0-3), midsleep fragmentation (days 6-13), late sleep fragmentation (days 20-27), and recovery/rest (days 28-34). Less sleep per hour during the sleep fragmentation period was associated with increased mean arterial pressure. Analyses of gut microbial communities and metabolites revealed that putative short chain fatty acid-producing bacteria were differentially abundant between control and intervention animals during mid-/late sleep fragmentation and recovery. Midsleep fragmentation was also characterized by lower alpha diversity, lower Firmicutes:Bacteroidetes ratio, and higher Proteobacteria in intervention rats. Elevated putative succinate-producing bacteria and acetate-producing bacteria were associated with lower and higher mean arterial pressure, respectively, and untargeted metabolomics analysis demonstrates that certain fecal metabolites are significantly correlated with blood pressure. These data reveal associations between sleep fragmentation, mean arterial pressure, and the gut microbiome/fecal metabolome and provide insight to links between disrupted sleep and cardiovascular pathology.
Collapse
Affiliation(s)
- Katherine A Maki
- Department of Biobehavioral Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
- Nursing Department, Nursing Research and Translational Science, National Institutes of Health, Clinical Center, Bethesda, Maryland
| | - Larisa A Burke
- Office of Research Facilitation, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
| | - Michael W Calik
- Department of Biobehavioral Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
| | - Miki Watanabe-Chailland
- NMR-Based Metabolomics Core, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dagmar Sweeney
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois
| | | | - Stefan J Green
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois
| | - Anne M Fink
- Department of Biobehavioral Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
37
|
Chen D, Qiu YB, Gao ZQ, Wu YX, Wan BB, Liu G, Chen JL, Zhou Q, Yu RQ, Pang QF. Sodium Propionate Attenuates the Lipopolysaccharide-Induced Epithelial-Mesenchymal Transition via the PI3K/Akt/mTOR Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6554-6563. [PMID: 32452677 DOI: 10.1021/acs.jafc.0c01302] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Short-chain fatty acids (SCFAs), especially propionate, originate from the fermentation of dietary fiber in the gut and play a key role in inhibiting pulmonary inflammation. Chronic inflammation may induce an epithelial-mesenchymal transition (EMT) in alveolar epithelial cells and result in fibrotic disorders. This study was designed to investigate the beneficial effect of sodium propionate (SP) on lipopolysaccharide (LPS)-induced EMT. In cultured BEAS-2B cells, the protein expression levels of E-cadherin, α-smooth muscle actin (SMA), and vimentin were 0.66 ± 0.20, 1.44 ± 0.23, and 1.32 ± 0.21 in the LPS group vs 1.11 ± 0.36 (P < 0.05), 1.04 ± 0.30 (P < 0.05), and 0.96 ± 0.13 (P < 0.01) in the LPS + SP group (mean ± standard deviation), respectively. Meanwhile, LPS-triggered inflammatory cytokines and extracellular proteins were also reduced by SP administration in BEAS-2B cells. Moreover, SP treatment attenuated inflammation, EMT, extracellular matrix (ECM) deposition, and even fibrosis in a mouse EMT model. In terms of mechanism, LPS-treated BEAS-2B cells exhibited a higher level of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) phosphorylation, which was interrupted by SP treatment. It is worth noting that the blockade of the PI3K/Akt/mTOR signaling cascade reduced the LPS-evoked EMT process in BEAS-2B cells. These results suggest that SP can block LPS-induced EMT via inhibition of the PI3K/Akt/mTOR signaling cascade, which provides a basis for possible clinical use of SP in airway and lung diseases.
