51
|
Souders CL, Zubcevic J, Martyniuk CJ. Tumor Necrosis Factor Alpha and the Gastrointestinal Epithelium: Implications for the Gut-Brain Axis and Hypertension. Cell Mol Neurobiol 2022; 42:419-437. [PMID: 33594519 PMCID: PMC8364923 DOI: 10.1007/s10571-021-01044-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/11/2021] [Indexed: 12/17/2022]
Abstract
The colonic epithelium is the site of production and transport of many vasoactive metabolites and neurotransmitters that can modulate the immune system, affect cellular metabolism, and subsequently regulate blood pressure. As an important interface between the microbiome and its host, the colon can contribute to the development of hypertension. In this critical review, we highlight the role of colonic inflammation and microbial metabolites on the gut brain axis in the pathology of hypertension, with special emphasis on the interaction between tumor necrosis factor α (TNFα) and short chain fatty acid (SCFA) metabolites. Here, we review the current literature and identify novel pathways in the colonic epithelium related to hypertension. A network analysis on transcriptome data previously generated in spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats reveals differences in several pathways associated with inflammation involving TNFα (NF-κB and STAT Expression Targets) as well as oxidative stress. We also identify down-regulation of networks associated with gastrointestinal function, cardiovascular function, enteric nervous system function, and cholinergic and adrenergic transmission. The analysis also uncovered transcriptome responses related to glycolysis, butyrate oxidation, and mitochondrial function, in addition to gut neuropeptides that serve as modulators of blood pressure and metabolic function. We present a model for the role of TNFα in regulating bacterial metabolite transport and neuropeptide signaling in the gastrointestinal system, highlighting the complexity of host-microbiota interactions in hypertension.
Collapse
Affiliation(s)
- Christopher L. Souders
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611 USA
| | - Jasenka Zubcevic
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA. .,Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, PO BOX 100274, Gainesville, FL, 32611, USA.
| | - Christopher J. Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611 USA,Corresponding authors contact information: Department of Physiological Sciences, College of Veterinary Medicine, University of Florida PO BOX 100274 GAINESVILLE FL 326100274 United States; and
| |
Collapse
|
52
|
Liu H, Li X, Zhu Y, Huang Y, Zhang Q, Lin S, Fang C, Li L, Lv Y, Mei W, Peng X, Yin J, Liu L. Effect of Plant-Derived n-3 Polyunsaturated Fatty Acids on Blood Lipids and Gut Microbiota: A Double-Blind Randomized Controlled Trial. Front Nutr 2022; 9:830960. [PMID: 35223959 PMCID: PMC8873928 DOI: 10.3389/fnut.2022.830960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/21/2022] [Indexed: 12/29/2022] Open
Abstract
Background Several cardioprotective mechanisms attributed to n-3 polyunsaturated fatty acids (PUFAs) have been widely documented. Significant interest has recently focused on the role of human gut microbiota in metabolic disorders. However, the role of plant-derived n-3 PUFAs on blood lipid profiles is controversial and the effect on gut microbiota is still unclear. Objectives We aimed to perform a double-blind randomized controlled trial to test the effect of plant-derived n-3 PUFAs on the blood lipids and gut microbiota of patients with marginal hyperlipidemia. Methods According to the inclusion and exclusion criteria, 75 participants with marginal hyperlipidemia were randomly assigned to the intervention group (supplied with n-3 PUFA-enriched plant oil) or control group (supplied with corn oil), respectively, for a 3-month treatment. Participants and assessors were blinded to the allocation. The primary outcomes of the trial were the changes in serum lipid levels. Secondary outcomes were changes in gut microbiota and metabolites. For the primary outcomes, we conducted both an intent-to-treat (ITT) analysis and a per protocol (PP) analysis. For the secondary outcomes, we only conducted the PP analysis among the participants who provided fecal sample. Results Fifty-one participants completed the trial. Relative to the control group, the n-3 PUFA supplementation resulted in significant reduction in total cholesterol (TC) levels (−0.43 mmol/L, 95% CI−0.84 to−0.01 mmol/L, P < 0.05). The n-3 PUFA supplementation was also associated with significantly increased relative abundance of Bacteroidetes in phylum level (P < 0.01; false discovery rate (FDR) corrected p = 0.11), and decreased the ratio between Firmicutes and Bacteroidetes (P < 0.05; FDR corrected p = 0.16). At genus level, the intervention of plant derived n-3 PUFAs resulted in a significant decrease in relative abundance of Phascolarctobacterium (P < 0.01; FDR corrected p = 0.18) and Veillonella (P < 0.01; FDR corrected p = 0.18) after the intervention. Conclusions Our results demonstrated that plant-derived n-3 PUFAs beneficially affected the serum levels of TC and decreased the ratio between Firmicutes and Bacteroidetes during the 12-week intervention period, which might confer advantageous consequences for lipid metabolism and intestinal health.
Collapse
Affiliation(s)
- Hongjie Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yalun Zhu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can Fang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linyan Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanling Lv
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhua Mei
- Zhuhai Center for Disease Control and Prevention, Zhuhai, China
| | - Xiaolin Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Nanshan Centre for Chronic Disease Control, Shenzhen, China
| | - Jiawei Yin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jiawei Yin
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Liegang Liu
| |
Collapse
|
53
|
Little M, Dutta M, Li H, Matson A, Shi X, Mascarinas G, Molla B, Weigel K, Gu H, Mani S, Cui JY. Understanding the physiological functions of the host xenobiotic-sensing nuclear receptors PXR and CAR on the gut microbiome using genetically modified mice. Acta Pharm Sin B 2022; 12:801-820. [PMID: 35256948 PMCID: PMC8897037 DOI: 10.1016/j.apsb.2021.07.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/29/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological activation of the xenobiotic-sensing nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) is well-known to increase drug metabolism and reduce inflammation. Little is known regarding their physiological functions on the gut microbiome. In this study, we discovered bivalent hormetic functions of PXR/CAR modulating the richness of the gut microbiome using genetically engineered mice. The absence of PXR or CAR increased microbial richness, and absence of both receptors synergistically increased microbial richness. PXR and CAR deficiency increased the pro-inflammatory bacteria Helicobacteraceae and Helicobacter. Deficiency in both PXR and CAR increased the relative abundance of Lactobacillus, which has bile salt hydrolase activity, corresponding to decreased primary taurine-conjugated bile acids (BAs) in feces, which may lead to higher internal burden of taurine and unconjugated BAs, both of which are linked to inflammation, oxidative stress, and cytotoxicity. The basal effect of PXR/CAR on the gut microbiome was distinct from pharmacological and toxicological activation of these receptors. Common PXR/CAR-targeted bacteria were identified, the majority of which were suppressed by these receptors. hPXR-TG mice had a distinct microbial profile as compared to wild-type mice. This study is the first to unveil the basal functions of PXR and CAR on the gut microbiome.
Collapse
Key Words
- BA, bile acid
- BSH, bile salt hydrolase
- Bile acids
- CA, cholic acid
- CAR
- CAR, constitutive androstane receptor
- CDCA, chenodeoxycholic acid
- CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
- CV, conventional
- CYP, cytochrome P450
- DCA, deoxycholic acid
- EGF, epidermal growth factor
- Feces
- GF, germ free
- GLP-1, glucagon-like peptide-1
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- Gut microbiome
- HDCA, hyodeoxycholic acid
- IBD, inflammatory bowel disease
- IFNγ, interferon-gamma
- IL, interleukin
- IS, internal standards
- Inflammation
- LCA, lithocholic acid
- LC–MS/MS, liquid chromatography–tandem mass spectrometry
- MCA, muricholic acid
- MCP-1, monocyte chemoattractant protein-1
- Mice
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NSAID, non-steroidal anti-inflammatory drug
- Nuclear receptor
- OH, hydroxylated
- OTUs, operational taxonomy units
- PA, indole-3 propionic acid
- PBDEs, polybrominated diphenyl ethers
- PCBs, polychlorinated biphenyls
- PCoA, Principle Coordinate Analysis
- PXR
- PXR, pregnane X receptor
- PiCRUSt, Phylogenetic Investigation of Communities by Reconstruction of Observed States
- QIIME, Quantitative Insights Into Microbial Ecology
- SCFAs, short-chain fatty acids
- SNP, single-nucleotide polymorphism
- SPF, specific-pathogen-free
- T, wild type
- T-, taurine conjugated
- TCPOBOP, 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene, 3,3′,5,5′-Tetrachloro-1,4-bis(pyridyloxy)benzene
- TGR-5, Takeda G-protein-coupled receptor 5
- TLR4, toll-like receptor 4
- TNF, tumor necrosis factor
- UDCA, ursodeoxycholic acid
- YAP, yes-associated protein
- hPXR-TG, humanized PXR transgenic
Collapse
Affiliation(s)
- Mallory Little
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Hao Li
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Adam Matson
- University of Connecticut, Hartford, CT 06106, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Gabby Mascarinas
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Bruk Molla
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Kris Weigel
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Sridhar Mani
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
54
|
Chewing the Fat with Microbes: Lipid Crosstalk in the Gut. Nutrients 2022; 14:nu14030573. [PMID: 35276931 PMCID: PMC8840455 DOI: 10.3390/nu14030573] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
It is becoming increasingly important for any project aimed at understanding the effects of diet on human health, to also consider the combined effect of the trillions of microbes within the gut which modify and are modified by dietary nutrients. A healthy microbiome is diverse and contributes to host health, partly via the production and subsequent host absorption of secondary metabolites. Many of the beneficial bacteria in the gut rely on specific nutrients, such as dietary fiber, to survive and thrive. In the absence of those nutrients, the relative proportion of good commensal bacteria dwindles while communities of opportunistic, and potentially pathogenic, bacteria expand. Therefore, it is unsurprising that both diet and the gut microbiome have been associated with numerous human diseases. Inflammatory bowel diseases and colorectal cancer are associated with the presence of certain pathogenic bacteria and risk increases with consumption of a Western diet, which is typically high in fat, protein, and refined carbohydrates, but low in plant-based fibers. Indeed, despite increased screening and better care, colorectal cancer is still the 2nd leading cause of cancer death in the US and is the 3rd most diagnosed cancer among US men and women. Rates are rising worldwide as diets are becoming more westernized, alongside rising rates of metabolic diseases like obesity and diabetes. Understanding how a modern diet influences the microbiota and how subsequent microbial alterations effect human health will become essential in guiding personalized nutrition and healthcare in the future. Herein, we will summarize some of the latest advances in understanding of the three-way interaction between the human host, the gut microbiome, and the specific class of dietary nutrients, lipids.
Collapse
|
55
|
Colonic Microbiota Profile Characterization of the Responsiveness to Dietary Fibre Treatment in Hypercholesterolemia. Nutrients 2022; 14:nu14030525. [PMID: 35276884 PMCID: PMC8839280 DOI: 10.3390/nu14030525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/12/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
This study aimed to determine how the microbiota profile might be predisposed to a better response in blood lipid profiles due to dietary fibre supplementation. A three-arm intervention study that included three different fibre types (mainly insoluble, soluble, and antioxidant fibre) supplemented (19.2 g/day) during 2 months in individuals with hypercholesterolemia was developed. Changes in faecal microbiota and blood lipid profile after fibre supplementation were determined. In all volunteers, regardless of fibre type, an increase in the abundance of Bifidobacterium was observed, and similarly, an inverse relationship between faecal propionic acid and blood LDL-cholesterol, LDL particle size, and LDL/HDL particle ratio (p-values 0.0067, 0.0002, and 0.0067, respectively) was observed. However, not all volunteers presented an improvement in lipid profile. The non-responders to fibre treatment showed a decrease in microbiota diversity (Shannon and Simpson diversity index p-values of 0.0110 and 0.0255, respectively) after the intervention; where the reduction in short-chain fatty acids (SCFAs) producing bacterial genera such as Clostridium XIVa and Ruminococcus after dietary fibre treatment was the main difference. It was concluded that the non-responsiveness to dietary fibre treatment might be mediated by the lack of ability to maintain a stable SCFA producing bacteria diversity and composition after extra fibre intake.
Collapse
|
56
|
Cheng X, Zhou L, Li Z, Shen S, Zhao Y, Liu C, Zhong X, Chang Y, Kermode AG, Qiu W. Gut Microbiome and Bile Acid Metabolism Induced the Activation of CXCR5+ CD4+ T Follicular Helper Cells to Participate in Neuromyelitis Optica Spectrum Disorder Recurrence. Front Immunol 2022; 13:827865. [PMID: 35126400 PMCID: PMC8811147 DOI: 10.3389/fimmu.2022.827865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/03/2022] [Indexed: 12/29/2022] Open
Abstract
From the perspective of the role of T follicular helper (Tfh) cells in the destruction of tolerance in disease progression, more attention has been paid to their role in autoimmunity. To address the role of Tfh cells in neuromyelitis optica spectrum disorder (NMOSD) recurrence, serum C-X-C motif ligand 13 (CXCL13) levels reflect the effects of the Tfh cells on B-cell-mediated humoral immunity. We evaluated the immunobiology of the CXCR5+CD4+ Tfh cells in 46 patients with NMOSD, including 37 patients with NMOSD with an annual recurrence rate (ARR) of<1 and 9 patients with NMOSD with an ARR of ≥1. Herein, we reported several key observations. First, there was a lower frequency of circulating Tfh cells in patients with an ARR of<1 than in those with an ARR of ≥1 (P< 0.05). Second, the serum CXCL13 levels were downregulated in individuals with an ARR<1 (P< 0.05), processing the ability to promote Tfh maturation and chemotaxis. Third, the level of the primary bile acid, glycoursodeoxycholic acid (GUDCA), was higher in patients with NMOSD with an ARR of<1 than in those with NMOSD with an ARR of ≥1, which was positively correlated with CXCL13. Lastly, the frequency of the Tfh precursor cells decreased in the spleen of keyhole limpet haemocyanin-stimulated animals following GUDCA intervention. These findings significantly broaden our understanding of Tfh cells and CXCL13 in NMOSD. Our data also reveal the potential mechanism of intestinal microbiota and metabolites involved in NMOSD recurrence.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Luyao Zhou
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhibin Li
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shishi Shen
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yipeng Zhao
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunxin Liu
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaonan Zhong
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanyu Chang
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Allan G. Kermode
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Centre for Neuromuscular and Neurological Disorders, Perron Institute, The University of Western Australia, Perth, WA, Australia
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Wei Qiu
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
57
|
Haran JP, Ward DV, Bhattarai SK, Loew E, Dutta P, Higgins A, McCormick BA, Bucci V. The high prevalence of Clostridioides difficile among nursing home elders associates with a dysbiotic microbiome. Gut Microbes 2022; 13:1-15. [PMID: 33764826 PMCID: PMC8007149 DOI: 10.1080/19490976.2021.1897209] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Clostridioides difficile disproportionally affects the elderly living in nursing homes (NHs). Our objective was to explore the prevalence of C. difficile in NH elders, over time and to determine whether the microbiome or other clinical factors are associated with C. difficile colonization.We collected serial stool samples from NH residents. C. difficile prevalence was determined by quantitative polymerase-chain reaction detection of Toxin genes tcdA and tcdB; microbiome composition was determined by shotgun metagenomic sequencing. We used mixed-effect random forest modeling machine to determine bacterial taxa whose abundance is associated with C. difficile prevalence while controlling for clinical covariates including demographics, medications, and past medical history.We enrolled 167 NH elders who contributed 506 stool samples. Of the 123 elders providing multiple samples, 30 (24.4%) elders yielded multiple samples in which C. difficile was detected and 78 (46.7%) had at least one C. difficile positive sample. Elders with C. difficile positive samples were characterized by increased abundances of pathogenic or inflammatory-associated bacterial taxa and by lower abundances of taxa with anti-inflammatory or symbiotic properties. Proton pump inhibitor (PPI) use is associated with lower prevalence of C. difficile (Odds Ratio 0.46; 95%CI, 0.22-0.99) and the abundance of bacterial species with known beneficial effects was higher in PPI users and markedly lower in elders with high C. difficile prevalence.C. difficile is prevalent among NH elders and a dysbiotic gut microbiome associates with C. difficile colonization status. Manipulating the gut microbiome may prove to be a key strategy in the reduction of C. difficile in the NH.
