1
|
Hong Y, Cui J, Xu G, Li N, Peng G. Intestinal IL-17 family orchestrates microbiota-driven histone deacetylation and promotes Treg differentiation to mediate the alleviation of asthma by Ma-Xing-Shi-Gan decoction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156656. [PMID: 40311598 DOI: 10.1016/j.phymed.2025.156656] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/08/2025] [Accepted: 03/15/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Gut microbiota imbalance is well-known as one important trigger of allergic asthma. Ma-Xing-Shi-Gan decoction (MXSG) is a traditional Chinese medicine prescription with ideal clinical efficacy on asthma. However, whether and how MXSG exerts its efficacy on asthma through gut microbiota remains unclear. PURPOSE To investigate the underlying mechanism of MXSG against asthma using multi-omics technologies. METHODS An asthma model was established using 8-week-old C57BL/6 J mice, after which they were daily administrated with high-, medium- and low-dose MXSG for 7 days. Histopathological examinations and flow cytometry were performed to evaluate the effects of MXSG on lung immune injury. Key regulatory pathways were predicted via network pharmacology and verified using 16S rRNA sequencing, metagenomics, metabolomics, and in vivo experiments including the knockout of the targeting gene. RESULTS MXSG alleviated asthma symptoms, elevated intestinal microbial diversities, and enriched potential beneficial microbes such as Lactococcus, Lactobacillus, and Limosilactobacillus. Network pharmacology and experimental validation highlighted the IL-17/Treg signaling as crucial for asthma treatment. IL-17 knockout experiments revealed its necessity for Treg differentiation during asthma. Moreover, IL-17-deficient asthmatic mice exhibited lower levels of Lactobacillus and significant changes in microbial genes involving histone deacetylases (HDAC) and short-chain fatty acids (SCFAs). Finally, MXSG significantly boosted SCFA production and reduced HDAC9 expression, which were correlated with Treg cell ratios. CONCLUSION Our study delineates a novel mechanism where MXSG synergizes with the IL-17 family to enrich intestinal beneficial microbes (e.g. Lactobacillus) and SCFAs. This inhibits the expression of SCFA-downstream HDAC9 to promote Treg differentiation, and thus potentially alleviates asthma.
Collapse
Affiliation(s)
- Yanfei Hong
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China
| | - Jiaqi Cui
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China
| | - Guichuan Xu
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China
| | - Na Li
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China.
| | - Guiying Peng
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China.
| |
Collapse
|
2
|
Zeng H, Safratowich BD, Liu Z, Bukowski MR. Resistant starch inhibits high-fat diet-induced oncogenic responses in the colon of C57BL/6 mice. J Nutr Biochem 2025; 139:109838. [PMID: 39788163 DOI: 10.1016/j.jnutbio.2025.109838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
The beneficial effects of dietary fiber for colon health may be due to short chain fatty acids (SCFAs), such as butyrate, produced by colonic bacterial fermentation. In contrast, obesogenic diet induced obesity is linked to increased colon cancer incidence. We hypothesize that increasing fiber intake promotes healthy microbiome and reduces bacterial dysbiosis and oncogenic signaling in the colon of mice fed an obesogenic diet. About 5-week-old male C57BL/6 mice were assigned to 5 dietary groups (n=22/group) for 24 weeks:(1) AIN93G as a control diet (AIN); (2) a high fat diet (HFD, 45% energy fat); (3) HFD+5% resistant starch enriched dietary fiber (RSF) from maize; (4) HFD+10%RSF; or (5) HFD+20%RSF. Compared to the AIN group, mice receiving the HFD exhibited more than 15% increase in body mass and body fat composition irrespective of RSF dosage. However, the HFD+RSF groups exhibited an increase (>300%) of fecal butyrate but a decrease (>45%) of secondary bile acids in a RSF dose-dependent manner over the HFD group. Similarly, there were concomitant decreases (>25%) in pro-inflammatory plasma cytokines (TNFα, IL-6 and MCP-1), β-catenin and Ki67 protein staining in the colon of the HFD+20%RSF group relative to the HFD group. Furthermore, the abundance of colonic Proteobacteria, signatures of dysbiosis, was decreased (>63%) in a RSF dose-dependent manner compared to the HFD. Collectively, these data indicate that RSF not only increases butyrate but also reduces secondary bile acids, bacterial dysbiosis and β-catenin in the colon of mice fed a HFD.
Collapse
Affiliation(s)
- Huawei Zeng
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203.
| | - Bryan D Safratowich
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203
| | - Zhenhua Liu
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003
| | - Michael R Bukowski
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203
| |
Collapse
|
3
|
Agyin-Birikorang A, Lennon S, Smith KS, Van Der Pol W, Smith MA, Sexton CL, Lamb DA, Young KC, Mobley CB, Huggins KW, Roberts MD, Frugé AD. Fecal Microbiota and Associated Metabolites Are Minimally Affected by Ten Weeks of Resistance Training in Younger and Older Adults. Sports (Basel) 2025; 13:98. [PMID: 40278724 PMCID: PMC12031165 DOI: 10.3390/sports13040098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Preclinical evidence suggests that short chain fatty acids (SCFAs) produced by gut microbiota may impact body composition and muscle growth. While aging is implicated in negative alterations to the gut microbiome, exercise may mitigate these changes. Limited human evidence indicates that resistance training (RT) does not appreciably alter the gut microbiome in older adults, and no human study has examined whether resistance training differentially alters the gut microbiome and associated SCFAs between younger and older individuals. Therefore, we examined whether 10 weeks of RT differentially altered fecal microbiota composition, fecal and circulating SCFAs, and serum markers associated with gastrointestinal integrity in two cohorts of adults. Fecal and serum samples were obtained from untrained younger (22 ± 2 years, n = 12) and older (58 ± 8 years, n = 12) participants prior to and following 10 weeks of supervised twice-weekly full-body RT. Outcome measures immediately before (PRE) and after the intervention (POST) included dual X-ray absorptiometry for body composition, ultrasound for vastus lateralis (VL) thickness, 16S rRNA gene sequencing fecal microbiome data, serum and fecal SCFAs measured by gas chromatography, and serum intestinal fatty acid-binding protein 2 (FABP2), lipopolysaccharide-binding protein (LBP), and leucine-rich alpha-2 glycoprotein (LRG-1) quantified by enzyme-linked immunosorbent assays. Main effects and interactions were measured by repeated measures analysis of variance (group × time; G × T) for all dependent variables, and Spearman correlations were used to explore relationships among changes in relevant outcomes. The intervention significantly increased VL thickness and lean body mass (p < 0.05) equally in both groups. Although group differences in microbiome beta diversity were identified, no effects of age, time, or their interaction were observed for the alpha diversity measures. Seven SCFAs were detected in the fecal samples, albeit no significant age, time, or interaction effects were evident. In serum, acetic acid was the only SCFA detected, with no significant age, time, or interaction effects. Serum LRG1 decreased for all participants (p = 0.007) with higher levels in younger adults (p = 0.015), but no G × T interactions were observed for this marker, serum FABP2, or LBP. No significant correlations were observed among RT-induced changes in muscle mass-related outcomes and changes in fecal microbiome diversity, total or individual SCFAs, or serum FABP2/LBP/LRG-1. These results highlight that 10 weeks of RT largely does not affect fecal microbiota, associated SCFAs, or select markers of gastrointestinal integrity in untrained younger or older adults.