Collapse
Affiliation(s)
- Dan Chen
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Yu-Bao Qiu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Zhi-Qi Gao
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Ya-Xian Wu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Bin-Bin Wan
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Gang Liu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Jun-Liang Chen
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Ren-Qiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Qing-Feng Pang
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| |
Collapse
|
38
|
Li B, Chen H, Cao L, Hu Y, Chen D, Yin Y. Effects of an Escherichia coli exopolysaccharide on human and mouse gut microbiota in vitro. Int J Biol Macromol 2020; 150:991-999. [DOI: 10.1016/j.ijbiomac.2019.10.186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
|
39
|
Bica R, Palarea-Albaladejo J, Kew W, Uhrin D, Pacheco D, Macrae A, Dewhurst RJ. Nuclear Magnetic Resonance to Detect Rumen Metabolites Associated with Enteric Methane Emissions from Beef Cattle. Sci Rep 2020; 10:5578. [PMID: 32221381 PMCID: PMC7101347 DOI: 10.1038/s41598-020-62485-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/13/2020] [Indexed: 11/10/2022] Open
Abstract
This study presents the application of metabolomics to evaluate changes in the rumen metabolites of beef cattle fed with three different diet types: forage-rich, mixed and concentrate-rich. Rumen fluid samples were analysed by 1H-NMR spectroscopy and the resulting spectra were used to characterise and compare metabolomic profiles between diet types and assess the potential for NMR metabolite signals to be used as proxies of methane emissions (CH4 in g/kg DMI). The dataset available consisted of 128 measurements taken from 4 experiments with CH4 measurements taken in respiration chambers. Predictive modelling of CH4 was conducted by partial least squares (PLS) regression, fitting calibration models either using metabolite signals only as predictors or using metabolite signals as well as other diet and animal covariates (DMI, ME, weight, BW0.75, DMI/BW0.75). Cross-validated R2 were 0.57 and 0.70 for the two models respectively. The cattle offered the concentrate-rich diet showed increases in alanine, valerate, propionate, glucose, tyrosine, proline and isoleucine. Lower methane yield was associated with the concentrate-rich diet (p < 0.001). The results provided new insight into the relationship between rumen metabolites, CH4 production and diets, as well as showing that metabolites alone have an acceptable association with the variation in CH4 production from beef cattle.
Collapse
Affiliation(s)
- R Bica
- Scotland's Rural College, SRUC, West Mains Rd, Edinburgh, EH9 3JG, United Kingdom. .,Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, United Kingdom. .,AgResearch Grasslands Research Centre, Tennent Drive, 11 Dairy Farm Road, Palmerston North, 4442, New Zealand.
| | - J Palarea-Albaladejo
- Biomathematics and Statistics Scotland, JCMB, Peter Guthrie Tait Road, The King's Buildings, Edinburgh, EH9 3FD, United Kingdom
| | - W Kew
- The University of Edinburgh, EaStCHEM School of Chemistry, The King's Buildings, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - D Uhrin
- The University of Edinburgh, EaStCHEM School of Chemistry, The King's Buildings, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - D Pacheco
- AgResearch Grasslands Research Centre, Tennent Drive, 11 Dairy Farm Road, Palmerston North, 4442, New Zealand
| | - A Macrae
- Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, United Kingdom
| | - R J Dewhurst
- Scotland's Rural College, SRUC, West Mains Rd, Edinburgh, EH9 3JG, United Kingdom
| |
Collapse
|
40
|
Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne) 2020; 11:25. [PMID: 32082260 PMCID: PMC7005631 DOI: 10.3389/fendo.2020.00025] [Citation(s) in RCA: 1261] [Impact Index Per Article: 315.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
A substantial body of evidence supports that the gut microbiota plays a pivotal role in the regulation of metabolic, endocrine and immune functions. In recent years, there has been growing recognition of the involvement of the gut microbiota in the modulation of multiple neurochemical pathways through the highly interconnected gut-brain axis. Although amazing scientific breakthroughs over the last few years have expanded our knowledge on the communication between microbes and their hosts, the underpinnings of microbiota-gut-brain crosstalk remain to be determined. Short-chain fatty acids (SCFAs), the main metabolites produced in the colon by bacterial fermentation of dietary fibers and resistant starch, are speculated to play a key role in neuro-immunoendocrine regulation. However, the underlying mechanisms through which SCFAs might influence brain physiology and behavior have not been fully elucidated. In this review, we outline the current knowledge about the involvement of SCFAs in microbiota-gut-brain interactions. We also highlight how the development of future treatments for central nervous system (CNS) disorders can take advantage of the intimate and mutual interactions of the gut microbiota with the brain by exploring the role of SCFAs in the regulation of neuro-immunoendocrine function.