Collapse
Affiliation(s)
- John P. Haran
- Department of Emergency Medicine, University of Massachusetts Medical School, Worcester, MA, USA,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, University of Massachusetts Medical School, Worcester, MA, USA,CONTACT John P. Haran Department of Emergency Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA01655
| | - Doyle V. Ward
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ethan Loew
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Protiva Dutta
- Department of Emergency Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Amanda Higgins
- Department of Emergency Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
58
|
Qi Q, Sun K, Rong Y, Li Z, Wu Y, Zhang D, Song S, Wang H, Feng L. Body composition of the upper limb associated with hypertension, hypercholesterolemia, and diabetes. Front Endocrinol (Lausanne) 2022; 13:985031. [PMID: 36120449 PMCID: PMC9471382 DOI: 10.3389/fendo.2022.985031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The associations between segmental body composition and metabolic diseases remain equivocal. This study aimed to investigate this association using the example of U.S. adults. This cross-sectional study included 12,148 participants from the National Health and Nutrition Examination Survey (NHANES) (2011-2018). Multivariable logistic regression models were used to estimate associations between segmental body composition quartiles of hypertension, hypercholesterolemia, and diabetes. Among 12,148 participants, 3,569, 5,683, and 1,212 had hypertension, hypercholesterolemia, and diabetes, respectively. After adjusting for potential confounders, increased percent upper limb lean body mass was associated with a lower risk of hypertension (OR= 0.88, 95%CI: 0.84, 0.92, P trend<0.001), hypercholesterolemia (OR= 0.93, 95%CI: 0.89, 0.96, P trend<0.001), and diabetes (OR= 0.96, 95%CI: 0.95, 0.98, P trend<0.001). Increased upper limb fat mass is associated with an increased risk of hypertension (OR= 1.11, 95%CI: 1.07, 1.15, P trend<0.001), hypercholesterolemia (OR= 1.05, 95%CI: 1.01, 1.09, P trend=0.07), and diabetes (OR= 1.03, 95%CI: 1.01, 1.05, P trend=0.014). The same correlations were found in the torso and whole-body composition parameters. We observed that for women, lean body mass has a better protective effect on metabolic diseases [hypertension (OR= 0.88, 95%CI: 0.82, 0.93), hypercholesteremia (OR =0.86, 95%CI: 0.81, 0.92), diabetes (OR= 0.97, 95%CI: 0.85, 0.99)]; for men, increased body fat is associated with greater risk of metabolic disease[hypertension (OR= 1.24, 95%CI: 1.15, 1.33), hypercholesteremia (OR =1.09, 95%CI: 1.01, 1.18), diabetes (OR= 1.06, 95%CI: 1.01, 1.10)]. There were significant differences between different gender. These findings suggested that upper limb and torso adiposity should be considered when assessing chronic metabolic disease risk using body composition.
Collapse
Affiliation(s)
- Qianjin Qi
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Kui Sun
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Rong
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhaoping Li
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yixia Wu
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Di Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuaihua Song
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haoran Wang
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Feng
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
59
|
Hossain MN, Senaka Ranadheera C, Fang Z, Masum A, Ajlouni S. Viability of Lactobacillus delbrueckii in chocolates during storage and in-vitro bioaccessibility of polyphenols and SCFAs. Curr Res Food Sci 2022; 5:1266-1275. [PMID: 36061408 PMCID: PMC9428806 DOI: 10.1016/j.crfs.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022] Open
Abstract
This study evaluated the viability of encapsulated Lactobacillus delbrueckii subsp. bulgaricus in chocolate during storage and in-vitro gastrointestinal transit. Flavonoid contents and short chain fatty acids (SCFAs) production during gastrointestinal transit were also assessed. Encapsulated L. delbrueckii subsp. bulgaricus survived well in chocolates >7 logs both after 120 days of storage at 4 °C and 25 °C, and during in-vitro gastrointestinal transit. The release of SCFAs through in-vitro gastrointestinal digestion and colonic fermentation revealed that probiotic-chocolates could be an excellent source of nutrients for the gut microbiota. Encapsulated probiotic in chocolates with 70% cocoa produced significantly (P < 0.05) more acetic, propionic, isobutyric, butyric and isovaleric acids than that with 45% cocoa. The bioconversion results of a specific polyphenol by L. delbrueckii subsp. bulgaricus exhibited that chocolate polyphenols could be utilized by probiotics for their metabolism. These findings confirmed that chocolate could be successfully fortified with L. delbrueckii subsp. bulgaricus encapsulation to improve health promoting properties of chocolates. Chocolates enhance the biosynthesis of SCFAs and Vit B12 in colonic fermentation. Chocolates served as a prebiotic source for gut microbiota proliferation. Chocolate with probiotics would favor the bioconversion of a specific polyphenols. Chocolates nutritional value can be enhanced via fortification with probiotics.
Collapse
|
60
|
Kang J, Li C, Gao X, Liu Z, Chen C, Luo D. Metformin inhibits tumor growth and affects intestinal flora in diabetic tumor-bearing mice. Eur J Pharmacol 2021; 912:174605. [PMID: 34757071 DOI: 10.1016/j.ejphar.2021.174605] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/31/2022]
Abstract
Many studies have found that diabetes increases the risk of some cancers such as hepatocellular carcinoma. However, there are few studies on the relationship between the two diseases and their effects on intestinal flora. Therefore, we used streptozotocin and high-fat diet to establish a mouse model of type 2 diabetes, and then inoculated the Huh-7 hepatocellular carcinoma cells to obtain mouse diabetic tumor model. Mice inoculated with Huh-7 cells alone served as control. The tumor size in the diabetic tumor group was significantly higher than that in the tumor group. Our study also showed that the expression levels of inflammation-related factors (TNFα, IL-1β, IL-6, TLR4 and MCP1) in the diabetic tumor group were significantly higher than that in the tumor group. We found that metformin alleviated blood glucose level, reduced the expressions of inflammation-related factors and retarded xenograft tumor growth in the diabetic tumor group, but it couldn't reduce the tumor growth in the tumor group. Subsequent studies found that the content of some short chain fatty acids (SCFAs) including acetic acid, propionic acid and isobutyric acid decreased significantly in diabetic tumor group. Metformin increased short chain fatty acid levels (acetic acid, butyic acid and valeric acid) and enriched the abundance of SCFA-producing bacterial genera such as Ruminococcaceae, Clostridiales, Anaerovorax, Odoribacter and Marvinbryantia. In conclusion, type 2 diabetes could promote the growth of hepatoma cells in mice. Metformin could inhibit the growth of tumor under the condition of diabetes and play a role in the intestinal homeostasis in mice.
Collapse
Affiliation(s)
- Jie Kang
- College of Life Science, Hebei University, Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, 071002, China
| | - Chunqing Li
- College of Life Science, Hebei University, Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, 071002, China
| | - Xuehui Gao
- College of Life Science, Hebei University, Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, 071002, China
| | - Zhiqin Liu
- College of Life Science, Hebei University, Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, 071002, China
| | - Chuan Chen
- College of Life Science, Hebei University, Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, 071002, China.
| | - Duqiang Luo
- College of Life Science, Hebei University, Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, 071002, China.
| |
Collapse
|
61
|
Macías-Acosta MP, Valerdi-Contreras L, Bustos-Angel ED, García-Reyes RA, Alvarez-Zavala M, González-Ávila M. Involvement of the fecal amino acid profile in a clinical and anthropometric study of Mexican patients with insulin resistance and type 2 diabetes mellitus. Amino Acids 2021; 54:47-55. [PMID: 34821993 DOI: 10.1007/s00726-021-03107-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022]
Abstract
The amino acids synthesized by the intestinal microbiota have been correlated with metabolic diseases, such as type 2 diabetes mellitus and insulin resistance; both are high incidence conditions in Mexico. However, the knowledge of the relationship of fecal amino acids with the development of both diseases in the Mexican population is scarce. The clinical study was descriptive; the study was carried out in the Antiguo Civil Hospital of Guadalajara. Samples were taken from a total of 48 participants with insulin resistance, diabetes, and a control group (n = 16 each). Anthropometric and biochemical measures were evaluated. HPLC carried out the quantification of fecal amino acids. A strong correlation between alanine and HOMA-IR (r = 0.5416) was found and between phenylalanine and HOMA-IR (r = 0.4258). Other interesting correlations were between alanine and glucose (r = 0.5854) and isoleucine and glucose (r = 0.5008). The diabetic group and the insulin-resistant group had increased fecal values of valine and isoleucine (branched-chain amino acids), which were positively correlated with the progression of both conditions. Likewise, alanine and phenylalanine can help predict the development of the disease in the Mexican population. Registry number: 037/19.
Collapse
Affiliation(s)
- Mayra Paloma Macías-Acosta
- Department Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco (CIATEJ), A.C., 44270, Guadalajara, JAL, Mexico
| | - Lorena Valerdi-Contreras
- Head of the Medical Division and Assigned to Internal Medicine Department of Antiguo Civil Hospital of Guadalajara "Fray Antonio Alcalde", 44280, Guadalajara, JAL, Mexico
| | - Ericka Denise Bustos-Angel
- Assigned to the Internal Medicine Department of Antiguo Civil Hospital of Guadalajara "Fray Antonio Alcalde", 44280, Guadalajara, JAL, Mexico
| | - Rudy Antonio García-Reyes
- Department Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco (CIATEJ), A.C., 44270, Guadalajara, JAL, Mexico
| | - Monserrat Alvarez-Zavala
- Clinical Medicine Department, University Center of Health Sciences-University of Guadalajara, 44340, Guadalajara, JAL, Mexico
| | - Marisela González-Ávila
- Department Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco (CIATEJ), A.C., 44270, Guadalajara, JAL, Mexico.
| |
Collapse
|
62
|
Oh JK, Vasquez R, Kim SH, Hwang IC, Song JH, Park JH, Kim IH, Kang DK. Multispecies probiotics alter fecal short-chain fatty acids and lactate levels in weaned pigs by modulating gut microbiota. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:1142-1158. [PMID: 34796353 PMCID: PMC8564300 DOI: 10.5187/jast.2021.e94] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 12/18/2022]
Abstract
Short-chain fatty acids (SCFAs) are metabolic products produced during the
microbial fermentation of non-digestible fibers and play an important role in
metabolic homeostasis and overall gut health. In this study, we investigated the
effects of supplementation with multispecies probiotics (MSPs) containing
Bacillus amyloliquefaciens, Limosilactobacillus
reuteri, and Levilactobacillus brevis on the gut
microbiota, and fecal SCFAs and lactate levels of weaned pigs. A total of 38
pigs weaned at 4 weeks of age were fed either a basal diet or a diet
supplemented with MSPs for 6 weeks. MSP administration significantly increased
the fecal concentrations of lactate (2.3-fold; p <
0.01), acetate (1.8-fold; p < 0.05), and formate
(1.4-fold; p < 0.05). Moreover, MSP supplementation
altered the gut microbiota of the pigs by significantly increasing the
population of potentially beneficial bacteria such as
Olsenella, Catonella,
Catenibacterium, Acidaminococcus, and
Ruminococcaceae. MSP supplementation also decreased the
abundance of pathogenic bacteria such as Escherichia and
Chlamydia. The modulation of the gut microbiota was
observed to be strongly correlated with the changes in fecal SCFAs and lactate
levels. Furthermore, we found changes in the functional pathways present within
the gut, which supports our findings that MSP modulates the gut microbiota and
SCFAs levels in pigs. The results support the potential use of MSPs to improve
the gut health of animals by modulating SCFAs production.
Collapse
Affiliation(s)
- Ju Kyoung Oh
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm 17177, Sweden
| | - Robie Vasquez
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| | - Sang Hoon Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| | - In-Chan Hwang
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| | - Ji Hoon Song
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| | - Jae Hong Park
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| | - In Ho Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
63
|
Xu N, Zhou Y, Lu X, Chang Y. Auricularia auricula-judae (Bull.) polysaccharides improve type 2 diabetes in HFD/STZ-induced mice by regulating the AKT/AMPK signaling pathways and the gut microbiota. J Food Sci 2021; 86:5479-5494. [PMID: 34787328 DOI: 10.1111/1750-3841.15963] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/01/2021] [Accepted: 10/10/2021] [Indexed: 10/19/2022]
Abstract
Auricularia auricula-judae is an edible fungus with high nutritional value due to abundant polysaccharides, and is acknowledged as traditional food and medicine in Asia. Polysaccharides from A. auricula (AAPs) are typically fungal polysaccharides and have a wide range of biological activities. It has been shown the potential of AAPs to improve diabetes as an effective adjuvant, but the underlying mechanism remains unclear. In this study, we explored the effects and potential mechanism of AAPs on type 2 diabetes (T2D) using a high-fat diet and streptozotocin (STZ) induced C57BL/6J mice. The results indicated that 50 and 100 mg/kg AAPs significantly decreased inflammation, liver injury, and insulin resistance. In addition, AAPs improved glycolipid metabolism disorders by activating the AKT and adenosine 5`monophosphate-activated protein kinase (AMPK) signaling pathways in T2D mice. Furthermore, we investigated the association between changes of gut microbiota and AAPs effects using high-throughput sequencing of 16S rDNA for fecal samples. In our study, AAPs elevated gut microbiota diversity and optimized microbial composition and function in T2D mice, characterized by increased Lactobacillus and Bacteroides abundance and decreased Clostridium and Allobaculum abundance. Particularly, AAPs intervention mainly affected the amino acid metabolism and glycolipid metabolism pathways. Overall, this study confirms that AAPs can improve type 2 diabetes by regulating the AKT and AMPK pathways and modulating intestinal microbiota. PRACTICAL APPLICATION: The article systematically verified the positive effects of AAPs on insulin resistance, glycolipid metabolism disorder, inflammation, and liver injury, key factors closely related to T2D. Furthermore, our study firstly determined the specific underlying mechanism that AAPs ameliorates T2D through regulating AKT/AMPK pathways and modifying the gut microbiota. These results could offer a full explanation and a potential option for the adjuvant therapy of diabetes with AAPs.