Collapse
Affiliation(s)
| | - Sarah Lennon
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Kristen S. Smith
- Ocean Spray Cranberries, Inc., Middleboro-Lakeville, MA 02347, USA
| | - William Van Der Pol
- Biomedical Informatics, UAB Center for Clinical and Translational Science, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan A. Smith
- School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Casey L. Sexton
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Donald A. Lamb
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Kaelin C. Young
- Department of Physiology, Pacific Northwest University of Health Sciences, Yakima, WA 98901, USA
| | | | - Kevin W. Huggins
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | | | | |
Collapse
|
4
|
Cui X, Yang Y, Zhang M, Bao L, Jiao F, Wei X, Qian W, Shi X, Su C, Qian Y. Mulberry leaves supplementation modulates ruminal and fecal bacterial community and metabolites in growing mutton sheep. Sci Rep 2025; 15:7923. [PMID: 40050295 PMCID: PMC11885437 DOI: 10.1038/s41598-025-87298-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 01/17/2025] [Indexed: 03/09/2025] Open
Abstract
Mulberry leaves, a traditional Chinese medicine with the values of medicine and food, are rich in various active ingredients and nutrients. Gastrointestinal bacteria are recognized to play a crucial role in food digestion, nutrient metabolism and the host immune health. However, the modulations of mulberry leaves on microbiota and metabolites remain insufficiently understood. In this study, a feeding experiment was conducted involving three groups on growing mutton sheep: one fed with dried mulberry leaves, another with fermented mulberry leaves and one without mulberry leaves (as control), afterwards, 16s sequencing of ruminal and fecal microbiota was performed. The present study revealed that the addition of mulberry leaves significantly modifies rumen bacterial community and promote the production of volatile fatty acids, which in turn promotes rumen digestion and metabolism of growing mutton sheep to ensure the supply of nutrients and promote immune health. Furthermore, mulberry leaves supplementation helps regulate the fecal microbial community and reduce the emission of odorous gas (hydrogen sulfide and harmful Benzene, 1, 2, 3, 5-tetramethyl). Unexpectedly, feeding with fermented mulberry leaves could reduce the emission of fecal smelly or harmful volatile gas (ammonia nitrogen, 3-methyl indole, p-Cresol and DL-Lactamide, propyl ether) which is caused by the immediate supplementation with dried mulberry leaves. The present study revealed that added mulberry leaves could promote absorption of animal's nutrients and immune health. Furthermore, the addition of mulberry leaves, especially fermented mulberry leaves, contributed to the optimization of the livestock environment and strengthen environmental protection. These findings provide valuable insights into the potential applications of mulberry leaves as forage in animal husbandry.
Collapse
Affiliation(s)
- Xiaopeng Cui
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212000, Jiangsu, China
| | - Yuxin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Minjuan Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Lijun Bao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Feng Jiao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xinlan Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Wei Qian
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiang Shi
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chao Su
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Yonghua Qian
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Shenzhen Fengnong Holding Co., Ltd, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
5
|
Beckers KF, Schulz CJ, Flanagan JP, Blair RV, Liu CC, Childers GW, Sones JL. Pregnancy-specific shifts in the maternal microbiome and metabolome in the BPH/5 mouse model of superimposed preeclampsia. Physiol Genomics 2025; 57:115-124. [PMID: 39773069 DOI: 10.1152/physiolgenomics.00106.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Preeclampsia (PE) is a life-threatening hypertensive disorder of pregnancy with an incidence rate of up to 8% worldwide. However, the complete pathogenesis is still unknown. Obesity increases the risk of developing PE threefold. To better understand the relationship of maternal risk factors, the BPH/5 mouse was described as a model of superimposed PE. Previous research demonstrated that adult BPH/5 female mice have an adverse cardiometabolic phenotype characterized by hypertension, obesity with increased white adipose tissue, and dyslipidemia, exaggerated by pregnancy. We hypothesize that BPH/5 mice have gut dysbiosis characterized by changes in alpha and beta diversity of bacterial community structure as well as perturbed short-chain fatty acids (SCFAs) compared with controls in pregnancy. Fecal samples were used for Illumina sequencing of 16S v4 rRNA amplicons. Microbial community composition of the pregnant BPH/5 mice compared with C57 controls was different using permutational multivariate analysis of variance (PERMANOVA) with Bray-Curtis dissimilarity. Alpha diversity was increased in pregnant BPH/5 dams compared with controls. Alistipes and Helicobacter were increased, whereas Bacteroides, Lactobacillus, Parasutterella, and Parabacteroides were decreased compared with controls. Fecal SCFAs were not different between groups, but BPH/5 serum acetic and butyric acids were decreased, whereas isobutyric and isovaleric acids were increased specifically in pregnancy. BPH/5 pregnant colons had decreased expression of free fatty acid receptor, GPR41. In conclusion, the BPH/5 maternal fecal microbiome demonstrates microbial dysbiosis characterized by community structure and diversity changes before and after the onset of pregnancy. Gut dysbiosis may be a key mechanism linking SCFA signaling and obesity to the BPH/5 PE-like phenotype.NEW & NOTEWORTHY This is the first time the pregnant fecal microbiome has been identified in the BPH/5 spontaneous mouse model of superimposed PE. Community composition changed with the onset of pregnancy in this model. BPH/5 showed an altered colonic signaling with decreased GPR41 expression, suggesting that gut dysbiosis may link SCFA signaling to the PE phenotype. This data highlights the importance of the maternal obesogenic gut microbiome in pregnancy.