Collapse
Affiliation(s)
- Ygor Parladore Silva
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Andressa Bernardi
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Rudimar Luiz Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- *Correspondence: Rudimar Luiz Frozza
| |
Collapse
|
41
|
Effects of Dietary Energy on Growth Performance, Rumen Fermentation and Bacterial Community, and Meat Quality of Holstein-Friesians Bulls Slaughtered at Different Ages. Animals (Basel) 2019; 9:ani9121123. [PMID: 31835883 PMCID: PMC6941049 DOI: 10.3390/ani9121123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Beef tenderness, flavour and juiciness are quality characteristics of consumer eating satisfaction. Marbling level of beef could determine quality characteristics by concentrating water soluble flavour molecules and releasing low fat-solubility volatiles faster. Increasing number of Holstein-Friesians bulls are used for beef production, and this breed is considered to have lower meat quality than purebred and crossbred beef cattle. Therefore, in this study we designed different dietary energy levels and slaughter ages to find ways to improve the meat quality of Holstein-Friesians bulls in beef production. We found that Holstein-Friesians bulls fed with 10.90 and 11.68 MJ/kg metabolizable energy diets, compared to a diet with 10.12 MJ/kg of metabolizable energy, had higher average daily gain, dressing percentage, intramuscular fat content and water holding capacity at 23 or 26 months of age. This study provides a basis to use bulls to produce beef by providing indicators of growth performance, rumen fermentation and ruminal bacteria community, and meat quality. Abstract The objective of this study was to evaluate the effects of dietary energy levels on growth performance, rumen fermentation and bacterial community, and meat quality of Holstein-Friesians bulls slaughtered at different ages. Thirty-six Holstein-Friesians bulls (17 months of age) were divided into a 3 × 3 factorial experiment with three energy levels (LE, ME and HE; metabolizable energy is 10.12, 10.90 and 11.68 MJ/kg, respectively) of diets, and three slaughter ages (20, 23 and 26 months). Results indicated that bulls fed with ME and HE diets had higher dry matter intake, average daily gain, and dressing percentage at 23 or 26 months of age. The ME and HE diets also reduced bacterial diversity, altered relative abundances of bacteria and produced lower concentrations of acetate, but higher butyrate and valerate concentrations in rumen fluid. Increasing in dietary energy and slaughter age increased the intramuscular fat (IMF) and water holding capacity. In summary, Holstein-Friesians bulls fed with ME and HE diets, slaughtered at 23 and 26 months of age could be a good choice to produce beef with high IMF. Slaughter age may have less influence than dietary energy in altering fermentation by increasing amylolytic bacteria and decreasing cellulolytic bacteria, and thus, further affecting meat quality.
Collapse
|
42
|
Niacin Protects against Butyrate-Induced Apoptosis in Rumen Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2179738. [PMID: 31737165 PMCID: PMC6815573 DOI: 10.1155/2019/2179738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/04/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023]
Abstract
The effects and underlying mechanisms of butyrate and butyrate+niacin on apoptosis in sheep rumen epithelial cells were investigated. Cells were exposed to butyrate (0-140 mM) for 6 h. A low concentration (20 mM) of butyrate increased cell viability and promoted growth whereas high concentrations (40-140 mM) inhibited proliferation. Cells were then cocultured with 120 mM butyrate and niacin (0-100 mM) for 6 h. Niacin addition attenuated butyrate-induced cellular damage and promoted proliferation at 20-80 mM; 40 mM presented the optimal effect. Higher concentrations (100 mM) of niacin resulted in low cell viability. Subsequent experiments confirmed that 120 mM butyrate increased intracellular reactive oxygen species (ROS) production and reduced the intracellular total antioxidant capacity (T-AOC) versus the untreated control. Compared with 120 mM butyrate, cotreatment with 40 mM niacin significantly reduced the intracellular ROS content and increased the intracellular T-AOC. Flow cytometry analysis revealed that 120 mM butyrate increased the proportion of apoptotic cells by 17.8% versus the untreated control, and 120 mM butyrate+40 mM niacin treatment reduced the proportion of apoptotic cells by 28.6% and 39.4% versus the untreated control and butyrate treatment, respectively. Treatment with 120 mM butyrate increased caspase-9 and p53 mRNA levels and decreased the expression of Bcl-2 and Bax, and the Bcl-2/Bax ratio versus the untreated control. Treatment with 120 mM butyrate+40 mM niacin downregulated the expression of caspase-3 and p53 and increased the expression of Bcl-2 and Bax versus butyrate treatment alone but had no effect on the Bcl-2/Bax ratio. Thus, high concentrations of butyrate may induce rumen epithelial cell apoptosis by increasing oxidative stress and inducing caspase-9 and p53 expression. Cotreatment with niacin regulates apoptosis-related gene expression by reducing intracellular ROS production and DNA damage and downregulating caspase-3 and p53 expressions to protect rumen epithelial cells against butyrate-induced apoptosis.