Collapse
Affiliation(s)
- Nuo Xu
- The State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, Xuhui District, 200237, P. R. China
| | - Yingjun Zhou
- The State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, Xuhui District, 200237, P. R. China
| | - Xinyang Lu
- The State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, Xuhui District, 200237, P. R. China
| | - Yaning Chang
- The State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, Xuhui District, 200237, P. R. China
| |
Collapse
|
64
|
Probiotic Enhancement of Antioxidant Capacity and Alterations of Gut Microbiota Composition in 6-Hydroxydopamin-Induced Parkinson's Disease Rats. Antioxidants (Basel) 2021; 10:antiox10111823. [PMID: 34829694 PMCID: PMC8615185 DOI: 10.3390/antiox10111823] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 01/15/2023] Open
Abstract
Oxidative stress plays a key role in the degeneration of dopaminergic neurons in Parkinson's disease (PD), which may be aggravated by concomitant PD-associated gut dysbiosis. Probiotics and prebiotics are therapeutically relevant to these conditions due to their antioxidant, anti-inflammatory, and gut microbiome modulation properties. However, the mechanisms by which probiotic/prebiotic supplementation affects antioxidant capacity and the gut microbiome in PD remains poorly characterized. In this study, we assessed the effects of a Lactobacillus salivarius AP-32 probiotic, a prebiotic (dried AP-32 culture medium supernatant), and a probiotic/prebiotic cocktail in rats with unilateral 6-hydroxydopamine (6-OHDA)-induced PD. The neuroprotective effects and levels of oxidative stress were evaluated after eight weeks of daily supplementation. Fecal microbiota composition was analyzed by fecal 16S rRNA gene sequencing. The supplements were associated with direct increases in host antioxidant enzyme activities and short-chain fatty acid production, protected dopaminergic neurons, and improved motor functions. The supplements also altered the fecal microbiota composition, and some specifically enriched commensal taxa correlated positively with superoxide dismutase, glutathione peroxidase, and catalase activity, indicating supplementation also promotes antioxidant activity via an indirect pathway. Therefore, L. salivarius AP-32 supplementation enhanced the activity of host antioxidant enzymes via direct and indirect modes of action in rats with 6-OHDA-induced PD.
Collapse
|
65
|
Evidence of MHC class I and II influencing viral and helminth infection via the microbiome in a non-human primate. PLoS Pathog 2021; 17:e1009675. [PMID: 34748618 PMCID: PMC8601626 DOI: 10.1371/journal.ppat.1009675] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/18/2021] [Accepted: 10/05/2021] [Indexed: 01/04/2023] Open
Abstract
Until recently, the study of major histocompability complex (MHC) mediated immunity has focused on the direct link between MHC diversity and susceptibility to parasite infection. However, MHC genes can also influence host health indirectly through the sculpting of the bacterial community that in turn shape immune responses. We investigated the links between MHC class I and II gene diversity gut microbiome diversity and micro- (adenovirus, AdV) and macro- (helminth) parasite infection probabilities in a wild population of non-human primates, mouse lemurs of Madagascar. This setup encompasses a plethora of underlying interactions between parasites, microbes and adaptive immunity in natural populations. Both MHC classes explained shifts in microbiome composition and the effect was driven by a few select microbial taxa. Among them were three taxa (Odoribacter, Campylobacter and Prevotellaceae-UCG-001) which were in turn linked to AdV and helminth infection status, correlative evidence of the indirect effect of the MHC via the microbiome. Our study provides support for the coupled role of MHC diversity and microbial flora as contributing factors of parasite infection. The selective pressure of the major histocompatibility complex (MHC) on microbial communities, and the potential role of this interaction in driving parasite resistance has been largely neglected. Using a natural population of the primate Microcebus griseorufus, we provide correlative evidence of two outstanding findings: that MHCI and MHCII diversity shapes the composition of the gut microbiota; and that select taxa associated with MHC diversity predicted adenovirus and helminth infection status. Our study highlights the importance of incorporating the microbiome when investigating parasite-mediated MHC selection.
Collapse
|
66
|
Urinary Metabolomic Profile of Neonates Born to Women with Gestational Diabetes Mellitus. Metabolites 2021; 11:metabo11110723. [PMID: 34822382 PMCID: PMC8621167 DOI: 10.3390/metabo11110723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most frequent pregnancy complications with potential adverse outcomes for mothers and newborns. Its effects on the newborn appear during the neonatal period or early childhood. Therefore, an early diagnosis is crucial to prevent the development of chronic diseases later in adult life. In this study, the urinary metabolome of babies born to GDM mothers was characterized. In total, 144 neonatal and maternal (second and third trimesters of pregnancy) urinary samples were analyzed using targeted metabolomics, combining liquid chromatographic mass spectrometry (LC-MS/MS) and flow injection analysis mass spectrometry (FIA-MS/MS) techniques. We provide here the neonatal urinary concentration values of 101 metabolites for 26 newborns born to GDM mothers and 22 newborns born to healthy mothers. The univariate analysis of these metabolites revealed statistical differences in 11 metabolites. Multivariate analyses revealed a differential metabolic profile in newborns of GDM mothers characterized by dysregulation of acylcarnitines, amino acids, and polyamine metabolism. Levels of hexadecenoylcarnitine (C16:1) and spermine were also higher in newborns of GDM mothers. The maternal urinary metabolome revealed significant differences in butyric, isobutyric, and uric acid in the second and third trimesters of pregnancy. These metabolic alterations point to the impact of GDM in the neonatal period.
Collapse
|
67
|
de Maria YNLF, Aciole Barbosa D, Menegidio FB, Santos KBNH, Humberto AC, Alencar VC, Silva JFS, Costa de Oliveira R, Batista ML, Nunes LR, Jabes DL. Analysis of mouse faecal dysbiosis, during the development of cachexia, induced by transplantation with Lewis lung carcinoma cells. MICROBIOLOGY (READING, ENGLAND) 2021; 167. [PMID: 34596506 DOI: 10.1099/mic.0.001088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cachexia (CC) is a complex wasting syndrome that significantly affects life quality and life expectancy among cancer patients. Original studies, in which CC was induced in mouse models through inoculation with BaF and C26 tumour cells, demonstrated that CC development correlates with bacterial gut dysbiosis in these animals. In both cases, a common microbial signature was observed, based on the expansion of Enterobacteriaceae in the gut of CC animals. However, these two types of tumours induce unique microbial profiles, suggesting that different CC induction mechanisms significantly impact the outcome of gut dysbiosis. The present study sought to expand the scope of such analyses by characterizing the CC-associated dysbiosis that develops when mice are inoculated with Lewis lung carcinoma (LLC) cells, which constitutes one of the most widely employed mechanisms for CC induction. Interestingly, Enterobacteriaceae expansion is also observed in LLC-induced CC. However, the dysbiosis identified herein displays a more complex pattern, involving representatives from seven different bacterial phyla, which were consistently identified across successive levels of taxonomic hierarchy. These results are supported by a predictive analysis of gene content, which identified a series of functional/structural changes that potentially occur in the gut bacterial population of these animals, providing a complementary and alternative approach to microbiome analyses based solely on taxonomic classification.
Collapse
Affiliation(s)
- Yara N L F de Maria
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| | - David Aciole Barbosa
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| | - Fabiano B Menegidio
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| | | | | | - Valquíria C Alencar
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Brazil
| | - Juliana F S Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Brazil
| | | | - Miguel L Batista
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
- Department of Biochemistry, Boston University School of Medicine, USA
| | - Luiz R Nunes
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Brazil
| | - Daniela L Jabes
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| |
Collapse
|
68
|
de Luna Freire MO, do Nascimento LCP, de Oliveira KÁR, de Oliveira AM, Napoleão TH, Lima MDS, Lagranha CJ, de Souza EL, de Brito Alves JL. Effects of a Mixed Limosilactobacillus fermentum Formulation with Claimed Probiotic Properties on Cardiometabolic Variables, Biomarkers of Inflammation and Oxidative Stress in Male Rats Fed a High-Fat Diet. Foods 2021; 10:foods10092202. [PMID: 34574313 PMCID: PMC8471400 DOI: 10.3390/foods10092202] [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: 06/29/2021] [Revised: 07/31/2021] [Accepted: 08/19/2021] [Indexed: 12/20/2022] Open
Abstract
High-fat diet (HFD) consumption has been linked to dyslipidemia, low-grade inflammation and oxidative stress. This study investigated the effects of a mixed formulation with Limosilactobacillusfermentum 139, L. fermentum 263 and L. fermentum 296 on cardiometabolic parameters, fecal short-chain fatty acid (SCFA) contents and biomarkers of inflammation and oxidative stress in colon and heart tissues of male rats fed an HFD. Male Wistar rats were grouped into control diet (CTL, n = 6), HFD (n = 6) and HFD with L. fermentum formulation (HFD-Lf, n = 6) groups. The L.fermentum formulation (1 × 109 CFU/mL of each strain) was administered twice a day for 4 weeks. After a 4-week follow-up, biochemical parameters, fecal SCFA, cytokines and oxidative stress variables were evaluated. HFD consumption caused hyperlipidemia, hyperglycemia, low-grade inflammation, reduced fecal acetate and propionate contents and increased biomarkers of oxidative stress in colon and heart tissues when compared to the CTL group. Rats receiving the L. fermentum formulation had reduced hyperlipidemia and hyperglycemia, but similar SCFA contents in comparison with the HFD group (p < 0.05). Rats receiving the L. fermentum formulation had increased antioxidant capacity throughout the colon and heart tissues when compared with the control group. Administration of a mixed L. fermentum formulation prevented hyperlipidemia, inflammation and oxidative stress in colon and heart tissues induced by HFD consumption.
Collapse
Affiliation(s)
| | - Luciana Caroline Paulino do Nascimento
- Health Sciences Center, Department of Nutrition, Federal University of Paraiba, João Pessoa 58051900, PB, Brazil; (L.C.P.d.N.); (K.Á.R.d.O.); (E.L.d.S.)
| | - Kataryne Árabe Rimá de Oliveira
- Health Sciences Center, Department of Nutrition, Federal University of Paraiba, João Pessoa 58051900, PB, Brazil; (L.C.P.d.N.); (K.Á.R.d.O.); (E.L.d.S.)
| | - Alisson Macário de Oliveira
- Biological Sciences Center, Department of Biochemistry, Federal University of Pernambuco, Recife 50670901, PE, Brazil; (A.M.d.O.); (T.H.N.)
| | - Thiago Henrique Napoleão
- Biological Sciences Center, Department of Biochemistry, Federal University of Pernambuco, Recife 50670901, PE, Brazil; (A.M.d.O.); (T.H.N.)
| | - Marcos dos Santos Lima
- Department of Food Technology, Federal Institute of Sertão Pernambucano, Petrolina 56302100, PE, Brazil;
| | - Cláudia Jacques Lagranha
- Laboratory of Biochemistry and Exercise Biochemistry, Federal University of Pernambuco, Vitória de Santo Antão 55608680, PE, Brazil;
| | - Evandro Leite de Souza
- Health Sciences Center, Department of Nutrition, Federal University of Paraiba, João Pessoa 58051900, PB, Brazil; (L.C.P.d.N.); (K.Á.R.d.O.); (E.L.d.S.)
| | - José Luiz de Brito Alves
- Health Sciences Center, Department of Nutrition, Federal University of Paraiba, João Pessoa 58051900, PB, Brazil; (L.C.P.d.N.); (K.Á.R.d.O.); (E.L.d.S.)
- Correspondence: or ; Tel./Fax: +55-81998495485
| |
Collapse
|
69
|
Del Chierico F, Manco M, Gardini S, Guarrasi V, Russo A, Bianchi M, Tortosa V, Quagliariello A, Shashaj B, Fintini D, Putignani L. Fecal microbiota signatures of insulin resistance, inflammation, and metabolic syndrome in youth with obesity: a pilot study. Acta Diabetol 2021; 58:1009-1022. [PMID: 33754165 DOI: 10.1007/s00592-020-01669-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022]
Abstract
AIMS To identify fecal microbiota profiles associated with metabolic abnormalities belonging to the metabolic syndrome (MS), high count of white blood cells (WBCs) and insulin resistance (IR). METHODS Sixty-eight young patients with obesity were stratified for percentile distribution of MS abnormalities. A MS risk score was defined as low, medium, and high MS risk. High WBCs were defined as a count ≥ 7.0 103/µL; severe obesity as body mass index Z-score ≥ 2 standard deviations; IR as homeostatic assessment model algorithm of IR (HOMA) ≥ 3.7. Stool samples were analyzed by 16S rRNA-based metagenomics. RESULTS We found reduced bacterial richness of fecal microbiota in patients with IR and high diastolic blood pressure (BP). Distinct microbial markers were associated to high BP (Clostridium and Clostridiaceae), low high-density lipoprotein cholesterol (Lachnospiraceae, Gemellaceae, Turicibacter), and high MS risk (Coriobacteriaceae), WBCs (Bacteroides caccae, Gemellaceae), severe obesity (Lachnospiraceae), and impaired glucose tolerance (Bacteroides ovatus and Enterobacteriaceae). Conversely, taxa such as Faecalibacterium prausnitzii, Parabacterodes, Bacteroides caccae, Oscillospira, Parabacterodes distasonis, Coprococcus, and Haemophilus parainfluenzae were associated to low MS risk score, triglycerides, fasting glucose and HOMA-IR, respectively. Supervised multilevel analysis grouped clearly "variable" patients based on the MS risk. CONCLUSIONS This was a proof-of-concept study opening the way at the identification of fecal microbiota signatures, precisely associated with cardiometabolic risk factors in young patients with obesity. These evidences led us to infer, while some gut bacteria have a detrimental role in exacerbating metabolic risk factors some others are beneficial ameliorating cardiovascular host health.