Collapse
Affiliation(s)
- Kalie F Beckers
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States
- Division of Veterinary Medicine, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States
| | - Christopher J Schulz
- Department of Biological Sciences, Southeastern Louisiana University, Hammond, Louisiana, United States
| | - Juliet P Flanagan
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Robert V Blair
- Division of Veterinary Medicine, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States
| | - Chin-Chi Liu
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Gary W Childers
- Department of Biological Sciences, Southeastern Louisiana University, Hammond, Louisiana, United States
| | - Jenny L Sones
- Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| |
Collapse
|
6
|
Chen M, Li Y, Zhai Z, Wang H, Lin Y, Chang F, Ge S, Sun X, Wei W, Wang D, Zhang M, Chen R, Yu H, Feng T, Huang X, Cheng D, Liu J, Di W, Hao Y, Yin P, Tang P. Bifidobacterium animalis subsp. lactis A6 ameliorates bone and muscle loss via modulating gut microbiota composition and enhancing butyrate production. Bone Res 2025; 13:28. [PMID: 40000617 PMCID: PMC11862215 DOI: 10.1038/s41413-024-00381-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 02/27/2025] Open
Abstract
Systematic bone and muscle loss is a complex metabolic disease, which is frequently linked to gut dysfunction, yet its etiology and treatment remain elusive. While probiotics show promise in managing diseases through microbiome modulation, their therapeutic impact on gut dysfunction-induced bone and muscle loss remains to be elucidated. Employing dextran sulfate sodium (DSS)-induced gut dysfunction model and wide-spectrum antibiotics (ABX)-treated mice model, our study revealed that gut dysfunction instigates muscle and bone loss, accompanied by microbial imbalances. Importantly, Bifidobacterium animalis subsp. lactis A6 (B. lactis A6) administration significantly ameliorated muscle and bone loss by modulating gut microbiota composition and enhancing butyrate-producing bacteria. This intervention effectively restored depleted butyrate levels in serum, muscle, and bone tissues caused by gut dysfunction. Furthermore, butyrate supplementation mitigated musculoskeletal loss by repairing the damaged intestinal barrier and enriching beneficial butyrate-producing bacteria. Importantly, butyrate inhibited the NF-κB pathway activation, and reduced the secretion of corresponding inflammatory factors in T cells. Our study highlights the critical role of dysbiosis in gut dysfunction-induced musculoskeletal loss and underscores the therapeutic potential of B. lactis A6. These discoveries offer new microbiome directions for translational and clinical research, providing promising strategies for preventing and managing musculoskeletal diseases.
Collapse
Affiliation(s)
- Ming Chen
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Zhengyuan Zhai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Hui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yuan Lin
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feifan Chang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Siliang Ge
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xinyu Sun
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Wei Wei
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Duanyang Wang
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingming Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ruijing Chen
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Haikuan Yu
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Taojin Feng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Dongliang Cheng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Jiang Liu
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenxuan Di
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Pengbin Yin
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| |
Collapse
|
7
|
Bascuñán KA, Araya M, Rodríguez JM, Roncoroni L, Elli L, Alvarez JDPL, Valenzuela R. Interplay of n-3 Polyunsaturated Fatty Acids, Intestinal Inflammation, and Gut Microbiota in Celiac Disease Pathogenesis. Nutrients 2025; 17:621. [PMID: 40004950 PMCID: PMC11858531 DOI: 10.3390/nu17040621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Celiac disease (CD) is a chronic autoimmune disorder driven by both genetic and environmental factors, with the HLA DQ2/DQ8 genotypes playing a central role in its development. Despite the genetic predisposition, only a small percentage of individuals carrying these genotypes develop the disease. Gluten, a protein found in wheat, rye, and barley, is the primary environmental trigger, but other factors, such as the intestinal microbiota, may also contribute to disease progression. While the gluten-free diet (GFD) remains the cornerstone of treatment, many CD patients experience persistent inflammation and gut dysbiosis, leading to ongoing symptoms and complications. This chronic inflammation, which impairs nutrient absorption, increases the risk of malnutrition, anemia, and other autoimmune disorders. Recent studies have identified an altered gut microbiota in CD patients, both on and off the GFD, highlighting the potential role of the microbiota in disease pathogenesis. An emerging area of interest is the supplementation of n-3 polyunsaturated fatty acids (PUFAs), known for their anti-inflammatory properties, as a potential therapeutic strategy. n-3 PUFAs, found in fish oil and certain plant oils, modulate the immune cell function and cytokine production, making them a promising intervention for controlling chronic inflammation in CD. This review explores the current understanding of n-3 PUFAs' effects on the gut microbiota's composition and inflammation in CD, with the goal of identifying new avenues for complementary treatments to improve disease management.
Collapse
Affiliation(s)
- Karla A. Bascuñán
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (K.A.B.); (J.D.P.L.A.)
| | - Magdalena Araya
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile (J.M.R.)
| | - Juan Manuel Rodríguez
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile (J.M.R.)
| | - Leda Roncoroni
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.R.); (L.E.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.R.); (L.E.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | | | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (K.A.B.); (J.D.P.L.A.)
| |
Collapse
|
8
|
Dardi P, Coutinho CP, de Oliveira S, Teixeira SA, Gacek RRF, Purgatto E, Vinolo MAR, Muscará MN, Rossoni LV. Changes in the intestinal microbiota induced by the postnatal environment and their association with hypertension. Pharmacol Res 2025; 212:107621. [PMID: 39848350 DOI: 10.1016/j.phrs.2025.107621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
It has been established that cross-fostering impacts the development of hypertension in spontaneously hypertensive rats (SHR). However, the ability of the cross-fostering protocol to shape gut microbiota profile in SHR and impact hypertension is not known. In this sense, the current study explored the influence of normotensive and hypertensive postnatal environments on the intestinal microbiota structure, composition, and functional capacity of SHR and Wistar rats. Our findings revealed significant differences in the microbiota's composition and its metabolic activity in young non-fostered SHR (SS) vs. Wistar (WW) rats, even before hypertension onset, characterized by a reduction of the "low-abundance" bacterial genera, a diminished availability of fecal butyrate and elevated hydrogen sulfide (H2S) production by the SS gut microbiota. Despite influencing the microbiota of both strains, cross-fostering did not fully replicate the microbiota composition of the naturally reared groups in the SHR nursed by Wistar mothers (SW), or in the Wistar rats breastfed by SHR mothers (WS). The SW group had fewer significant genera identified at the Partial Least Squares Discriminant Analysis (PLS-DA), despite resembling the genera profile identified in the normotensive group. While sharing bacterial genera with both SS and WW groups, the WS group is distinguished by its unique microbial composition, particularly by a greater diversity of the 'low-abundance' bacterial genera. Moreover, decreased systolic blood pressure was observed in the SW group compared to the SS group in adulthood. Thus, we could establish a link between microbiota composition and hypertension development, associating it with the loss of the "low-abundance" bacterial taxa. Our data suggest that the postnatal environment is pivotal to promoting gut microbiota compositional changes and contributes to hypertension development.