Collapse
|
43
|
Yamamura R, Nakamura K, Kitada N, Aizawa T, Shimizu Y, Nakamura K, Ayabe T, Kimura T, Tamakoshi A. Associations of gut microbiota, dietary intake, and serum short-chain fatty acids with fecal short-chain fatty acids. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2019; 39:11-17. [PMID: 32010539 PMCID: PMC6971417 DOI: 10.12938/bmfh.19-010] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022]
Abstract
In recent years, short-chain fatty acids (SCFAs) have been reported to play an important role in maintaining human health. Fecal SCFA concentrations correlate well with colonic SCFA status
and gut microbiota composition. However, the associations with the gut microbiota functional pathway, dietary intake, blood SCFAs, and fecal SCFAs remain uncertain. To clarify these
relationships, we collected fecal samples, blood samples, and dietary habit data from 12 healthy adults aged 22–51 years. The relative abundance of several SCFA-producing bacteria, gut
microbiota diversity, and functional pathways related to SCFA biosynthesis were positively associated with fecal SCFAs even after adjusting for age and sex. Furthermore, fecal acetate was
likely to be positively associated with serum acetate. By contrast, dietary intake was not associated with fecal SCFAs. Overall, the present study highlights the potential usefulness of
fecal SCFAs as an indicator of the gut microbiota ecosystem and dynamics of SCFAs in the human body.
Collapse
Affiliation(s)
- Ryodai Yamamura
- Graduate School of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Koshi Nakamura
- Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan.,Faculty of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Naoya Kitada
- Graduate School of Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo 060-0810, Japan
| | - Tomoyasu Aizawa
- Faculty of Advanced Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo 060-0810, Japan
| | - Yu Shimizu
- Graduate School of Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo 060-0810, Japan
| | - Kiminori Nakamura
- Faculty of Advanced Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo 060-0810, Japan
| | - Tokiyoshi Ayabe
- Faculty of Advanced Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo 060-0810, Japan
| | - Takashi Kimura
- Faculty of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Akiko Tamakoshi
- Faculty of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
44
|
Yue SJ, Liu J, Wang WX, Wang AT, Yang XY, Guan HS, Wang CY, Yan D. Berberine treatment-emergent mild diarrhea associated with gut microbiota dysbiosis. Biomed Pharmacother 2019; 116:109002. [DOI: 10.1016/j.biopha.2019.109002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/28/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
|
45
|
Abstract
Short-chain fatty acids (SCFAs), the main metabolites produced by bacterial fermentation of dietary fibre in the gastrointestinal tract, are speculated to have a key role in microbiota-gut-brain crosstalk. However, the pathways through which SCFAs might influence psychological functioning, including affective and cognitive processes and their neural basis, have not been fully elucidated. Furthermore, research directly exploring the role of SCFAs as potential mediators of the effects of microbiota-targeted interventions on affective and cognitive functioning is sparse, especially in humans. This Review summarizes existing knowledge on the potential of SCFAs to directly or indirectly mediate microbiota-gut-brain interactions. The effects of SCFAs on cellular systems and their interaction with gut-brain signalling pathways including immune, endocrine, neural and humoral routes are described. The effects of microbiota-targeted interventions such as prebiotics, probiotics and diet on psychological functioning and the putative mediating role of SCFA signalling will also be discussed, as well as the relationship between SCFAs and psychobiological processes. Finally, future directions to facilitate direct investigation of the effect of SCFAs on psychological functioning are outlined.