Collapse
Affiliation(s)
| | - Melania Manco
- Research Area for Multifactorial Diseases and Complex Phenotypes, Obesity and Diabetes, Bambino Gesù Children's Hospital, IRCCS, Via Ferdinando Baldelli 38, 00146, Rome, Italy.
| | | | - Valerio Guarrasi
- GenomeUp SRL, Rome, Italy
- Department of Computer, Control, and Management Engineering Antonio Ruberti, Sapienza University, Rome, Italy
| | - Alessandra Russo
- Unit of Parasitology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marzia Bianchi
- Research Area for Multifactorial Diseases and Complex Phenotypes, Obesity and Diabetes, Bambino Gesù Children's Hospital, IRCCS, Via Ferdinando Baldelli 38, 00146, Rome, Italy
| | - Valentina Tortosa
- Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Blegina Shashaj
- Research Area for Multifactorial Diseases and Complex Phenotypes, Obesity and Diabetes, Bambino Gesù Children's Hospital, IRCCS, Via Ferdinando Baldelli 38, 00146, Rome, Italy
| | - Danilo Fintini
- Endocrinology Unit, Bambino Gesù Children's Hospital, IRCCS, Palidoro, Rome, Italy
| | - Lorenza Putignani
- Unit of Parasitology and Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
70
|
Polysaccharide Structures and Their Hypocholesterolemic Potential. Molecules 2021; 26:molecules26154559. [PMID: 34361718 PMCID: PMC8348680 DOI: 10.3390/molecules26154559] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Several classes of polysaccharides have been described to have hypocholesterolemic potential, namely cholesterol bioaccessibility and bioavailability. This review will highlight the main mechanisms by which polysaccharides are known to affect cholesterol homeostasis at the intestine, namely the effect (i) of polysaccharide viscosity and its influence on cholesterol bioaccessibility; (ii) on bile salt sequestration and its dependence on the structural diversity of polysaccharides; (iii) of bio-transformations of polysaccharides and bile salts by the gut microbiota. Different quantitative structure–hypocholesterolemic activity relationships have been explored depending on the mechanism involved, and these were based on polysaccharide physicochemical properties, such as sugar composition and ramification degree, linkage type, size/molecular weight, and charge. The information gathered will support the rationalization of polysaccharides’ effect on cholesterol homeostasis and highlight predictive rules towards the development of customized hypocholesterolemic functional food.
Collapse
|
71
|
Winther SA, Mannerla MM, Frimodt-Møller M, Persson F, Hansen TW, Lehto M, Hörkkö S, Blaut M, Forsblom C, Groop PH, Rossing P. Faecal biomarkers in type 1 diabetes with and without diabetic nephropathy. Sci Rep 2021; 11:15208. [PMID: 34312454 PMCID: PMC8313679 DOI: 10.1038/s41598-021-94747-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal dysbiosis is common among persons with type 1 diabetes (T1D), but its potential impact on diabetic nephropathy (DN) remains obscure. We examined whether faecal biomarkers, previously associated with low-grade gastrointestinal inflammation, differ between healthy controls and T1D subjects with and without DN. Faecal samples were analyzed for levels of calprotectin, intestinal alkaline phosphatase (IAP), short-chain fatty acids (SCFA) and immunoglobulins in subjects with T1D (n = 159) and healthy controls (NDC; n = 50). The subjects with T1D were stratified based on albuminuria: normoalbuminuria (< 30 mg/g; n = 49), microalbuminuria (30-299 mg/g; n = 50) and macroalbuminuria (≥ 300 mg/g; n = 60). aecal calprotectin, IAP and immunoglobulin levels did not differ between the T1D albuminuria groups. However, when subjects were stratified based on faecal calprotectin cut-off level (50 µg/g), macroalbuminuric T1D subjects exceeded the threshold more frequently than NDC (p = 0.02). Concentrations of faecal propionate and butyrate were lower in T1D subjects compared with NDC (p = 0.04 and p = 0.03, respectively). Among T1D subjects, levels of branched SCFA (BCFA) correlated positively with current albuminuria level (isobutyrate, p = 0.03; isovalerate, p = 0.005). In our study cohort, fatty acid metabolism seemed to be altered among T1D subjects and those with albuminuria compared to NDC. This may reflect gastrointestinal imbalances associated with T1D and renal complications.
Collapse
Affiliation(s)
- Signe Abitz Winther
- Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, 2820, Gentofte, Denmark
- Novo Nordisk A/S, Måløv, Denmark
| | - Miia Maininki Mannerla
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marie Frimodt-Møller
- Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, 2820, Gentofte, Denmark
| | - Frederik Persson
- Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, 2820, Gentofte, Denmark
| | - Tine Willum Hansen
- Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, 2820, Gentofte, Denmark
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Unit of Biomedicine, University of Oulu, Oulu, Finland
- Medical Research Center, Nordlab Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Michael Blaut
- Department of Gastrointestinal Microbiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, 2820, Gentofte, Denmark.
- University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
72
|
Wastyk HC, Fragiadakis GK, Perelman D, Dahan D, Merrill BD, Yu FB, Topf M, Gonzalez CG, Van Treuren W, Han S, Robinson JL, Elias JE, Sonnenburg ED, Gardner CD, Sonnenburg JL. Gut-microbiota-targeted diets modulate human immune status. Cell 2021; 184:4137-4153.e14. [PMID: 34256014 DOI: 10.1016/j.cell.2021.06.019] [Citation(s) in RCA: 487] [Impact Index Per Article: 162.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/13/2021] [Accepted: 06/11/2021] [Indexed: 12/21/2022]
Abstract
Diet modulates the gut microbiome, which in turn can impact the immune system. Here, we determined how two microbiota-targeted dietary interventions, plant-based fiber and fermented foods, influence the human microbiome and immune system in healthy adults. Using a 17-week randomized, prospective study (n = 18/arm) combined with -omics measurements of microbiome and host, including extensive immune profiling, we found diet-specific effects. The high-fiber diet increased microbiome-encoded glycan-degrading carbohydrate active enzymes (CAZymes) despite stable microbial community diversity. Although cytokine response score (primary outcome) was unchanged, three distinct immunological trajectories in high-fiber consumers corresponded to baseline microbiota diversity. Alternatively, the high-fermented-food diet steadily increased microbiota diversity and decreased inflammatory markers. The data highlight how coupling dietary interventions to deep and longitudinal immune and microbiome profiling can provide individualized and population-wide insight. Fermented foods may be valuable in countering the decreased microbiome diversity and increased inflammation pervasive in industrialized society.
Collapse
Affiliation(s)
- Hannah C Wastyk
- Department of Bioengineering, Stanford School of Medicine, Stanford, CA 94305, USA
| | | | - Dalia Perelman
- Stanford Prevention Research Center, Department of Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dylan Dahan
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Bryan D Merrill
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Feiqiao B Yu
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Madeline Topf
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Carlos G Gonzalez
- Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - William Van Treuren
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Shuo Han
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jennifer L Robinson
- Stanford Prevention Research Center, Department of Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | | | - Erica D Sonnenburg
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA; Center for Human Microbiome Studies, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA.
| | - Christopher D Gardner
- Stanford Prevention Research Center, Department of Medicine, Stanford School of Medicine, Stanford, CA 94305, USA.
| | - Justin L Sonnenburg
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA; Center for Human Microbiome Studies, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
73
|
Disease Differentiation and Monitoring of Anti-TNF Treatment in Rheumatoid Arthritis and Spondyloarthropathies. Int J Mol Sci 2021; 22:ijms22147389. [PMID: 34299006 PMCID: PMC8307996 DOI: 10.3390/ijms22147389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 01/16/2023] Open
Abstract
Rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) are comprehensive immunological disorders. The treatment of these disorders is limited to ameliorating the symptoms and improving the quality of life of patients. In this study, serum samples from RA, AS, and PsA patients were analyzed with metabolomic tools employing the 1H NMR method in combination with univariate and multivariate analyses. The results obtained in this study showed that the changes in metabolites were the highest for AS > RA > PsA. The study demonstrated that the time until remission or until low disease activity is achieved is shortest (approximately three months) for AS, longer for RA and longest for PsA. The statistically common metabolite that was found to be negatively correlated with the healing processes of these disorders is ethanol, which may indicate the involvement of the gut microflora and/or the breakdown of malondialdehyde as a cell membrane lipid peroxide product.
Collapse
|
74
|
Stothart MR, Newman AEM. Shades of grey: host phenotype dependent effect of urbanization on the bacterial microbiome of a wild mammal. Anim Microbiome 2021; 3:46. [PMID: 34225812 PMCID: PMC8256534 DOI: 10.1186/s42523-021-00105-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/31/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Host-associated microbiota are integral to the ecology of their host and may help wildlife species cope with rapid environmental change. Urbanization is a globally replicated form of severe environmental change which we can leverage to better understand wildlife microbiomes. Does the colonization of separate cities result in parallel changes in the intestinal microbiome of wildlife, and if so, does within-city habitat heterogeneity matter? Using 16S rRNA gene amplicon sequencing, we quantified the effect of urbanization (across three cities) on the microbiome of eastern grey squirrels (Sciurus carolinensis). Grey squirrels are ubiquitous in rural and urban environments throughout their native range, across which they display an apparent coat colour polymorphism (agouti, black, intermediate). RESULTS Grey squirrel microbiomes differed between rural and city environments; however, comparable variation was explained by habitat heterogeneity within cities. Our analyses suggest that operational taxonomic unit (OTU) community structure was more strongly influenced by local environmental conditions (rural and city forests versus human built habitats) than urbanization of the broader landscape (city versus rural). The bacterial genera characterizing the microbiomes of built-environment squirrels are thought to specialize on host-derived products and have been linked in previous research to low fibre diets. However, despite an effect of urbanization at fine spatial scales, phylogenetic patterns in the microbiome were coat colour phenotype dependent. City and built-environment agouti squirrels displayed greater phylogenetic beta-dispersion than those in rural or forest environments, and null modelling results indicated that the phylogenetic structure of urban agouti squirrels did not differ greatly from stochastic expectations. CONCLUSIONS Squirrel microbiomes differed between city and rural environments, but differences of comparable magnitude were observed between land classes at a within-city scale. We did not observe strong evidence that inter-environmental differences were the result of disparate selective pressures. Rather, our results suggest that microbiota dispersal and ecological drift are integral to shaping the inter-environmental differences we observed. However, these processes were partly mediated by squirrel coat colour phenotype. Given a well-known urban cline in squirrel coat colour melanism, grey squirrels provide a useful free-living system with which to study how host genetics mediate environment x microbiome interactions.
Collapse
Affiliation(s)
- Mason R. Stothart
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, T2N 4Z6 Canada
| | - Amy E. M. Newman
- Department of Integrative Biology, College of Biological Sciences, University of Guelph, Guelph, N1G 2W1 Canada
| |
Collapse
|
75
|
Fan X, Liu B, Zhou J, Gu X, Zhou Y, Yang Y, Guo F, Wei X, Wang H, Si N, Yang J, Bian B, Zhao H. High-Fat Diet Alleviates Neuroinflammation and Metabolic Disorders of APP/PS1 Mice and the Intervention With Chinese Medicine. Front Aging Neurosci 2021; 13:658376. [PMID: 34168550 PMCID: PMC8217439 DOI: 10.3389/fnagi.2021.658376] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease caused by the complex interaction of multiple mechanisms. Recent studies examining the effect of high-fat diet (HFD) on the AD phenotype have demonstrated a significant influence on both inflammation and cognition. However, different studies on the effect of high-fat diet on AD pathology have reported conflicting conclusions. To explore the involvement of HFD in AD, we investigated phenotypic and metabolic changes in an AD mouse model in response to HFD. The results indicated there was no significant effect on Aβ levels or contextual memory due to HFD treatment. Of note, HFD did moderate neuroinflammation, despite spurring inflammation and increasing cholesterol levels in the periphery. In addition, diet affected gut microbiota symbiosis, altering the production of bacterial metabolites. HFD created a favorable microenvironment for bile acid alteration and arachidonic acid metabolism in APP/PS1 mice, which may be related to the observed improvement in LXR/PPAR expression. Our previous research demonstrated that Huanglian Jiedu decoction (HLJDD) significantly ameliorated impaired learning and memory. Furthermore, HLJDD may globally suppress inflammation and lipid accumulation to relieve cognitive impairment after HFD intervention. It was difficult to define the effect of HFD on AD progression because the results were influenced by confounding factors and biases. Although there was still obvious damage in AD mice treated with HFD, there was no deterioration and there was even a slight remission of neuroinflammation. Moreover, HLJDD represents a potential AD drug based on its anti-inflammatory and lipid-lowering effects.
Collapse
Affiliation(s)
- Xiaorui Fan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Junyi Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinru Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yifei Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
76
|
Feeding diversified protein sources exacerbates hepatic insulin resistance via increased gut microbial branched-chain fatty acids and mTORC1 signaling in obese mice. Nat Commun 2021; 12:3377. [PMID: 34099716 PMCID: PMC8184893 DOI: 10.1038/s41467-021-23782-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Animal models of human diseases are classically fed purified diets that contain casein as the unique protein source. We show that provision of a mixed protein source mirroring that found in the western diet exacerbates diet-induced obesity and insulin resistance by potentiating hepatic mTORC1/S6K1 signaling as compared to casein alone. These effects involve alterations in gut microbiota as shown by fecal microbiota transplantation studies. The detrimental impact of the mixed protein source is also linked with early changes in microbial production of branched-chain fatty acids (BCFA) and elevated plasma and hepatic acylcarnitines, indicative of aberrant mitochondrial fatty acid oxidation. We further show that the BCFA, isobutyric and isovaleric acid, increase glucose production and activate mTORC1/S6K1 in hepatocytes. Our findings demonstrate that alteration of dietary protein source exerts a rapid and robust impact on gut microbiota and BCFA with significant consequences for the development of obesity and insulin resistance.