Collapse
Affiliation(s)
- Patrizia Dardi
- Laboratory of Vascular Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Camille Perella Coutinho
- Department of Food and Experimental Nutrition /FoRC - Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Simone Aparecida Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Eduardo Purgatto
- Department of Food and Experimental Nutrition /FoRC - Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Marcelo Nicolás Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luciana Venturini Rossoni
- Laboratory of Vascular Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
9
|
Ismail HM, Perera D, Mandal R, DiMeglio LA, Evans-Molina C, Hannon T, Petrosino J, Javornik Cregeen S, Schmidt NW. Gut Microbial Changes Associated With Obesity in Youth With Type 1 Diabetes. J Clin Endocrinol Metab 2025; 110:364-373. [PMID: 39078977 PMCID: PMC11747672 DOI: 10.1210/clinem/dgae529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
CONTEXT Obesity is prevalent in type 1 diabetes (T1D) and is problematic with higher risk for diabetes complications. It is unknown to what extent gut microbiome changes are associated with obesity and T1D. OBJECTIVE This work aimed to describe the gut microbiome and microbial metabolite changes associated with obesity in T1D. We hypothesized statistically significant gut microbial and metabolite differences in lean T1D youth (body mass index [BMI]: 5%-<85%) vs those with obesity (BMI: ≥95%). METHODS We analyzed stool samples for gut microbial (using metagenomic shotgun sequencing) and short-chain fatty acid (SCFA) differences in lean (n = 27) and obese (n = 21) T1D youth in a pilot study. The mean ± SD age was 15.3 ± 2.2 years, glycated hemoglobin A1c 7.8 ± 1.3%, diabetes duration 5.1 ± 4.4 years, 42.0% female, and 94.0% were White. RESULTS Bacterial community composition showed between sample diversity differences (β-diversity) by BMI group (P = .013). There was a higher ratio of Prevotella to Bacteroides in the obese group (P = .0058). There was a differential distribution of significantly abundant taxa in either the lean or obese groups, including increased relative abundance of Prevotella copri, among other taxa in the obese group. Functional profiling showed an upregulation of branched-chain amino acid (BCAA) biosynthesis in the obese group and upregulation of BCAA degradation, tyrosine metabolism, and secondary bile acid biosynthesis in the lean group. Stool SCFAs were higher in the obese vs the lean group (P < .05 for all). CONCLUSION Our findings identify a gut microbiome and microbial metabolite signature associated with obesity in T1D. These findings could help identify gut microbiome-targeted therapies to manage obesity in T1D.
Collapse
Affiliation(s)
- Heba M Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dimuthu Perera
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rabindra Mandal
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Linda A DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tamara Hannon
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joseph Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sara Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nathan W Schmidt
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Heras-Molina A, Estellé J, Vázquez-Gómez M, López-García A, Pesantez-Pacheco JL, Astiz S, Garcia-Contreras C, Escudero R, Isabel B, Gonzalez-Bulnes A, Óvilo C. The impact of host genetics on porcine gut microbiota composition excluding maternal and postnatal environmental influences. PLoS One 2024; 19:e0315199. [PMID: 39652543 PMCID: PMC11627362 DOI: 10.1371/journal.pone.0315199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
The gut microbiota of the pig is being increasingly studied due to its implications for host homeostasis and the importance of the pig as a meat source and biomedical model of human diseases. However, most studies comparing the microbiome between different breeds do not consider the influence of maternal environment during the colonization of the microbiota. The aim of the present study was to compare the gut microbiota during postnatal growth between two pig genotypes (purebred Iberian vs. crossbreds Iberian x Large White pigs), gestated in a single maternal environment (pure Iberian mothers) inseminated with heterospermic semen. Postnatally, piglets were maintained in the same environmental conditions, and their microbiota was studied at 60 and 210 days old. Results showed that age had the greatest influence on alpha and beta diversity, and genotype also affected beta diversity at both ages. There were differences in the microbiome profile between genotypes at the ASV and genus levels when jointly analyzing the total number of samples, which may help to explain phenotypical differences. When each time-point was analyzed individually, there were more differences at 210 days-old than 60 days-old. Fecal short-chain fatty acids (SCFA) were also affected by age, but not by genotype. These results may be a basis for further research on host genotype interactions with the gut microbiota.
Collapse
Affiliation(s)
- Ana Heras-Molina
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n, Madrid, Spain
- CSIC-INIA, Madrid, Spain
| | - Jordi Estellé
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Marta Vázquez-Gómez
- Sorbonne université, INSERM, Nutrition et obésités: approaches systémiques, Nutriomics, Paris, France
| | | | - José-Luis Pesantez-Pacheco
- CSIC-INIA, Madrid, Spain
- School of Veterinary Medicine and Zootechnics, Faculty of Agricultural Sciences, University of Cuenca, Cuenca, Ecuador
| | | | | | - Rosa Escudero
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n, Madrid, Spain
| | - Beatriz Isabel
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n, Madrid, Spain
| | - Antonio Gonzalez-Bulnes
- Faculty of Veterinary Medicine, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | | |
Collapse
|
11
|
Manokasemsan W, Jariyasopit N, Poungsombat P, Kaewnarin K, Wanichthanarak K, Kurilung A, Duangkumpha K, Limjiasahapong S, Pomyen Y, Chaiteerakij R, Tansawat R, Srisawat C, Sirivatanauksorn Y, Sirivatanauksorn V, Khoomrung S. Quantifying fecal and plasma short-chain fatty acids in healthy Thai individuals. Comput Struct Biotechnol J 2024; 23:2163-2172. [PMID: 38827233 PMCID: PMC11141283 DOI: 10.1016/j.csbj.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are involved in important physiological processes such as gut health and immune response, and changes in SCFA levels can be indicative of disease. Despite the importance of SCFAs in human health and disease, reference values for fecal and plasma SCFA concentrations in healthy individuals are scarce. To address this gap in current knowledge, we developed a simple and reliable derivatization-free GC-TOFMS method for quantifying fecal and plasma SCFAs in healthy individuals. We targeted six linear- and seven branched-SCFAs, obtaining method recoveries of 73-88% and 83-134% in fecal and plasma matrices, respectively. The developed methods are simpler, faster, and more sensitive than previously published methods and are well suited for large-scale studies. Analysis of samples from 157 medically confirmed healthy individuals showed that the total SCFAs in the feces and plasma were 34.1 ± 15.3 µmol/g and 60.0 ± 45.9 µM, respectively. In fecal samples, acetic acid (Ace), propionic acid (Pro), and butanoic acid (But) were all significant, collectively accounting for 89% of the total SCFAs, whereas the only major SCFA in plasma samples was Ace, constituting of 93% of the total plasma SCFAs. There were no statistically significant differences in the total fecal and plasma SCFA concentrations between sexes or among age groups. The data revealed, however, a positive correlation for several nutrients, such as carbohydrate, fat, iron from vegetables, and water, to most of the targeted SCFAs. This is the first large-scale study to report SCFA reference intervals in the plasma and feces of healthy individuals, and thereby delivers valuable data for microbiome, metabolomics, and biomarker research.