Collapse
|
46
|
Huart J, Leenders J, Taminiau B, Descy J, Saint-Remy A, Daube G, Krzesinski JM, Melin P, de Tullio P, Jouret F. Gut Microbiota and Fecal Levels of Short-Chain Fatty Acids Differ Upon 24-Hour Blood Pressure Levels in Men. Hypertension 2019; 74:1005-1013. [PMID: 31352822 DOI: 10.1161/hypertensionaha.118.12588] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gut microbiota may influence blood pressure (BP), namely via end products of carbohydrate fermentation. After informed consent, male volunteers were prospectively categorized into 3 groups upon European Society of Hypertension criteria based on 24-hour ambulatory BP measurements: (1) hypertension, (2) borderline hypertension, and (3) normotension. Stool, urine and serum samples were collected in fasting conditions. Gut microbiota was characterized by 16S amplicon sequencing. Metabolomics, including quantification of short-chain fatty acids, was conducted using nuclear magnetic resonance. Two-way ANOVA combined with Tukey post hoc test, as well as multiple permutation test and Benjamini-Hochberg-Yekutieli false discovery rate procedure, was used. The cohort included 54 males: 38 hypertensive (including 21 under treatment), 7 borderline, and 9 normotensive. No significant difference was observed between groups concerning age, body mass index, smoking habits, and weekly alcohol consumption. The genus Clostridium sensu stricto 1 positively correlated with BP levels in nontreated patients (n=33). This correlation was significant after multiple permutation tests but was not substantiated following false discovery rate adjustment. Short-chain fatty acid levels were significantly different among groups, with higher stool levels of acetate, butyrate, and propionate in hypertensive versus normotensive individuals. No difference was observed in serum and urine metabolomes. Correlation between stool metabolome and 24-hour BP levels was evidenced, with R2 reaching 0.9. Our pilot study based on 24-hour ambulatory BP measurements, 16S amplicon sequencing, and metabolomics supports an association between gut microbiota and BP homeostasis, with changes in stool abundance of short-chain fatty acids.
Collapse
Affiliation(s)
- Justine Huart
- From the Division of Nephrology, University of Liège Hospital (J.H., A.S.-R., J.-M.K., F.J.), University of Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée, Cardiovascular Sciences (J.H., J.-M.K., F.J.), University of Liège, Belgium
| | - Justine Leenders
- Center for Interdisciplinary Research on Medicines, Metabolomics Group (J.L., P.d.T.), University of Liège, Belgium
| | - Bernard Taminiau
- Department of Food Sciences, Faculty of Veterinary Medicine, Fundamental and Applied Research Center for Animal and Health (B.T., G.D.), University of Liège, Belgium
| | - Julie Descy
- Clinical Microbiology, University of Liège Hospital (J.D., P.M.), University of Liège, Belgium
| | - Annie Saint-Remy
- From the Division of Nephrology, University of Liège Hospital (J.H., A.S.-R., J.-M.K., F.J.), University of Liège, Belgium
| | - Georges Daube
- Department of Food Sciences, Faculty of Veterinary Medicine, Fundamental and Applied Research Center for Animal and Health (B.T., G.D.), University of Liège, Belgium
| | - Jean-Marie Krzesinski
- From the Division of Nephrology, University of Liège Hospital (J.H., A.S.-R., J.-M.K., F.J.), University of Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée, Cardiovascular Sciences (J.H., J.-M.K., F.J.), University of Liège, Belgium
| | - Pierrette Melin
- Clinical Microbiology, University of Liège Hospital (J.D., P.M.), University of Liège, Belgium
| | - Pascal de Tullio
- Center for Interdisciplinary Research on Medicines, Metabolomics Group (J.L., P.d.T.), University of Liège, Belgium
| | - François Jouret
- From the Division of Nephrology, University of Liège Hospital (J.H., A.S.-R., J.-M.K., F.J.), University of Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée, Cardiovascular Sciences (J.H., J.-M.K., F.J.), University of Liège, Belgium
| |
Collapse
|
47
|