Collapse
|
77
|
Yang C, Peng C, Jin H, You L, Wang J, Xu H, Sun Z. Comparison of the composition and function of the gut microbiome in herdsmen from two pasture regions, Hongyuan and Xilingol. Food Sci Nutr 2021; 9:3258-3268. [PMID: 34136190 PMCID: PMC8194741 DOI: 10.1002/fsn3.2290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
There is a close relationship between the gut microbiome and health in humans including regulation of immunity and energy metabolism. This study investigated differences in the gut microbiome of herdsmen from two regions: Hongyuan pasture in Sichuan and Xilingol pasture in Inner Mongolia. We found significant differences in the gut microbiome between the two groups. The main discriminatory species between the two groups were Bifidobacterium longum, Bifidobacterium breve, Phascolarctobacterium succinatutens, Prevotella stercorea, Prevotella copri, Eubacterium biforme, and Fusobacterium prausnitzii. The abundances of Bifidobacterium longum and Bifidobacterium breve were significantly lower in the gut microbiomes of Hongyuan herdsmen than in the gut microbiomes of Xilingol herdsmen. Functional metagenomic analysis showed that more genes were enriched in glycoside hydrolase and transposase in the gut microbiome of Hongyuan herdsmen compared with Xilingol herdsmen, suggesting a higher energy demand in the gut microbiome of Hongyuan herdsmen. Significantly more genes associated with glycolysis, starch degradation, and sucrose degradation were also found in the gut microbiome of Hong yuan herdsmen compared with Xilingol herdsmen. These results indicate that herdsmen from different pastoral regions had distinct gut microbiome composition and functions.
Collapse
Affiliation(s)
- Chengcong Yang
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
| | - Chuantao Peng
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
- Qingdao Special Food Research InstituteQingdao Agricultural UniversityQingdaoChina
| | - Hao Jin
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
| | - Lijun You
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
| | - Jiao Wang
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
| | - Haiyan Xu
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and EngineeringKey Laboratory of Dairy Products ProcessingInner Mongolia Agricultural UniversityHohhotChina
- Key Laboratory of Dairy Products ProcessingMinistry of Agriculture and Rural AffairsInner Mongolia Agricultural UniversityInner MongoliaChina
| |
Collapse
|
78
|
Daniel N, Rossi Perazza L, Varin TV, Trottier J, Marcotte B, St-Pierre P, Barbier O, Chassaing B, Marette A. Dietary fat and low fiber in purified diets differently impact the gut-liver axis to promote obesity-linked metabolic impairments. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1014-G1033. [PMID: 33881354 DOI: 10.1152/ajpgi.00028.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Selecting the most relevant control diet is of critical importance for metabolic and intestinal studies in animal models. Chow and LF-purified diet differentially impact metabolic and gut microbiome outcomes resulting in major changes in intestinal integrity in LF-fed animals which contributes to altering metabolic homeostasis. Dietary fat and low fiber both contribute to the deleterious metabolic effect of purified HF diets through both selective and overlapping mechanisms.
Collapse
Affiliation(s)
- Noëmie Daniel
- Faculty of Food Science, Laval University, Québec City, Québec, Canada.,Cardiology axis of the Québec Heart and Lung Institute Research Center, Québec City, Québec, Canada.,Institute of Nutrition and Functional Foods (INAF), Laval University, Québec City, Québec, Canada
| | - Laίs Rossi Perazza
- Faculty of Medicine, Laval University, Québec City, Québec, Canada.,Cardiology axis of the Québec Heart and Lung Institute Research Center, Québec City, Québec, Canada.,Institute of Nutrition and Functional Foods (INAF), Laval University, Québec City, Québec, Canada
| | - Thibault V Varin
- Institute of Nutrition and Functional Foods (INAF), Laval University, Québec City, Québec, Canada
| | - Jocelyn Trottier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Center, and Faculty of Pharmacy, Laval University, Québec City, Québec, Canada
| | - Bruno Marcotte
- Cardiology axis of the Québec Heart and Lung Institute Research Center, Québec City, Québec, Canada.,Institute of Nutrition and Functional Foods (INAF), Laval University, Québec City, Québec, Canada
| | - Philippe St-Pierre
- Cardiology axis of the Québec Heart and Lung Institute Research Center, Québec City, Québec, Canada.,Institute of Nutrition and Functional Foods (INAF), Laval University, Québec City, Québec, Canada
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Center, and Faculty of Pharmacy, Laval University, Québec City, Québec, Canada
| | - Benoit Chassaing
- INSERM U1016, team "Mucosal microbiota in chronic inflammatory diseases," CNRS UMR 8104, Université de Paris, Paris, France
| | - André Marette
- Faculty of Medicine, Laval University, Québec City, Québec, Canada.,Cardiology axis of the Québec Heart and Lung Institute Research Center, Québec City, Québec, Canada.,Institute of Nutrition and Functional Foods (INAF), Laval University, Québec City, Québec, Canada
| |
Collapse
|
79
|
Implications of SCFAs on the Parameters of the Lipid and Hepatic Profile in Pregnant Women. Nutrients 2021; 13:nu13061749. [PMID: 34063900 PMCID: PMC8224042 DOI: 10.3390/nu13061749] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/27/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are the product of the anaerobic intestinal bacterial fermentation of dietary fiber and resistant starch. An abnormal intestinal microbiota may cause a reduction in the production of SCFAs, which stimulate the development of intestinal epithelial cells, nourish enterocytes, influence their maturation and proper differentiation, reduce the pH, and are an additional source of energy for the host. There have been reports of the special role of SCFAs in the regulation of glucose and lipid metabolism during pregnancy. AIM The aim of the study was to analyze the correlation of SCFAs with lipid and hepatic metabolism during pregnancy in relation to the body weight of pregnant women. MATERIAL AND METHODS This study was conducted in pregnant women divided into two groups: Obese (OW-overweight and obese women; n = 48) and lean (CG-control group; n = 48) individuals. The biochemical plasma parameters of lipid metabolism (TG, CH, LDL, HDL), inflammation (CRP), and liver function (ALT, AST, GGT) were determined in all of the subjects. SCFA analysis was performed in the stool samples to measure acetic acid (C 2:0), propionic acid (C 3:0), isobutyric acid (C 4:0 i), butyric acid (C 4:0 n), isovaleric acid (C 5:0 i) valeric acid (C 5:0 n), isocaproic acid (C 6:0 i), caproic acid (C 6:0 n), and heptanoic acid (C 7:0). RESULTS Statistically significant differences in the concentrations of C 3:0 and C 6:0 n were found between women in the OW group compared to the CG group. The other SCFAs tested did not differ significantly depending on BMI. The C 2:0, C 3:0, and C 4:0 n ratios showed differences in both OW and CG groups. In the OW group, no relationship was observed between the concentrations of the SCFAs tested and CRP, ALT, AST. A surprising positive relationship between C 5:0 n and all fractions of the tested lipids and branched C 5:0 with CHL, HDL, and LDL was demonstrated. In the OW group, HDL showed a positive correlation with C 3:0. However, lower GGT concentrations were accompanied by higher C 4:0 and C 5:0 values, and this tendency was statistically significant. CONCLUSIONS The results of our research show that some SCFAs are associated with hepatic lipid metabolism and CRP concentrations, which may vary with gestational weight. Obesity in pregnancy reduces the amount of SCFAs in the stool, and a decrease in the level of butyrate reduces liver function.
Collapse
|
80
|
Effect of Lactobacillus paracasei HII01 Supplementation on Total Cholesterol, and on the Parameters of Lipid and Carbohydrate Metabolism, Oxidative Stress, Inflammation and Digestion in Thai Hypercholesterolemic Subjects. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11104333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hypercholesterolemia is one of the leading causes of cardiovascular disease. Probiotics can help to improve high blood lipid levels in hypercholesterolemia patients. Lactobacillus paracasei has been reported to have beneficial effects in several subjects; however, there is a lack of studies on Thai hypercholesterolemic subjects. Thus, this study was conducted in order to investigate the effect of L. paracasei HII01 on cholesterol, oxidative stress, and other biomarkers. Fifty-two subjects were randomized into two groups: the L. paracasei treatment group and the placebo group. The study was conducted over an intervention period of 12 weeks of supplementation. The results show that L. paracasei HII01 significantly reduced the total cholesterol (TCH), triglycerides (TGs), tumor necrosis factor (TNF)-α and lipopolysaccharide (LPS) of the patients, and increased their HDL, total antioxidant capacity (TAC) and propionic acid compared to the placebo group. Moreover, the supplementation of L. paracasei HII01 significantly increased lactic acid, IL-10 and IFN-γ, and substantially decreased malondialdehyde (MDA) at the end of the treatment. The results suggest that L. paracasei HII01 improves the blood lipid profile, reduces oxidative stress, and is beneficial for health among Thai hypercholesterolemic subjects.
Collapse
|
81
|
Nurrahma BA, Tsao SP, Wu CH, Yeh TH, Hsieh PS, Panunggal B, Huang HY. Probiotic Supplementation Facilitates Recovery of 6-OHDA-Induced Motor Deficit via Improving Mitochondrial Function and Energy Metabolism. Front Aging Neurosci 2021; 13:668775. [PMID: 34025392 PMCID: PMC8137830 DOI: 10.3389/fnagi.2021.668775] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease associated with progressive impairment of motor and non-motor functions in aging people. Overwhelming evidence indicate that mitochondrial dysfunction is a central factor in PD pathophysiology, which impairs energy metabolism. While, several other studies have shown probiotic supplementations to improve host energy metabolism, alleviate the disease progression, prevent gut microbiota dysbiosis and alter commensal bacterial metabolites. But, whether probiotic and/or prebiotic supplementation can affect energy metabolism and cause the impediment of PD progression remains poorly characterized. Therefore, we investigated 8-weeks supplementation effects of probiotic [Lactobacillus salivarius subsp. salicinius AP-32 (AP-32)], residual medium (RM) obtained from the AP-32 culture medium, and combination of AP-32 and RM (A-RM) on unilateral 6-hydroxydopamine (6-OHDA)-induced PD rats. We found that AP-32, RM and A-RM supplementation induced neuroprotective effects on dopaminergic neurons along with improved motor functions in PD rats. These effects were accompanied by significant increases in mitochondrial activities in the brain and muscle, antioxidative enzymes level in serum, and altered SCFAs profile in fecal samples. Importantly, the AP-32 supplement restored muscle mass along with improved motor function in PD rats, and produced the best results among the supplements. Our results demonstrate that probiotic AP-32 and A-RM supplementations can recover energy metabolism via increasing SCFAs producing and mitochondria function. This restoring of mitochondrial function in the brain and muscles with improved energy metabolism might additionally be potentiated by ROS suppression by the elevated generation of antioxidants, and which finally leads to facilitated recovery of 6-OHDA-induced motor deficit. Taken together, this work demonstrates that probiotic AP-32 supplementation could be a potential candidate for alternate treatment strategy to avert PD progression.
Collapse
Affiliation(s)
- Bira Arumndari Nurrahma
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan
| | - Shu-Ping Tsao
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Chieh-Hsi Wu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan.,School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Tu-Hsueh Yeh
- Department of Neurology, Taipei Medical University Hospital, Taipei City, Taiwan.,Department of Neurology, College of Medicine and Taipei Neuroscience Institute, Taipei Medical University, Taipei City, Taiwan
| | | | - Binar Panunggal
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan.,Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Central Java, Indonesia
| | - Hui-Yu Huang
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan
| |
Collapse
|
82
|
Hernández MAG, Canfora EE, Blaak EE. Faecal microbial metabolites of proteolytic and saccharolytic fermentation in relation to degree of insulin resistance in adult individuals. Benef Microbes 2021; 12:259-266. [PMID: 33880973 DOI: 10.3920/bm2020.0179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The gut microbiota may affect host metabolic health through microbial metabolites. The balance between the production of microbial metabolites by saccharolytic and proteolytic fermentation may be an important determinant of metabolic health. Amongst the best-studied saccharolytic microbial metabolites are the short-chain fatty acids acetate, propionate and butyrate. However, human data on the role of other microbial fermentation by-products in metabolic health are greatly lacking. Therefore, we compared in a cross-sectional study the faecal microbial metabolites (caproate, lactate, valerate, succinate, and the branched-chain fatty acids (BCFA) (isobutyrate, isovalerate)) between insulin sensitive (homeostatic model assessment of insulin resistance (HOMA-IR), HOMA-IR<1.85, IS) and insulin resistant (HOMA-IR>1.85, IR) individuals. Additionally, we assessed the relationships between faecal metabolites and markers of metabolic health including fasting glucose, insulin, free fatty acids, insulin resistance (HOMA-IR) and fasting substrate oxidation in 86 individuals with a wide range of body mass index. Faecal metabolite concentrations did not significantly differ between IS and IR. Furthermore, there were no associations between microbial metabolites and metabolic health markers, except for a slight positive association of isovalerate with carbohydrate oxidation (E%, std β 0.194, P=0.011) and fat oxidation (E%, std β -0.075, P=0.047), also after adjustment for age, sex and BMI. In summary, faecal caproate, lactate, valerate, succinate, and BCFA (isobutyrate, isovalerate) were not different between IR and IS individuals, nor was there any association between these faecal metabolites and parameters of metabolic health. Further human intervention studies are warranted to investigate the role of these microbially-derived fermentation products and their kinetics in metabolic health and insulin sensitivity.
Collapse
Affiliation(s)
- M A González Hernández
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Universiteitssingel 40, 6229 ET Maastricht, the Netherlands
| | - E E Canfora
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Universiteitssingel 40, 6229 ET Maastricht, the Netherlands
| | - E E Blaak
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Universiteitssingel 40, 6229 ET Maastricht, the Netherlands
| |
Collapse
|
83
|
Ratajczak W, Mizerski A, Rył A, Słojewski M, Sipak O, Piasecka M, Laszczyńska M. Alterations in fecal short chain fatty acids (SCFAs) and branched short-chain fatty acids (BCFAs) in men with benign prostatic hyperplasia (BPH) and metabolic syndrome (MetS). Aging (Albany NY) 2021; 13:10934-10954. [PMID: 33847600 PMCID: PMC8109139 DOI: 10.18632/aging.202968] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/02/2021] [Indexed: 12/12/2022]
Abstract
Gut microbiome-derived short-chain fatty acids (SCFAs) emerge in the process of fermentation of polysaccharides that resist digestion (dietary fiber, resistant starch). SCFAs have a very high immunomodulatory potential and ensure local homeostasis of the intestinal epithelium, which helps maintain the intestinal barrier. We analyzed the association between stool SCFAs levels acetic acid (C 2:0), propionic acid (C 3:0), isobutyric acid (C 4:0i), butyric acid (C 4:0n), isovaleric acid (C 5:0i) valeric acid (C 5:0n), isocaproic acid (C 6:0i), and caproic acid (C 6:0n)) in aging man with benign prostatic hyperplasia (BPH) and healthy controls. The study involved 183 men (with BPH, n = 103; healthy controls, n = 80). We assessed the content of SCFAs in the stool samples of the study participants using gas chromatography. The levels of branched SCFAs (branched-chain fatty acids, BCFAs): isobutyric acid (C4:0i) (p = 0.008) and isovaleric acid (C5:0i) (p < 0.001) were significantly higher in patients with BPH than in the control group. In healthy participants isocaproic acid (C6:0i) predominated (p = 0.038). We also analyzed the relationship between stool SCFA levels and serum diagnostic parameters for MetS. We noticed a relationship between C3:0 and serum lipid parameters (mainly triglycerides) in both healthy individuals and patients with BPH with regard to MetS. Moreover we noticed relationship between C4:0i, C5:0i and C6:0i and MetS in both groups. Our research results suggest that metabolites of the intestinal microflora (SCFAs) may indicate the proper function of the intestines in aging men, and increased BCFAs levels are associated with the presence of BPH.