Collapse
Affiliation(s)
- Weerawan Manokasemsan
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Narumol Jariyasopit
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Patcha Poungsombat
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Khwanta Kaewnarin
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- SingHealth Duke-NUS Institute of Biodiversity Medicine, National Cancer Centre Singapore, Singapore
| | - Kwanjeera Wanichthanarak
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Alongkorn Kurilung
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kassaporn Duangkumpha
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Suphitcha Limjiasahapong
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Yotsawat Pomyen
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Roongruedee Chaiteerakij
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Division of Gastroenterology, Department of Medicine, Faculty of Medicine Chulalongkorn University, Chulalongkorn University, Bangkok, Thailand
| | - Rossarin Tansawat
- Thailand Metabolomics Society, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Chulalongkorn University, Bangkok, Thailand
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Yongyut Sirivatanauksorn
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vorapan Sirivatanauksorn
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Sakda Khoomrung
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
12
|
Szczuko M, Duliban G, Drozd A, Sochaczewska D, Pokorska-Niewiada K, Ziętek M. The Association of Short-Chain Fatty Acids with the Occurrence of Gastrointestinal Symptoms in Infants. Int J Mol Sci 2024; 25:12487. [PMID: 39684199 DOI: 10.3390/ijms252312487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are produced by the fermentation of undigested polysaccharides; they are a group of metabolites resulting from the activity of intestinal bacteria. The main SCFAs are acetic, butyric, propionic, valeric, and caproic acid, and their levels and proportions depend on various factors. The aim of this study was to investigate the relationship between the concentration of SCFAs and the occurrence of specific gastrointestinal symptoms in infants. This study was conducted using faecal samples obtained at 1, 3, 6, and 12 months of age. The SCFA content was measured using gas chromatography. At 1 month, an association was found between butyric acid and flatulence. At 3 months, an association was found between butyric acid and flatulence/gas and between 3,4-methylovaleric acid and mucus in the stool. At 6 months, an association was found between butyric and valeric acids and flatulence. By 12 months, the gastrointestinal symptoms had decreased significantly. This study confirms that there is an association between SCFA levels and the presence of bloating, gas, mucus in the stool, and constipation in the gastrointestinal tract. Higher levels of butyric and valeric acids may lead to an increase in troublesome symptoms, such as flatulence and gas, in the first few months of life but are not associated with the occurrence of intestinal colic. The level of 3,4-methylovaleric acid is associated with the presence of allergies, whereas a decrease in acetic acid and an increase in isovaleric acid may exacerbate defecation problems in infants.
Collapse
Affiliation(s)
- Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
- Department of Human Nutrition and Bromatology, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Gabriela Duliban
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Arleta Drozd
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Diana Sochaczewska
- Department of Neonatology, Pomeranian Medical University in Szczecin, 72-009 Police, Poland
| | - Kamila Pokorska-Niewiada
- Department of Toxicology, Dairy Technology and Food Storage, West Pomeranian University of Technology in Szczecin, 71-459 Szczecin, Poland
| | - Maciej Ziętek
- Department of Perinatology, Obstetrics and Gynecology Pomeranian Medical University in Szczecin, 72-009 Police, Poland
| |
Collapse
|
13
|
Kim SJ, Lee HK, Kang KS, Lee MG, Shin MS. Korean Red Ginseng Polysaccharides Enhance Intestinal IgA Production and Barrier Function via Peyer's Patch Activation in Mice. Nutrients 2024; 16:3816. [PMID: 39599603 PMCID: PMC11597691 DOI: 10.3390/nu16223816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Natural products are gaining attention for their potential benefits in gastrointestinal health. Plant-derived polysaccharides are essential for boosting intestinal immunity and maintaining gut homeostasis. This study investigated the effects of Korean red ginseng polysaccharides (KRG-P) on intestinal homeostasis including IgA and SCFA production and mucosal barrier integrity. Methods: Mice were orally administered KRG-P at doses of 50 mg/kg or 200 mg/kg for 10 days. Fecal IgA levels were measured on days 3, 5, and 11 and IgA from cultured Peyer's patch cells from KRG-P-treated mice were analyzed. Additionally, mRNA and protein expression levels of α-defensin, lysozyme, and E-cadherin in the small intestine were examined. Short-chain fatty acids (SCFAs) content in the cecum was also assessed. Results: KRG-P-treated groups showed a significant increase in fecal IgA levels on days 5 and 11, with no notable change on day 3. Cultured Peyer's patch cells from mice demonstrated heightened IgA production. Additionally, KRG-P administration upregulated α-defensin and lysozyme mRNA expression, along with elevated protein expression of E-cadherin, α-defensin, and lysozyme, in the small intestine. KRG-P treatment also led to increased cecal SCFA levels, including acetate, butyrate, and propionate. Conclusions: KRG-P may promote intestinal homeostasis and host defense mechanisms by activating immune cells in Peyer's patches, stimulating IgA production, enhancing antimicrobial peptide expression, and modulating gut microbiota metabolism through increased SCFA production.
Collapse
Affiliation(s)
- Sung Jin Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.J.K.); (K.S.K.)
| | - Hae-Kyung Lee
- Avison Biomedical Research Center, Yonsei University, Seoul 03722, Republic of Korea;
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.J.K.); (K.S.K.)
| | - Mi-Gi Lee
- Bio-Center, Gyeonggi-do Business and Science Accelerator, Suwon 16229, Republic of Korea
| | - Myoung-Sook Shin
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.J.K.); (K.S.K.)
| |
Collapse
|
14
|
Vidal PM, Brockie S, Farkas C, Hong J, Zhou C, Fehlings MG. Neuromotor decline is associated with gut dysbiosis following surgical decompression for Degenerative Cervical Myelopathy. Neurobiol Dis 2024; 200:106640. [PMID: 39159895 DOI: 10.1016/j.nbd.2024.106640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Abstract
Degenerative cervical myelopathy (DCM) describes a spectrum of disorders that cause progressive and chronic cervical spinal cord compression. The clinical presentation can be complex and can include locomotor impairment, hand and upper extremity dysfunction, pain, loss of bladder and bowel function, as well as gastrointestinal dysfunction. Once diagnosed, surgical decompression is the recommended treatment for DCM patients with moderate to severe impairment. Our body is composed of a large community of microorganisms, known as the microbiota. Traumatic and non-traumatic spinal cord injuries (SCIs) can induce changes in the gut microbiota and gut microbiota derived metabolites. These changes have been reported as important disease-modifying factors after injury. However, whether gut dysbiosis is associated with functional neurological recovery after surgical decompression has not been examined to date. Here, DCM was induced in C57BL/6 mice by implanting an aromatic polyether material underneath the C5-6 laminae. The extent of gut dysbiosis was assessed by gas chromatography and 16S rRNA sequencing from fecal samples before and after decompression. Neuromotor activity was assessed using the Catwalk test. Our results show that DCM pre- and post- surgical decompression is associated with gut dysbiosis, without altering short chain fatty acids (SCFAs) levels. Significant differences in Clostridia, Verrumicrobiae, Lachnospiracea, Firmicutes, Bacteroidales, and Clostridiaceae were observed between the DCM group (before decompression) and after surgical decompression (2 and 5 weeks). The changes in gut microbiota composition correlated with locomotor features of the Catwalk. For example, a longer duration of ground contact and dysfunctional swing in the forelimbs, were positively correlated with gut dysbiosis. These results show for the first time an association between gut dysbiosis and locomotor deterioration after delayed surgical decompression. Thus, providing a better understanding of the extent of changes in microbiota composition in the setting of DCM pre- and post- surgical decompression.