López-Colom P, Castillejos L, Barba-Vidal E, Zhu Y, Puyalto M, Mallo JJ, Martín-Orúe SM. Response of gastrointestinal fermentative activity and colonic microbiota to protected sodium butyrate and protected sodium heptanoate in weaned piglets challenged with ETEC F4 . Arch Anim Nutr 2019; 73:339-359. [PMID: 31342760 DOI: 10.1080/1745039x.2019.1641376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study aimed to evaluate the potential of two new fat-protected butyrate or heptanoate salts to improve gut health and control post-weaning colibacillosis in weaning piglets challenged with enterotoxigenic Escherichia coli (ETEC) F4+, particularly focusing on their impact on intestinal microbiota and fermentative activity along the gastrointestinal tract (GIT). Seventy-two 21-d-old pigs were fed a plain diet (CTR) or supplemented with sodium butyrate (BUT) or sodium heptanoate (HPT), both at 0.3%. After a week of adaptation, animals were orally challenged at days 8 and 9 with 5.8 · 109 and 6.6 · 1010 cfu, respectively, and were euthanised on d 4 and d 8 post-inoculation (PI) (n = 8) to collect blood, digesta and tissue samples and characterise microbial groups, pathogen loads (qPCR), fermentation, ileal histomorphometry and immune markers. Colonic microbiota was analysed by 16S rRNA gene MiSeq sequencing. Supplementing both acid salts did not compensate clinical challenge effects nor performance impairments and neither histomorphometry nor serum biomarkers. Changes in the gastric fermentative activity were registered, BUT reducing lactic acid concentrations (day 8 PI), and with HPT fewer animals presenting detectable concentrations of propionic, butyric and valeric acids. At ileum BUT increased acetic acid concentration (day 8 PI), and both additives reduced short-chain fatty acids (SCFA) in the colon. Increases in enterobacteria and coliforms counts in ileal digesta (day 4 PI, p < 0.10) and mucosa scrapes (p < 0.05) were registered although E. coli F4 gene copies were unaffected. Regarding changes in the colonic microbiota (day 4 PI), Prevotellaceae and Prevotella were promoted with BUT supplementation whereas only minor groups were modified in HPT-treated animals. Summarising, although the pathogen loads or inflammatory mediators remained unresponsive, butyrate and heptanoate showed a significant impact on microbial fermentation along the whole GIT, being able to modify different bacterial groups at the colon. It could be hypothesised that these effects might be mediated by a carry-over effect of the changes observed in gastric fermentation, but possibly also to a better nutrient digestion in the foregut as a result of the reduced colonic SCFA concentrations.
Collapse
Affiliation(s)
- Paola López-Colom
- a Animal Nutrition and Welfare Service, Animal and Food Science Department, Facultat de Veterinària, Universitat Autònoma de Barcelona , Bellaterra , Spain
| | - Lorena Castillejos
- a Animal Nutrition and Welfare Service, Animal and Food Science Department, Facultat de Veterinària, Universitat Autònoma de Barcelona , Bellaterra , Spain
| | - Emili Barba-Vidal
- a Animal Nutrition and Welfare Service, Animal and Food Science Department, Facultat de Veterinària, Universitat Autònoma de Barcelona , Bellaterra , Spain
| | - Yanan Zhu
- a Animal Nutrition and Welfare Service, Animal and Food Science Department, Facultat de Veterinària, Universitat Autònoma de Barcelona , Bellaterra , Spain
| | | | | | - Susana M Martín-Orúe
- a Animal Nutrition and Welfare Service, Animal and Food Science Department, Facultat de Veterinària, Universitat Autònoma de Barcelona , Bellaterra , Spain
| |
Collapse
|
48
|
Park CJ, Armenia SJ, Shaughnessy MP, Greig CJ, Cowles RA. Potentiation of serotonin signaling leads to increased carbohydrate and lipid absorption in the murine small intestine. J Pediatr Surg 2019; 54:1245-1249. [PMID: 30879746 DOI: 10.1016/j.jpedsurg.2019.