Collapse
Affiliation(s)
- Weronika Ratajczak
- Department of Histology and Development Biology, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland.,Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Arnold Mizerski
- Department of General and Gastroentereological Surgery, Pomeranian Medical University in Szczecin, Szczecin 71-252, Poland
| | - Aleksandra Rył
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Marcin Słojewski
- Department of Urology and Urological Oncology, Pomeranian Medical University in Szczecin, Szczecin 70-111, Poland
| | - Olimpia Sipak
- Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Małgorzata Piasecka
- Department of Histology and Development Biology, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Maria Laszczyńska
- Department of Histology and Development Biology, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| |
Collapse
|
84
|
Short-Chain Fatty Acids, Maternal Microbiota and Metabolism in Pregnancy. Nutrients 2021; 13:nu13041244. [PMID: 33918804 PMCID: PMC8069164 DOI: 10.3390/nu13041244] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Short-chain fatty acids (SCFAs), as products of intestinal bacterial metabolism, are particularly relevant in the diagnosis of intestinal dysbiosis. The most common studies of microbiome metabolites include butyric acid, propionic acid and acetic acid, which occur in varying proportions depending on diet, age, coexisting disease and other factors. During pregnancy, metabolic changes related to the protection of energy homeostasis are of fundamental importance for the developing fetus, its future metabolic fate and the mother’s health. SCFAs act as signaling molecules that regulate the body’s energy balance through G-protein receptors. GPR41 receptors affect metabolism through the microflora, while GPR43 receptors are recognized as a molecular link between diet, microflora, gastrointestinal tract, immunity and the inflammatory response. The possible mechanism by which the gut microflora may contribute to fat storage, as well as the occurrence of gestational insulin resistance, is blocking the expression of the fasting-induced adipose factor. SCFAs, in particular propionic acid via GPR, determine the development and metabolic programming of the fetus in pregnant women. The mechanisms regulating lipid metabolism during pregnancy are similar to those found in obese people and those with impaired microbiome and its metabolites. The implications of SCFAs and metabolic disorders during pregnancy are therefore critical to maternal health and neonatal development. In this review paper, we summarize the current knowledge about SCFAs, their potential impact and possible mechanisms of action in relation to maternal metabolism during pregnancy. Therefore, they constitute a contemporary challenge to practical nutritional therapy. Material and methods: The PubMed database were searched for “pregnancy”, “lipids”, “SCFA” in conjunction with “diabetes”, “hypertension”, and “microbiota”, and searches were limited to work published for a period not exceeding 20 years in the past. Out of 2927 publication items, 2778 papers were excluded from the analysis, due to being unrelated to the main topic, conference summaries and/or articles written in a language other than English, while the remaining 126 publications were included in the analysis.
Collapse
|
85
|
Kumar S, Khan MA, Beijer E, Liu J, Lowe KK, Young W, Mills DA, Moon CD. Effect of milk replacer allowance on calf faecal bacterial community profiles and fermentation. Anim Microbiome 2021; 3:27. [PMID: 33795026 PMCID: PMC8017768 DOI: 10.1186/s42523-021-00088-2] [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: 09/30/2020] [Accepted: 03/15/2021] [Indexed: 12/02/2022] Open
Abstract
Background The nutrition of calves from birth until weaning is predominantly from liquid (milk or milk-based) feeds. Liquid feed allowances are often restricted during artificial rearing to accelerate the development of the rumen by promoting solid feed intake. Liquid feeds bypass the rumen and are digested in the lower digestive tract, however, the influence of different types of milk feeds, and their allowances, on the calf hindgut microbiota is not well understood. In this study, faecal samples from 199 calves raised on three different allowances of milk replacer: 10% of initial bodyweight (LA), 20% of initial bodyweight (HA), and ad libitum (ADLIB), were collected just prior to weaning. Bacterial community structures and fermentation products were analysed, and their relationships with calf growth and health parameters were examined to identify potential interactions between diet, gut microbiota and calf performance. Results Differences in the total concentrations of short-chain fatty acids were not observed, but higher milk replacer allowances increased the concentrations of branched short-chain fatty acids and decreased acetate to propionate ratios. The bacterial communities were dominated by Ruminococcaceae, Lachnospiraceae and Bacteroides, and the bacterial diversity of the ADLIB diet group was greater than that of the other diet groups. Faecalibacterium was over three times more abundant in the ADLIB compared to the LA group, and its abundance correlated strongly with girth and body weight gains. Milk replacer intake correlated strongly with Peptococcus and Blautia, which also correlated with body weight gain. Bifidobacterium averaged less than 1% abundance, however its levels, and those of Clostridium sensu stricto 1, correlated strongly with initial serum protein levels, which are an indicator of colostrum intake and passive transfer of immunoglobulins in early life. Conclusions Higher milk replacer intakes in calves increased hindgut bacterial diversity and resulted in bacterial communities and short chain fatty acid profiles associated with greater protein fermentation. Increased abundances of beneficial bacteria such as Faecalibacterium, were also observed, which may contribute to development and growth. Moreover, correlations between microbial taxa and initial serum protein levels suggest that colostrum intake in the first days of life may influence microbiota composition at pre-weaning. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-021-00088-2.
Collapse
Affiliation(s)
- Sandeep Kumar
- AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - M Ajmal Khan
- AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Emma Beijer
- Animal Nutrition Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Jinxin Liu
- Department of Food Science and Technology, Robert Mondavi Institute for Wine and Food Science, University of California, Davis, One Shields Ave, Davis, CA, 95616, USA.,Foods for Health Institute, University of California, Davis, California, One Shields Ave, Davis, CA, 95616, USA
| | - Katherine K Lowe
- AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - Wayne Young
- AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand
| | - David A Mills
- Department of Food Science and Technology, Robert Mondavi Institute for Wine and Food Science, University of California, Davis, One Shields Ave, Davis, CA, 95616, USA.,Foods for Health Institute, University of California, Davis, California, One Shields Ave, Davis, CA, 95616, USA.,Department of Viticulture and Enology, Robert Mondavi Institute for Wine and Food Science, University of California, Davis, California, One Shields Ave, Davis, CA, 95616, USA
| | - Christina D Moon
- AgResearch Limited, Grasslands Research Centre, Palmerston North, New Zealand.
| |
Collapse
|
86
|
Pham VT, Calatayud M, Rotsaert C, Seifert N, Richard N, Van den Abbeele P, Marzorati M, Steinert RE. Antioxidant Vitamins and Prebiotic FOS and XOS Differentially Shift Microbiota Composition and Function and Improve Intestinal Epithelial Barrier In Vitro. Nutrients 2021; 13:nu13041125. [PMID: 33805552 PMCID: PMC8066074 DOI: 10.3390/nu13041125] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Human gut microbiota (HGM) play a significant role in health and disease. Dietary components, including fiber, fat, proteins and micronutrients, can modulate HGM. Much research has been performed on conventional prebiotics such as fructooligosaccharides (FOS) and galactooligosaccharides (GOS), however, novel prebiotics or micronutrients still require further validation. We assessed the effect of FOS, xylooligosaccharides (XOS) and a mixture of an antioxidant vitamin blend (AOB) on gut microbiota composition and activity, and intestinal barrier in vitro. We used batch fermentations and tested the short-term effect of different products on microbial activity in six donors. Next, fecal inocula from two donors were used to inoculate the simulator of the human microbial ecosystem (SHIME) and after long-term exposure of FOS, XOS and AOB, microbial activity (short- and branched-chain fatty acids and lactate) and HGM composition were evaluated. Finally, in vitro assessment of intestinal barrier was performed in a Transwell setup of differentiated Caco-2 and HT29-MTX-E12 cells exposed to fermentation supernatants. Despite some donor-dependent differences, all three tested products showed beneficial modulatory effects on microbial activity represented by an increase in lactate and SCFA levels (acetate, butyrate and to a lesser extent also propionate), while decreasing proteolytic markers. Bifidogenic effect of XOS was consistent, while AOB supplementation appears to exert a specific impact on reducing F. nucleatum and increasing butyrate-producing B. wexlerae. Functional and compositional microbial changes were translated to an in vitro host response by increases of the intestinal barrier integrity by all the products and a decrease of the redox potential by AOB supplementation.
Collapse
Affiliation(s)
- Van T. Pham
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., 4002 Basel, Switzerland; (N.S.); (N.R.); (R.E.S.)
- Correspondence: ; Tel.: +41-618-158-828
| | - Marta Calatayud
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.C.); (C.R.); (P.V.d.A.); (M.M.)
| | - Chloë Rotsaert
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.C.); (C.R.); (P.V.d.A.); (M.M.)
| | - Nicole Seifert
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., 4002 Basel, Switzerland; (N.S.); (N.R.); (R.E.S.)
| | - Nathalie Richard
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., 4002 Basel, Switzerland; (N.S.); (N.R.); (R.E.S.)
| | - Pieter Van den Abbeele
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.C.); (C.R.); (P.V.d.A.); (M.M.)
| | - Massimo Marzorati
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.C.); (C.R.); (P.V.d.A.); (M.M.)
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Robert E. Steinert
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., 4002 Basel, Switzerland; (N.S.); (N.R.); (R.E.S.)
- Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
87
|
Yu H, Fang C, Li P, Wu M, Shen S. The relevance of DHA with modulating of host-gut microbiome signatures alterations and repairing of lipids metabolism shifts. Eur J Pharmacol 2021; 895:173885. [PMID: 33482183 DOI: 10.1016/j.ejphar.2021.173885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
Huge of previous reports recommended that gut microbiome have a crucial role in the human health and its change was profound impact for the metabolic improvements associated with lipids metabolism. In order to explore the relevance of a direct dysbiosis effect of gut microbiome on lipids metabolism shifts and repaired position of DHA, we built the animal model for the study with gut microbiome dysbiosis administrated by i.g. with CRO and intervened by DHA in the present work. Gut microbiome was analyzed by high throughput sequencing and bioinformatics analyses of bacteria. The composition of fatty acids and short chain fatty acids (SCFAs) were determined by gas chromatography. Blood lipids and bile acids were assayed by kit and UPLC-MS/MS, respectively. The expressions of enzymes of long chain fatty acid metabolism were analyzed by qRT-PCR. The results showed that gut microbiome dysbiosis caused lipid metabolism abnormal, and DHA was able to repair the lipids metabolism shifts resulted from gut microbiome dysbiosis. DHA could modulate host-gut microbiome signatures, improve concentrations of SCFAs, regulate fatty acids metabolism but modify bile acid profiles. In conclusion, we considered that DHA repaired lipid metabolism by modulating gut microbiome and regulating fatty acids metabolism pathway.
Collapse
Affiliation(s)
- Haining Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| | - Chengjie Fang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Peng Li
- Department of Geratoloy, The Third People's Hospital of Hangzhou, Hangzhou, China
| | - Manman Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | | |
Collapse
|
88
|
Becattini S, Sorbara MT, Kim SG, Littmann EL, Dong Q, Walsh G, Wright R, Amoretti L, Fontana E, Hohl TM, Pamer EG. Rapid transcriptional and metabolic adaptation of intestinal microbes to host immune activation. Cell Host Microbe 2021; 29:378-393.e5. [PMID: 33539766 PMCID: PMC7954923 DOI: 10.1016/j.chom.2021.01.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/08/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022]
Abstract
The gut microbiota produces metabolites that regulate host immunity, thereby impacting disease resistance and susceptibility. The extent to which commensal bacteria reciprocally respond to immune activation, however, remains largely unexplored. Herein, we colonized mice with four anaerobic symbionts and show that acute immune responses result in dramatic transcriptional reprogramming of these commensals with minimal changes in their relative abundance. Transcriptomic changes include induction of stress-response mediators and downregulation of carbohydrate-degrading factors such as polysaccharide utilization loci (PULs). Flagellin and anti-CD3 antibody, two distinct immune stimuli, induced similar transcriptional profiles, suggesting that commensal bacteria detect common effectors or activate shared pathways when facing different host responses. Immune activation altered the intestinal metabolome within 6 hours, decreasing luminal short-chain fatty acid and increasing aromatic metabolite concentrations. Thus, intestinal bacteria, prior to detectable shifts in community composition, respond to acute host immune activation by rapidly changing gene transcription and immunomodulatory metabolite production.
Collapse
Affiliation(s)
- Simone Becattini
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology and Immunology, School of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| | - Matthew T Sorbara
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Sohn G Kim
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Eric L Littmann
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Qiwen Dong
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Gavin Walsh
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Roberta Wright
- Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Luigi Amoretti
- Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Emily Fontana
- Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Tobias M Hohl
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
89
|
Macho-González A, Garcimartín A, Redondo N, Cofrades S, Bastida S, Nova E, Benedí J, Sánchez-Muniz FJ, Marcos A, Elvira López-Oliva M. Carob fruit extract-enriched meat, as preventive and curative treatments, improves gut microbiota and colonic barrier integrity in a late-stage T2DM model. Food Res Int 2021; 141:110124. [PMID: 33641991 DOI: 10.1016/j.foodres.2021.110124] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/19/2020] [Accepted: 01/07/2021] [Indexed: 12/28/2022]
Abstract
Epidemiological and experimental studies have suggested that dietary fiber and proanthocyanidins play an important role on gut microbiota (GM), colonic integrity and body health. Type 2 Diabetes Mellitus (T2DM) is a prevalent disease in which the modifications in the GM and colonic markers stand out. This manuscript hypothesizes the consumption of functional meat enriched in carob fruit extract [CFE; CFE-restructured meat (RM)] ameliorates the dysbiosis and colonic barrier integrity loss in a late-stage T2DM rat model induced by the conjoint action of a high-saturated-fat/high-cholesterol diet (Chol-diet) and a low dose of streptozotocin (STZ) plus a nicotinamide (NAD) injection. Three groups of eight rats were used: (1) D group, a T2DM control group, fed the Chol-diet; (2) ED group, a T2DM preventive strategy group fed the CFE-Chol-diet since the beginning of the study; and (3) DE group, a T2DM curative treatment group, fed the CFE-Chol-diet once the diabetic state was confirmed. The study lasted 8 weeks. Amount and variety of GM, feces short-chain-fatty acids (SCFAs), colonic morphology [crypt depth and density, goblet cells, proliferating cell nuclear antigen (PCNA) and transferase dUTP nick end labelling (TUNEL) indexes] and tight junctions were evaluated. A global colonic index combining 17 markers (GCindex) was calculated. ED rats displayed higher levels of GM richness, SCFAs production, crypt depth, and goblet cells than the D group. DE group showed lower Enterobacteriaceae abundance and greater TUNEL index and occludin expression in the distal colon than D counterpart. GCindex differentiated the colonic health status of the experimental groups in the order (ED > DE > D; P < 0.001) as a 17-51 range-quotation, ED, DE, and D groups displayed the values 43, 32.5, and 27, respectively. Thus, CFE-RM used as a T2DM preventive therapy could induce higher GM richness, more adequate SCFAs production, and better colonic barrier integrity. Furthermore, CFE-RM used with curative purposes induced more modest changes and mainly at the distal colonic mucosa. Further studies are needed to confirm this study's results, to ascertain the benefits of consuming proanthocyanidins-rich fiber during different T2DM stages.