Collapse
Affiliation(s)
- Pia M Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| | - Sydney Brockie
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Carlos Farkas
- Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Cindy Zhou
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
15
|
Moya AMTM, Alexandrino TD, Morari J, Reguengo LM, Velloso LA, Leal RF, Junior SB, Pereira APA, Pastore GM, Bicas JL, Cazarin CBB. The Consumption of the Fibrous Fraction of Solanum lycocarpum St. Hil. Does Not Preserve the Intestinal Mucosa in TNBS-Induced Rats. Foods 2024; 13:2949. [PMID: 39335878 PMCID: PMC11431493 DOI: 10.3390/foods13182949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Solanum lycocarpum St. Hil. is considered a natural anti-inflammatory. In traditional medicine, it is used to reduce cholesterol levels in the treatment of obesity. Foods capable of conferring a protective and nutritious effect have been used to prevent or attenuate the clinical symptoms of inflammatory bowel diseases. Ulcerative colitis is a multifactorial inflammatory bowel disease. This study investigated the impact of the consumption of the fibrous fraction (FF) and resistant starch (RS) of fruta-do-lobo in an experimental model of colitis induced with the use 2,4,6-trinitrobenzene sulphonic acid (TNBS) in rats. The different colitis groups all experienced decreased weight gain, which could be linked to the inflammatory process (p = 0.603). Additionally, the experimental model led to increased oxidative stress, higher levels of pro-inflammatory cytokines, and the elevated gene expression of these cytokines. Despite this, consuming the fibrous fraction of fruta-do-lobo (RS and FF) did not appear to protect the animals against the inflammatory process. Regarding the expression of TNF-α, only the group treated with the drug mesalamine had a reduced serum level of this inflammatory marker (p = 0.03). Our results showed that the diet containing RS and FF did not protect the intestinal mucosa against TNBS inflammation. New studies on the variation in the time of consumption or the supplemented dose of fruta-do-lobo fibers could help to elucidate their effects in protecting the mucosa.
Collapse
Affiliation(s)
- Amanda Maria Tomazini Munhoz Moya
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Thaís Dolfini Alexandrino
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Joseane Morari
- School of Medical Sciences, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Campinas 13083-887, São Paulo, Brazil; (J.M.); (L.A.V.); (R.F.L.)
| | - Livia Mateus Reguengo
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Licio Augusto Velloso
- School of Medical Sciences, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Campinas 13083-887, São Paulo, Brazil; (J.M.); (L.A.V.); (R.F.L.)
| | - Raquel Franco Leal
- School of Medical Sciences, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Campinas 13083-887, São Paulo, Brazil; (J.M.); (L.A.V.); (R.F.L.)
| | - Stanislau Bogusz Junior
- São Carlos Institute of Chemistry (IQSC), University of São Paulo (USP), São Carlos 13566-590, São Paulo, Brazil;
| | - Ana Paula Aparecida Pereira
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
- Faculty of Nutrition, Federal University of Mato Grosso, Avenida Fernando Correa da Costa, 2367, Boa Esperança, Cuiabá 78068-600, Mato Grosso, Brazil
| | - Glaucia Maria Pastore
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Juliano Lemos Bicas
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Cinthia Baú Betim Cazarin
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| |
Collapse
|
16
|
Zhang L, Zhu T, Wang Y, Zhang B, Zhang H, Han L, Liu E, Fu Z. Effects of in vitro simulated digestion and fecal fermentation on the structure and regulating the glucose and lipid activity of a polysaccharide from Mori Folium. Int J Biol Macromol 2024; 280:135595. [PMID: 39276886 DOI: 10.1016/j.ijbiomac.2024.135595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/24/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Mori folium, as a homologous drug-food, has hypoglycemic and lipid-lowering activity. Polysaccharides are the main bioactive ingredient of the Mori folium that exhibit diverse biological activities. In this study, a homogeneous polysaccharide (MP4) was purified and characterized from Mori folium. The changes of MP4 affected by saliva, simulated gastrointestinal juice, and human fecal fermentation, including physicochemical property or its bioactivity, were systematically investigated. Meanwhile, the influence of fermentation on the bioactivity were evaluated. The results showed that the backbone of MP4 is mainly composed of →4)-α-D-GalpA-(1→ residues. The molecular weight, the levels of reducing sugar content and free monosaccharides of MP4 exhibited no significant differences indicating that gastrointestinal digestion has a minimal effect on the physicochemical characteristics of MP4. However, during in vitro gut microbiota fermentation, MP4 are significantly degraded and utilized by gut microbiota, showing increased the production of short-chain fatty acids, notably acetic acid and propionic acid. The relative abundance of beneficial bacteria such as Bacteroidetes and Actinobacteria were significantly increased, whereas the levels of pathogenic bacteria such as Fusobacteria and Megamonas were significantly decreased, which changed the composition of the gut microbiota. The Firmicutes/Bacteroides ratio was also decreased significantly. Interestingly, after in vitro fermentation, the α-glucosidase inhibitory activity was increased, the lipase inhibitory activity and cholesterol adsorption activity was decreased. Correlation analysis showed that the relative abundance of some bacteria was significantly correlated with the bioactivities. These results provide a basis for the development of Mori folium polysaccharides as functional probiotic products.