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/21/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Enteric serotonin influences intestinal homeostasis and functions as a mucosal growth factor. Previously, we demonstrated increased mucosal surface area and enhanced crypt cell proliferation in serotonin reuptake transporter (SERT)-deficient mice. Therefore, we hypothesized that serotonin-mediated mucosal growth would also result in enhanced carbohydrate and lipid absorption. MATERIAL AND METHODS Wild-type C57Bl/6 (WT) and SERT-knockout (SERTKO) mice were fasted then gavaged with D-xylose or boron-dipyrromethene (BODIPY) FL-C12 medium-chain fatty acid analog. Serum D-xylose and BODIPY concentrations were serially measured from blood drawn at 30 to 360 min post-gavage. Small intestine was harvested from both groups for comparison of morphometric parameters. Area under the curve of plotted graphs was calculated, and means were compared with Student's t-test to a significance of p < 0.05. RESULTS Villus height and crypt depth were significantly greater in the middle and distal small intestine of SERTKO animals compared to WT. Overall absorption of D-xylose and BODIPY was greater in SERTKO animals compared to WT animals. Absorption of D-xylose was persistently elevated in SERTKO animals, while there was an initial delay in BODIPY absorption followed by a sustained and significantly greater absorption in SERTKO animals at 60-360 min after gavage. CONCLUSION Potentiation of serotonin signaling in SERTKO mice results in small intestinal mucosal growth and enhanced carbohydrate and fat absorption in vivo. These functional increases support the concept of targeting the serotonin signaling system to augment intestinal adaptation in the setting of intestinal failure.
Collapse
Affiliation(s)
- Christine J Park
- Department of Surgery, Section of Pediatric Surgery at Yale University, New Haven, CT
| | - Sarah J Armenia
- Department of Surgery, Section of Pediatric Surgery at Yale University, New Haven, CT
| | - Matthew P Shaughnessy
- Department of Surgery, Section of Pediatric Surgery at Yale University, New Haven, CT
| | - Chasen J Greig
- Department of Surgery, Section of Pediatric Surgery at Yale University, New Haven, CT
| | - Robert A Cowles
- Department of Surgery, Section of Pediatric Surgery at Yale University, New Haven, CT.
| |
Collapse
|
49
|
Adaptation of intestinal fermentation over time in the growing pig is influenced by the amount of kiwi fruit consumed. Br J Nutr 2019; 121:601-614. [DOI: 10.1017/s0007114518003574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AbstractThe effect of kiwi fruit at two dietary levels on the adaptation of intestinal fermentation over time in the growing pig was studied. A semi-synthetic fibre-free diet and two semi-synthetic diets containing kiwi fruit as a model fibre source (133 or 266 g/kg (DM basis); 28 or 48 g fibre/kg) were formulated and the diets contained titanium dioxide as an indigestible marker. A total of fourteen ileal cannulated pigs (41 kg body weight) were fed the fibre-free diet for 7 d followed by either the low or high kiwi fruit-containing diets (n 7/diet) for a further 44 d. Ileal digesta and faeces were collected at five times throughout the study. Ileal digesta were fermented (in vitro) with a standard pooled human faecal inoculum, while fresh pig faeces were used as inocula to ferment in vitro a standard purified fibre. Observations were normalised for diet DM intake using the marker. The 16S ribosomal RNA gene copy number of ileal and total faecal bacteria were high for the high-kiwi fruit level diet (P<0·05). The ileal bacteria tended to decrease over time (P<0·1), while the faecal bacteria increased (P<0·05), at the same rate for both diets. The amounts of crude protein and insoluble dietary fibre entering the hindgut changed over time similarly for both diets, whereas for starch it changed only for the low kiwi fruit-containing diet (P<0·05). Changes over time were also observed for the predicted hindgut valeric acid production and butyric acid absorption (P<0·05). In conclusion, adaptational changes over time of some characteristics of intestinal fermentation depended on the dietary level of kiwi fruit.
Collapse
|
50
|
Aschenbach JR, Zebeli Q, Patra AK, Greco G, Amasheh S, Penner GB. Symposium review: The importance of the ruminal epithelial barrier for a healthy and productive cow. J Dairy Sci 2019; 102:1866-1882. [DOI: 10.3168/jds.2018-15243] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/04/2018] [Indexed: 12/22/2022]
|