Collapse
Affiliation(s)
- Adrián Macho-González
- Nutrition and Food Science Department (Nutrition), Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Alba Garcimartín
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Noemí Redondo
- Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), Spain
| | - Susana Cofrades
- Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), Spain
| | - Sara Bastida
- Nutrition and Food Science Department (Nutrition), Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Esther Nova
- Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), Spain
| | - Juana Benedí
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Francisco J Sánchez-Muniz
- Nutrition and Food Science Department (Nutrition), Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Ascensión Marcos
- Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), Spain
| | - M Elvira López-Oliva
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
90
|
Crittenden S, Goepp M, Pollock J, Robb CT, Smyth DJ, Zhou Y, Andrews R, Tyrrell V, Gkikas K, Adima A, O'Connor RA, Davies L, Li XF, Yao HX, Ho GT, Zheng X, Mair A, Vermeren S, Qian BZ, Mole DJ, Gerasimidis K, Schwarze JKJ, Breyer RM, Arends MJ, O'Donnell VB, Iredale JP, Anderton SM, Narumiya S, Maizels RM, Rossi AG, Howie SE, Yao C. Prostaglandin E 2 promotes intestinal inflammation via inhibiting microbiota-dependent regulatory T cells. SCIENCE ADVANCES 2021; 7:eabd7954. [PMID: 33579710 PMCID: PMC7880593 DOI: 10.1126/sciadv.abd7954] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/24/2020] [Indexed: 05/26/2023]
Abstract
The gut microbiota fundamentally regulates intestinal homeostasis and disease partially through mechanisms that involve modulation of regulatory T cells (Tregs), yet how the microbiota-Treg cross-talk is physiologically controlled is incompletely defined. Here, we report that prostaglandin E2 (PGE2), a well-known mediator of inflammation, inhibits mucosal Tregs in a manner depending on the gut microbiota. PGE2 through its receptor EP4 diminishes Treg-favorable commensal microbiota. Transfer of the gut microbiota that was modified by PGE2-EP4 signaling modulates mucosal Treg responses and exacerbates intestinal inflammation. Mechanistically, PGE2-modified microbiota regulates intestinal mononuclear phagocytes and type I interferon signaling. Depletion of mononuclear phagocytes or deficiency of type I interferon receptor diminishes PGE2-dependent Treg inhibition. Together, our findings provide emergent evidence that PGE2-mediated disruption of microbiota-Treg communication fosters intestinal inflammation.
Collapse
Affiliation(s)
- Siobhan Crittenden
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marie Goepp
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Jolinda Pollock
- SRUC Veterinary Services, Scotland's Rural College, Easter Bush Estate EH26 0PZ, UK
| | - Calum T Robb
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Danielle J Smyth
- Wellcome Centre for Molecular Parasitology, Institute for Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - You Zhou
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Robert Andrews
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Victoria Tyrrell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Konstantinos Gkikas
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow G31 2ER, UK
| | - Alexander Adima
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Richard A O'Connor
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Luke Davies
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Xue-Feng Li
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Hatti X Yao
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gwo-Tzer Ho
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Xiaozhong Zheng
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Amil Mair
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Sonja Vermeren
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Bin-Zhi Qian
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Damian J Mole
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow G31 2ER, UK
| | - Jürgen K J Schwarze
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark J Arends
- Division of Pathology, Cancer Research UK Edinburgh Centre, The University of Edinburgh, Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, UK
| | - Valerie B O'Donnell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - John P Iredale
- Senate House, University of Bristol, Bristol BS8 1TH, UK
| | - Stephen M Anderton
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Shuh Narumiya
- Alliance Laboratory for Advanced Medical Research and Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Rick M Maizels
- Wellcome Centre for Molecular Parasitology, Institute for Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Sarah E Howie
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Chengcan Yao
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
91
|
Li R, Yao Y, Gao P, Bu S. The Therapeutic Efficacy of Curcumin vs. Metformin in Modulating the Gut Microbiota in NAFLD Rats: A Comparative Study. Front Microbiol 2021; 11:555293. [PMID: 33584555 PMCID: PMC7874275 DOI: 10.3389/fmicb.2020.555293] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Structural disruption of gut microbiota is closely related to the occurrence of non-alcoholic fatty liver disease (NAFLD). Previous research has demonstrated that both curcumin (CUR) and metformin (MET) have a therapeutic effect against NAFLD and play a role in modulating the gut microbiota. However, there is a lack of direct comparison between the two medications in terms of the therapeutic efficacy and the regulatory effect on gut microbiota. In this study, we administered either CUR or MET to rats with high-fat diet (HFD)-induced obesity to observe changes in body parameters, biochemical parameters, liver, and ileum pathology and gut microbiota, and used next generation sequencing and multivariate analysis to evaluate the structural changes of gut microbiota in a NAFLD rat model before and after CUR and MET intervention. It was found that both CUR and MET attenuated hepatic ectopic fat deposition, alleviated inflammatory factors, and improved intestinal barrier integrity in HFD-fed rats. More importantly, CUR and MET reduced the Firmicutes/Bacteroidetes ratio and reverted the composition of the HFD-disrupted gut microbiota. Both CUR and MET treatments effectively modified the gut microbiome, enriched the abundance of beneficial bacteria and reduced opportunistic pathogens in obese rats. The abundance of Butyricicoccus was increased while the abundance of Dorea was decreased in HFD + CUR group. Besides, some beneficial bacteria such as Prevotella were increased in MET-treated animals. Spearman’s correlation analysis showed that Helicobacter, Akkermansia, Desulfovibrio, Romboutsia, Corynebacterium, Lactobacillus, Ruminococcaceae_unclassified, Lachnospiraceae_unclassified, and Clostridiales_unclassified showed significantly positive correlations with TG, TC, LDL-C, GLU, IL-6, IL-1β, and TNF-α, and negative correlations with HDL-C (both p < 0.05). However, Prevotella and Stomatobaculum showed an opposite trend. In summary, CUR and MET showed similar effects in alleviating hepatic steatosis, improving intestinal barrier integrity and modulating gut microbiota in HFD-induced obesity rats, and therefore may prove to be a novel adjunctive therapy for NAFLD.
Collapse
Affiliation(s)
- Ruifang Li
- Department of Gastroenterology, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yurong Yao
- Department of Infection, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Pengfei Gao
- Department of Traditional Chinese Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Shurui Bu
- Department of Gastroenterology, Jinshan Hospital Affiliated to Fudan University, Shanghai, China.,Department of Infection, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
92
|
Norheim F, Chella Krishnan K, Bjellaas T, Vergnes L, Pan C, Parks BW, Meng Y, Lang J, Ward JA, Reue K, Mehrabian M, Gundersen TE, Péterfy M, Dalen KT, Drevon CA, Hui ST, Lusis AJ, Seldin MM. Genetic regulation of liver lipids in a mouse model of insulin resistance and hepatic steatosis. Mol Syst Biol 2021; 17:e9684. [PMID: 33417276 PMCID: PMC7792507 DOI: 10.15252/msb.20209684] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/31/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
To elucidate the contributions of specific lipid species to metabolic traits, we integrated global hepatic lipid data with other omics measures and genetic data from a cohort of about 100 diverse inbred strains of mice fed a high-fat/high-sucrose diet for 8 weeks. Association mapping, correlation, structure analyses, and network modeling revealed pathways and genes underlying these interactions. In particular, our studies lead to the identification of Ifi203 and Map2k6 as regulators of hepatic phosphatidylcholine homeostasis and triacylglycerol accumulation, respectively. Our analyses highlight mechanisms for how genetic variation in hepatic lipidome can be linked to physiological and molecular phenotypes, such as microbiota composition.
Collapse
Affiliation(s)
- Frode Norheim
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Department of NutritionInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | | | | | - Laurent Vergnes
- Department of Human GeneticsUniversity of California at Los AngelesLos AngelesCAUSA
| | - Calvin Pan
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Brian W Parks
- Department of Nutritional SciencesUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Yonghong Meng
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Jennifer Lang
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - James A Ward
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Karen Reue
- Department of Human GeneticsUniversity of California at Los AngelesLos AngelesCAUSA
| | - Margarete Mehrabian
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | | | - Miklós Péterfy
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Depatrment of Basic Medical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Knut T Dalen
- Department of NutritionInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | - Christian A Drevon
- Department of NutritionInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
- Vitas ASOsloNorway
| | - Simon T Hui
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
| | - Aldons J Lusis
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Department of Human GeneticsUniversity of California at Los AngelesLos AngelesCAUSA
| | - Marcus M Seldin
- Division of CardiologyDepartment of MedicineUniversity of California at Los AngelesLos AngelesCAUSA
- Department of Biological Chemistry and Center for Epigenetics and MetabolismUniversity of California, IrvineIrvineCAUSA
| |
Collapse
|
93
|
Hiippala K, Barreto G, Burrello C, Diaz-Basabe A, Suutarinen M, Kainulainen V, Bowers JR, Lemmer D, Engelthaler DM, Eklund KK, Facciotti F, Satokari R. Novel Odoribacter splanchnicus Strain and Its Outer Membrane Vesicles Exert Immunoregulatory Effects in vitro. Front Microbiol 2020; 11:575455. [PMID: 33281770 PMCID: PMC7689251 DOI: 10.3389/fmicb.2020.575455] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022] Open
Abstract
Odoribacter splanchnicus, belonging to the order Bacteroidales, is a common, short-chain fatty acid producing member of the human intestinal microbiota. A decreased abundance of Odoribacter has been linked to different microbiota-associated diseases, such as non-alcoholic fatty liver disease, cystic fibrosis and inflammatory bowel disease (IBD). The type strain of O. splanchnicus has been genome-sequenced, but otherwise very little is known about this anaerobic bacterium. The species surfaces in many microbiota studies and, consequently, comprehension on its interactions with the host is needed. In this study, we isolated a novel strain of O. splanchnicus from a healthy fecal donor, identified it by genome sequencing and addressed its adhesive, epithelium reinforcing and immunoregulatory properties. Our results show that O. splanchnicus strain 57 is non-adherent to enterocytes or mucus, does not reinforce nor compromise Caco-2 monolayer integrity and most likely harbors penta-acylated, less endotoxic lipid A as part of its lipopolysaccharide (LPS) structure based on the lack of gene lpxM and in vitro results on low-level NF-κB activity. The studies by transmission electron microscopy revealed that O. splanchnicus produces outer membrane vesicles (OMV). O. splanchnicus cells, culture supernatant i.e., spent medium or OMVs did not induce interleukin-8 (IL-8) response in HT-29 enterocyte cells suggesting a very low proinflammatory capacity. On the contrary, the treatment of HT-29 cells with O. splanchnicus cells, spent medium or OMVs prior to exposure to Escherichia coli LPS elicited a significant decrease in IL-8 production as compared to E. coli LPS treatment alone. Moreover, O. splanchnicus spent supernatant induced IL-10 production by immune cells, suggesting anti-inflammatory activity. Our in vitro findings indicate that O. splanchnicus and its effector molecules transported in OMVs could potentially exert anti-inflammatory action in the gut epithelium. Taken together, O. splanchnicus seems to be a commensal with a primarily beneficial interaction with the host.
Collapse
Affiliation(s)
- Kaisa Hiippala
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Gonçalo Barreto
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Claudia Burrello
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Angelica Diaz-Basabe
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Maiju Suutarinen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Veera Kainulainen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jolene R Bowers
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, Arizona, AZ, United States
| | - Darrin Lemmer
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, Arizona, AZ, United States
| | - David M Engelthaler
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, Arizona, AZ, United States
| | - Kari K Eklund
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Helsinki University and Helsinki University Hospital, Department of Rheumatology, Helsinki, Finland and ORTON Orthopedic Hospital of the Orton Foundation, Helsinki, Finland
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Reetta Satokari
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
94
|
Caulerpa lentillifera (Sea Grapes) Improves Cardiovascular and Metabolic Health of Rats with Diet-Induced Metabolic Syndrome. Metabolites 2020; 10:metabo10120500. [PMID: 33297424 PMCID: PMC7762211 DOI: 10.3390/metabo10120500] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023] Open
Abstract
Caulerpa lentillifera (sea grapes) is widely consumed in South-East Asia as a low-energy food with high contents of vitamins and minerals. This study investigated dried sea grapes containing 16.6% insoluble fibre commercially produced in Vietnam as an intervention. We hypothesised that insoluble fibre is the primary metabolite that will reverse diet-induced metabolic syndrome. Male Wistar rats (n = 48) were randomly allocated to four groups in a 16 week protocol. Two groups were fed either corn starch (C) or high-carbohydrate, high-fat (H) diets for the full 16 weeks. The other two groups received C and H diets for eight weeks and then received C. lentillifera added to these diets for the final eight weeks (CCL and HCL, respectively). High-carbohydrate, high-fat diet-fed rats developed obesity, hypertension, dyslipidaemia, fatty liver disease and increased left ventricular collagen deposition. C. lentillifera supplementation in HCL rats decreased body weight, systolic blood pressure, plasma concentrations of total cholesterol and non-esterified fatty acids, inflammatory cells in heart and liver, and visceral adiposity. The Firmicutes to Bacteroidetes ratio decreased in the gut microbiota of HCL rats. Therefore, C. lentillifera attenuated cardiovascular and metabolic symptoms of metabolic syndrome in rats, possibly by preventing infiltration of inflammatory cells together with modulating gut microbiota.