Collapse
Affiliation(s)
- Lingyu Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Tongtong Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China
| | - Ying Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Boli Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Lifeng Han
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Erwei Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Zhifei Fu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai district, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
17
|
Archana, Gupta AK, Noumani A, Panday DK, Zaidi F, Sahu GK, Joshi G, Yadav M, Borah SJ, Susmitha V, Mohan A, Kumar A, Solanki PR. Gut microbiota derived short-chain fatty acids in physiology and pathology: An update. Cell Biochem Funct 2024; 42:e4108. [PMID: 39228159 DOI: 10.1002/cbf.4108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024]
Abstract
Short-chain fatty acids (SCFAs) are essential molecules produced by gut bacteria that fuel intestinal cells and may also influence overall health. An imbalance of SCFAs can result in various acute and chronic diseases, including diabetes, obesity and colorectal cancer (CRC). This review delves into the multifaceted roles of SCFAs, including a brief discussion on their source and various gut-residing bacteria. Primary techniques used for detection of SCFAs, including gas chromatography, high-performance gas chromatography, nuclear magnetic resonance and capillary electrophoresis are also discussed through this article. This review study also compiles various synthesis pathways of SCFAs from diverse substrates such as sugar, acetone, ethanol and amino acids. The different pathways through which SCFAs enter cells for immune response regulation are also highlighted. A major emphasis is the discussion on diseases associated with SCFA dysregulation, such as anaemia, brain development, CRC, depression, obesity and diabetes. This includes exploring the relationship between SCFA levels across ethnicities and their connection with blood pressure and CRC. In conclusion, this review highlights the critical role of SCFAs in maintaining gut health and their implications in various diseases, emphasizing the need for further research on SCFA detection, synthesis and their potential as diagnostic biomarkers. Future studies of SCFAs will pave the way for the development of novel diagnostic tools and therapeutic strategies for optimizing gut health and preventing diseases associated with SCFA dysregulation.
Collapse
Affiliation(s)
- Archana
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Abhijeet Kumar Gupta
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Ashab Noumani
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Dharmendra Kumar Panday
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Fareen Zaidi
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Gaurav Kumar Sahu
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Gunjan Joshi
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Manisha Yadav
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Shikha Jyoti Borah
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Vanne Susmitha
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Anand Mohan
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Anil Kumar
- National Institute of Immunology, New Delhi, India
| | - Pratima R Solanki
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
18
|
Hou Y, Luo S, Li Z, Zhang H, Chen T, Liu C. Extrusion treatment of rice bran insoluble fiber generates specific niches favorable for Bacteroides during in vitro fermentation. Food Res Int 2024; 190:114599. [PMID: 38945569 DOI: 10.1016/j.foodres.2024.114599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024]
Abstract
To investigate the morphological changes of insoluble fiber and their effects on microbiota modulation, particularly Bacteroides, rice bran insoluble fibers were extruded at different feed moisture levels (E20, E40, and E60). The physicochemical properties and SEM revealed that E20 exhibited the highest water holding capacity and displayed the most fragmented edges. E40 had the highest swelling holding capacity and displayed the most lamellar gaps. E60 showed minimal change in physicochemical properties but had a rough surface. After 48h fermentation, E40 showed the highest levels of Bacteroides and SCFAs. E20 and E60 resulted in a modest increase in Bacteroides abundance. SEM showed that bacteria were attached to fragmented edges, loosened lamellar gaps, and rough surfaces of the extruded insoluble fibers. The results suggested that Bacteroides gained a competitive advantage within the extrusion treatment created structural changes. Extrusion treatment can be used to generate specific niches favorable for Bacteroides.
Collapse
Affiliation(s)
- Yaqin Hou
- The State Key Laboratory of Food Science and Resources, School of Food Science & Technology, Nanchang University, 235 East Nanjing Road, Nanchang, Jiangxi 330047, China
| | - Shunjing Luo
- The State Key Laboratory of Food Science and Resources, School of Food Science & Technology, Nanchang University, 235 East Nanjing Road, Nanchang, Jiangxi 330047, China
| | - Zhongxia Li
- BYHEALTH Institute of Nutrition & Health, Guangzhou 510663, China
| | - Huibin Zhang
- The State Key Laboratory of Food Science and Resources, School of Food Science & Technology, Nanchang University, 235 East Nanjing Road, Nanchang, Jiangxi 330047, China
| | - Tingting Chen
- The State Key Laboratory of Food Science and Resources, School of Food Science & Technology, Nanchang University, 235 East Nanjing Road, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation Co., Ltd., Nanchang University, Luozhu Road, Xiaolan Economic and Technological Development Zone, Nanchang, Jiangxi 330200, China.
| | - Chengmei Liu
- The State Key Laboratory of Food Science and Resources, School of Food Science & Technology, Nanchang University, 235 East Nanjing Road, Nanchang, Jiangxi 330047, China; International Institute of Food Innovation Co., Ltd., Nanchang University, Luozhu Road, Xiaolan Economic and Technological Development Zone, Nanchang, Jiangxi 330200, China.
| |
Collapse
|
19
|
Stothart MR, McLoughlin PD, Medill SA, Greuel RJ, Wilson AJ, Poissant J. Methanogenic patterns in the gut microbiome are associated with survival in a population of feral horses. Nat Commun 2024; 15:6012. [PMID: 39039075 PMCID: PMC11263349 DOI: 10.1038/s41467-024-49963-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Gut microbiomes are widely hypothesised to influence host fitness and have been experimentally shown to affect host health and phenotypes under laboratory conditions. However, the extent to which they do so in free-living animal populations and the proximate mechanisms involved remain open questions. In this study, using long-term, individual-based life history and shallow shotgun metagenomic sequencing data (2394 fecal samples from 794 individuals collected between 2013-2019), we quantify relationships between gut microbiome variation and survival in a feral population of horses under natural food limitation (Sable Island, Canada), and test metagenome-derived predictions using short-chain fatty acid data. We report detailed evidence that variation in the gut microbiome is associated with a host fitness proxy in nature and outline hypotheses of pathogenesis and methanogenesis as key causal mechanisms which may underlie such patterns in feral horses, and perhaps, wild herbivores more generally.
Collapse
Affiliation(s)
- Mason R Stothart
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Biology, University of Oxford, Oxford, United Kingdom.
| | - Philip D McLoughlin
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sarah A Medill
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ruth J Greuel
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alastair J Wilson
- Centre for Ecology and Conservation, University of Exeter, Penryn, United Kingdom
| | - Jocelyn Poissant
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
20
|
Chen R, Zhang H, Cai J, Cai M, Dai T, Liu Y, Wu J. Germination-Induced Enhancement of Brown Rice Noodle Nutritional Profile and Gut Microbiota Modulation. Foods 2024; 13:2279. [PMID: 39063363 PMCID: PMC11275603 DOI: 10.3390/foods13142279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
This study explored how germination influences the starch digestion and intestinal fermentation characteristics of brown rice noodle. The study began with in vitro starch digestion tests to assess how germination affects starch digestibility in brown rice noodles, revealing an increase in rapidly digestible starch content and a decrease in resistant starch content. Subsequently, an in vitro human fecal fermentation model was used to simulate the human intestinal environment, showing that germination altered pH levels and the production of short-chain fatty acids, particularly by increasing propionate while decreasing acetate and butyrate. Additionally, the study noted a decrease in gut microbiota diversity following fermentation, accompanied by an increase in Megamonas growth and a decrease in Bacteroides and Bifidobacterium. In conclusion, these findings suggest that germination could enhance the nutritional value and intestinal probiotic properties of brown rice noodles. This research contributes valuable insights into the role of germination in improving the nutritional properties of rice-based products and provides a foundation for further exploration into the development of health-promoting rice noodles.