Collapse
|
95
|
Plaza-Díaz J, Solís-Urra P, Rodríguez-Rodríguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadía-Molina F, Álvarez-Mercado AI. The Gut Barrier, Intestinal Microbiota, and Liver Disease: Molecular Mechanisms and Strategies to Manage. Int J Mol Sci 2020; 21:E8351. [PMID: 33171747 PMCID: PMC7664383 DOI: 10.3390/ijms21218351] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Liver disease encompasses pathologies as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, alcohol liver disease, hepatocellular carcinoma, viral hepatitis, and autoimmune hepatitis. Nowadays, underlying mechanisms associating gut permeability and liver disease development are not well understood, although evidence points to the involvement of intestinal microbiota and their metabolites. Animal studies have shown alterations in Toll-like receptor signaling related to the leaky gut syndrome by the action of bacterial lipopolysaccharide. In humans, modifications of the intestinal microbiota in intestinal permeability have also been related to liver disease. Some of these changes were observed in bacterial species belonging Roseburia, Streptococcus, and Rothia. Currently, numerous strategies to treat liver disease are being assessed. This review summarizes and discusses studies addressed to determine mechanisms associated with the microbiota able to alter the intestinal barrier complementing the progress and advancement of liver disease, as well as the main strategies under development to manage these pathologies. We highlight those approaches that have shown improvement in intestinal microbiota and barrier function, namely lifestyle changes (diet and physical activity) and probiotics intervention. Nevertheless, knowledge about how such modifications are beneficial is still limited and specific mechanisms involved are not clear. Thus, further in-vitro, animal, and human studies are needed.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada;
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
| | - Patricio Solís-Urra
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2531015, Chile;
| | - Fernando Rodríguez-Rodríguez
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile; (F.R.-R.); (J.O.-A.)
| | - Jorge Olivares-Arancibia
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile; (F.R.-R.); (J.O.-A.)
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad de las Américas, Santiago 8370035, Chile
| | - Miguel Navarro-Oliveros
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain;
| | - Francisco Abadía-Molina
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain;
- Department of Cell Biology, School of Sciences, University of Granada, 18071 Granada, Spain
| | - Ana I. Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain;
| |
Collapse
|
96
|
Neijat M, Habtewold J, Li S, Jing M, House JD. Effect of dietary n-3 polyunsaturated fatty acids on the composition of cecal microbiome of Lohmann hens. Prostaglandins Leukot Essent Fatty Acids 2020; 162:102182. [PMID: 33038831 DOI: 10.1016/j.plefa.2020.102182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022]
Abstract
Supplementation of n-3 fatty acids to poultry diets is widely acknowledged for its role in enhancing poultry products, however, little is known about the compositional responses of gut microbial communities to type and dosage of these supplements. Here, we compared the effects of n-3 polyunsaturated fatty acids (PUFA), supplied as alpha-linolenic acid (ALA) or docosahexaenoic acid (DHA), on the composition of bacterial communities in ceca of laying hens. Corn-soybean basal diets were supplemented with either flaxseed oil (FO, ALA-rich) or marine algal biomass (MA, DHA-rich), and each supplied 0.20 and 0.60% of total n-3 PUFA in the diet. Lohmann LSL-Classic laying hens (n = 10/treatment) were randomly allocated to one of the 4 diets. After 8 weeks of feeding, blood, liver and cecal digesta samples were obtained for plasma glucose, fatty acids, and short chain fatty acids analyses, respectively. The gut bacterial communities were characterized using genomic DNA extracted from cecal contents, whereby the V3-V4 hypervariable region of the 16S rRNA gene was sequenced using the Illumina Miseq® platform. Firmicutes and Bacteroidetes were the predominant phyla in both the FO- and MA-fed groups. The relative abundance of Tenericutes, often associated with immunomodulation, was relatively higher (P<0.0001) in the FO than MA group. Although the relative abundance of Bacteroides was greater for the FO- than the MA-fed group, this genus was negatively correlated (P<0.05) with total n-3 PUFA in the liver at higher dosages of both FO- and MA-fed hens. Higher dose of FO (0.60%) and both dosages of MA (0.20 and 0.60%) substantially enriched several members of Firmicutes (e.g., Faecalibacterium, Clostridium and Ruminococcus) which are known to produce butyrate. Moreover, co-occurrence network analysis revealed that, in the FO 0.60- and MA 0.20-fed hens, Ruminococcaceae was the most influential taxon accounting for about 31% of the network complexity. These findings demonstrate that supplementation of different type and level of n-3 PUFA in hens' diets could enrich microbial communities with potential role in lipid metabolism and health.
Collapse
Affiliation(s)
- M Neijat
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - J Habtewold
- Agriculture and Agri-Food Canada (AAFC), Ottawa, Ontario, Canada
| | - S Li
- Department of Animal Sciences, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - M Jing
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - J D House
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada; Department of Animal Sciences, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada; Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, R3T 2E1, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Research Centre, Winnipeg, MB, R2H 2A6, Canada.
| |
Collapse
|
97
|
Tuck CJ, De Palma G, Takami K, Brant B, Caminero A, Reed DE, Muir JG, Gibson PR, Winterborn A, Verdu EF, Bercik P, Vanner S. Nutritional profile of rodent diets impacts experimental reproducibility in microbiome preclinical research. Sci Rep 2020; 10:17784. [PMID: 33082369 PMCID: PMC7575541 DOI: 10.1038/s41598-020-74460-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
The lack of reproducibility of animal experimental results between laboratories, particularly in studies investigating the microbiota, has raised concern among the scientific community. Factors such as environment, stress and sex have been identified as contributors, whereas dietary composition has received less attention. This study firstly evaluated the use of commercially available rodent diets across research institutions, with 28 different diets reported by 45 survey respondents. Secondly, highly variable ingredient, FODMAP (Fermentable Oligo-, Di-, Mono-saccharides And Polyols) and gluten content was found between different commercially available rodent diets. Finally, 40 mice were randomized to four groups, each receiving a different commercially available rodent diet, and the dietary impact on cecal microbiota, short- and branched-chain fatty acid profiles was evaluated. The gut microbiota composition differed significantly between diets and sexes, with significantly different clusters in β-diversity. Total BCFA were highest (p = 0.01) and SCFA were lowest (p = 0.03) in mice fed a diet lower in FODMAPs and gluten. These results suggest that nutritional composition of commercially available rodent diets impact gut microbiota profiles and fermentation patterns, with major implications for the reproducibility of results across laboratories. However, further studies are required to elucidate the specific dietary factors driving these changes.
Collapse
Affiliation(s)
- C J Tuck
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada.
- Department of Dietetics, Nutrition and Sport, La Trobe University, Bundoora, VIC, Australia.
| | - G De Palma
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - K Takami
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - B Brant
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - A Caminero
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - D E Reed
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - J G Muir
- Department of Gastroenterology, Monash University, Melbourne, Australia
| | - P R Gibson
- Department of Gastroenterology, Monash University, Melbourne, Australia
| | - A Winterborn
- Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - E F Verdu
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - P Bercik
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - S Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| |
Collapse
|
98
|
Muralitharan RR, Jama HA, Xie L, Peh A, Snelson M, Marques FZ. Microbial Peer Pressure: The Role of the Gut Microbiota in Hypertension and Its Complications. HYPERTENSION (DALLAS, TEX. : 1979) 2020; 76:1674-1687. [PMID: 33012206 DOI: 10.1161/hypertensionaha.120.14473] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is increasing evidence of the influence of the gut microbiota on hypertension and its complications, such as chronic kidney disease, stroke, heart failure, and myocardial infarction. This is not surprising considering that the most common risk factors for hypertension, such as age, sex, medication, and diet, can also impact the gut microbiota. For example, sodium and fermentable fiber have been studied in relation to both hypertension and the gut microbiota. By combining second- and, now, third-generation sequencing with metabolomics approaches, metabolites, such as short-chain fatty acids and trimethylamine N-oxide, and their producers, have been identified and are now known to affect host physiology and the cardiovascular system. The receptors that bind these metabolites have also been explored with positive findings-examples include known short-chain fatty acid receptors, such as G-protein coupled receptors GPR41, GPR43, GPR109a, and OLF78 in mice. GPR41 and OLF78 have been shown to have inverse roles in blood pressure regulation, whereas GPR43 and GPR109A have to date been demonstrated to impact cardiac function. New treatment options in the form of prebiotics (eg, dietary fiber), probiotics (eg, Lactobacillus spp.), and postbiotics (eg, the short-chain fatty acids acetate, propionate, and butyrate) have all been demonstrated to be beneficial in lowering blood pressure in animal models, but the underlying mechanisms remain poorly understood and translation to hypertensive patients is still lacking. Here, we review the evidence for the role of the gut microbiota in hypertension, its risk factors, and cardiorenal complications and identify future directions for this exciting and fast-evolving field.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia (R.R.M.)
| | - Hamdi A Jama
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.A.J., F.Z.M.)
| | - Liang Xie
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia (L.X.)
| | - Alex Peh
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School (M.S.), Monash University, Melbourne, Australia
| | - Francine Z Marques
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.A.J., F.Z.M.)
| |
Collapse
|
99
|
Myronovych A, Bhattacharjee J, Salazar-Gonzalez RM, Tan B, Mowery S, Ferguson D, Ryan KK, Zhang W, Zhao X, Oehrle M, Setchell KD, Seeley RJ, Sandoval DA, Kohli R. Assessment of the role of FGF15 in mediating the metabolic outcomes of murine Vertical Sleeve Gastrectomy (VSG). Am J Physiol Gastrointest Liver Physiol 2020; 319:G669-G684. [PMID: 32967428 PMCID: PMC7792670 DOI: 10.1152/ajpgi.00175.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 01/31/2023]
Abstract
Vertical sleeve gastrectomy (VSG) is the best current therapy for remission of obesity and its co-morbidities. It is understood to alter the enterohepatic circulation of bile acids in vivo. Fibroblast growth factor 19 (FGF19) in human and its murine orthologue Fgf15 plays a pivotal role in this bile acid driven enterohepatic signaling. The present study evaluated the metabolic outcomes of VSG in Fgf15 deficient mice. 6-8 weeks old male wildtype mice (WT) and Fgf15 deficient mice (KO) were fed a high fat diet (HFD) for 8 weeks. At 8th week of diet, both WT and KO mice were randomly distributed to VSG or sham surgery. Post-surgery, mice were observed for 8 weeks while fed a HFD and then euthanized to collect tissues for experimental analysis. Fgf15 deficient (KO) mice lost weight post VSG, but glucose tolerance in KO mice did not improve post VSG compared to WT mice. Enteroids derived from WT and KO mice proliferated with bile acid exposure in vitro. Post VSG both WT and KO mice had similarly altered bile acid enterohepatic flux, however Fgf15 deficient mice post VSG had increased hepatic accumulation of free and esterified cholesterol leading to lipotoxicity related ER stress, inflammasome activation, and increased Fgf21 expression. Intact Fgf15 mediated enterohepatic bile acid signaling, but not changes in bile acid flux, appear to be important for the metabolic improvements post-murine bariatric surgery. These novel data introduce a potential point of distinction between bile acids acting as ligands compared to their canonical downstream signaling pathways.
Collapse
Affiliation(s)
| | | | | | - Brandon Tan
- Pediatrics, Cincinnati Children's Hospital Medical Center, United States
| | - Sarah Mowery
- Pediatrics, Cincinnati Children's Hospital Medical Center, United States
| | - Danielle Ferguson
- Pediatrics, Cincinnati Children's Hospital Medical Center, United States
| | | | - Wujuan Zhang
- Human Genetics, Cincinnati Children's Hospital Medical Center, United States
| | - Xueheng Zhao
- Pediatrics, Cincinnati Children's Hospital Medical Center, United States
| | - Melissa Oehrle
- Pediatrics, Cincinnati Children's Hospital Medical Center, United States
| | | | - Randy J Seeley
- Surgery, University of Michigan-Ann Arbor, United States
| | - Darleen A Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Rohit Kohli
- Pediatrics, Children's Hospital of Los Angeles, United States
| |
Collapse
|
100
|
Yao Z, Zhao M, Gong Y, Chen W, Wang Q, Fu Y, Guo T, Zhao J, Gao L, Bo T. Relation of Gut Microbes and L-Thyroxine Through Altered Thyroxine Metabolism in Subclinical Hypothyroidism Subjects. Front Cell Infect Microbiol 2020; 10:495. [PMID: 33072620 PMCID: PMC7531258 DOI: 10.3389/fcimb.2020.00495] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
Thyroxine metabolism is an important topic of pathogenesis research and treatment schedule of subclinical hypothyroidism (SCH). L-Thyroxine replacement therapy (LRT) is usually recommended for severe SCH patients only. Our previous studies reported that disordered serum lipid of mild SCH people could also benefit from LRT. However, the benefits were different among individuals, as shown by the variations in drug dosage that required to maintain thyroid-stimulating hormone (TSH) stability. Alternative pathways, such as sulfation and glucuronidation of iodothyronine, may play a role in thyroid hormones metabolism in peripheral tissues aside from thyroid. Conjugated thyroxine can be hydrolyzed and reused in tissues including gastrointestinal tract, in which gut microbiota are one of the most attractive physiological components. On this site, the roles of gut microbiota in thyroidal metabolism should be valued. In this study, a cross-sectional study was performed by analyzing 16S rDNA of gut microbiota in mild SCH patients treated with L-thyroxine or not. Subjects were divided by serum lipid level, L-thyroxine treatment, or L-thyroxine dosage, respectively. Relationship between gut microbiome and serum profile, L-thyroxine treatment, and dose were discussed. Other metabolic disorders such as type 2 diabetes and hypertension were also taken into consideration. It turned out that microbiome varied among individuals divided by dose and the increment of L-thyroxine but not by serum lipid profile. Relative abundance of certain species that were associated with thyroxine metabolism were found varied among different L-thyroxine doses although in relatively low abundance. Moreover, serum cholesterol may perform relevance effects with L-thyroxine in shaping microbiome. Our findings suggested that the differences in L-thyroxine dosage required to maintain TSH level stability, as well as the SCH development, which was displayed by the increased L-thyroxine doses in subsequent follow-up, had relationship with gut microbial composition. The reason may due to the differences in thyroxine metabolic capacity in gut. In addition, the metabolic similarity of iodothyronines and bile acid in gut also provides possibilities for the correlation between host's thyroxine and cholesterol levels. This study was registered with ClinicalTrials.gov as number NCT01848171.
Collapse
Affiliation(s)
- Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meng Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Gong
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenbin Chen
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qian Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yilin Fu
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tian Guo
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiajun Zhao
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China.,Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tao Bo
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|