Collapse
Affiliation(s)
- Ruiyun Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- Jiangxi General Institute of Testing and Certification, Nanchang 330052, China
| | - Huibin Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jiamei Cai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Mingxi Cai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Taotao Dai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Yunfei Liu
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang 330096, China
| | - Jianyong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
21
|
Lin X, Peng Y, Guo Z, He W, Guo W, Feng J, Lu L, Liu Q, Xu P. Short-chain fatty acids suppresses astrocyte activation by amplifying Trp-AhR-AQP4 signaling in experimental autoimmune encephalomyelitis mice. Cell Mol Life Sci 2024; 81:293. [PMID: 38976012 PMCID: PMC11335219 DOI: 10.1007/s00018-024-05332-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 07/09/2024]
Abstract
The function of astrocytes in response to gut microbiota-derived signals has an important role in the pathophysiological processes of central nervous system (CNS) diseases. However, the specific effects of microbiota-derived metabolites on astrocyte activation have not been elucidated yet. Experimental autoimmune encephalomyelitis (EAE) was induced in female C57BL/6 mice as a classical MS model. The alterations of gut microbiota and the levels of short-chain fatty acids (SCFAs) were assessed after EAE induction. We observed that EAE mice exhibit low levels of Allobaculum, Clostridium_IV, Clostridium_XlVb, Lactobacillus genera, and microbial-derived SCFAs metabolites. SCFAs supplementation suppressed astrocyte activation by increasing the level of tryptophan (Trp)-derived AhR ligands that activating the AhR. The beneficial effects of SCFAs supplementation on the clinical scores, histopathological alterations, and the blood brain barrier (BBB)-glymphatic function were abolished by intracisterna magna injection of AAV-GFAP-shAhR. Moreover, SCFAs supplementation suppressed the loss of AQP4 polarity within astrocytes in an AhR-dependent manner. Together, SCFAs potentially suppresses astrocyte activation by amplifying Trp-AhR-AQP4 signaling in EAE mice. Our study demonstrates that SCFAs supplementation may serve as a viable therapy for inflammatory disorders of the CNS.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Astrocytes/metabolism
- Astrocytes/drug effects
- Fatty Acids, Volatile/pharmacology
- Fatty Acids, Volatile/metabolism
- Receptors, Aryl Hydrocarbon/metabolism
- Mice
- Mice, Inbred C57BL
- Tryptophan/metabolism
- Tryptophan/pharmacology
- Female
- Signal Transduction/drug effects
- Aquaporin 4/metabolism
- Aquaporin 4/genetics
- Gastrointestinal Microbiome/drug effects
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/drug effects
Collapse
Affiliation(s)
- Xiuli Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yufeng Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Zhimei Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Wuhui He
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Junmin Feng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Lin Lu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Qin Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China.
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
22
|
Tao M, Cao K, Pu X, Hou Y, He L, Liu W, Ren Y, Yang X. Cadmium exposure induces changes in gut microbial composition and metabolic function in long-tailed dwarf hamsters, Cricetulus longicaudatus. Ecol Evol 2024; 14:e11682. [PMID: 38966245 PMCID: PMC11222731 DOI: 10.1002/ece3.11682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
Numerous studies have demonstrated that exposure to cadmium disrupts the diversity and composition of the gut microbiota, resulting in damage to organ tissue. However, there remains a lack of comprehensive understanding regarding the broader ecological reality associated with this phenomenon. In this study, we conducted a thorough evaluation of the effects of different concentrations of Cd (6, 12, 24, and 48 mg/L) over a period of 35 consecutive days on the organ viscera and the gut microbiota of long-tailed dwarf hamsters, Cricetulus longicaudatus (Rodentia: Cricetidae), using histopathological analysis, 16S rDNA, and metagenome sequencing. Our findings revealed that the results suggest that Cd exposure induced liver, spleen, and kidney damage, potentially leading to increased intestinal permeability and inflammation. These alterations were accompanied by significant perturbations in the gut microbiota composition, particularly affecting potentially pathogenic bacteria such as Prevotella and Treponema within the gut ecosystem. Consequently, host susceptibility to underlying diseases was heightened due to these changes. Notably though, Cd exposure did not significantly impact the overall structure of the gut microbiota itself. Additionally, Cd exposure induced significant changes in the metabolic functions, with the pathways related to disease and environmental information processing notably enhanced, possibly indicating stronger innate defense mechanisms against external injuries among wild mammals exposed to Cd. This study offers a novel approach to comprehensively evaluate the significant impact of Cd pollution on ecosystems by investigating both structural and functional alterations in the digestive system, as well as disruptions in intestinal flora among wild mammals.
Collapse
Affiliation(s)
- Mengfan Tao
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Kanglin Cao
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Xinsheng Pu
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Yu Hou
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Lei He
- Shanxi Forestry and Grassland General Engineering StationTaiyuanChina
| | - Wei Liu
- Shanxi Forestry and Grassland General Engineering StationTaiyuanChina
| | - Yue Ren
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Xin'gen Yang
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| |
Collapse
|
23
|
Bukavina L, Ginwala R, Eltoukhi M, Sindhani M, Prunty M, Geynisman DM, Ghatalia P, Valentine H, Calaway A, Correa AF, Brown JR, Mishra K, Plimack ER, Kutikov A, Ghannoum M, Elshaer M, Retuerto M, Ponsky L, Uzzo RG, Abbosh PH. Role of Gut Microbiome in Neoadjuvant Chemotherapy Response in Urothelial Carcinoma: A Multi-institutional Prospective Cohort Evaluation. CANCER RESEARCH COMMUNICATIONS 2024; 4:1505-1516. [PMID: 38747616 PMCID: PMC11181990 DOI: 10.1158/2767-9764.crc-23-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/21/2024] [Accepted: 05/09/2024] [Indexed: 05/28/2024]
Abstract
Neoadjuvant chemotherapy (NAC) is linked with clinical advantages in urothelial carcinoma for patients with muscle-invasive bladder cancer (MIBC). Despite comprehensive research into the influence of tumor mutation expression profiles and clinicopathologic factors on chemotherapy response, the role of the gut microbiome (GM) in bladder cancer chemotherapy response remains poorly understood. This study examines the variance in the GM of patients with bladder cancer compared with healthy adults, and investigates GM compositional differences between patients who respond to chemotherapy versus those who exhibit residual disease.Our study reveals distinct clustering, effectively separating the bladder cancer and healthy cohorts. However, no significant differences were observed between chemotherapy responders and nonresponders within community subgroups. Machine learning models based on responder status outperformed clinical variables in predicting complete response (AUC 0.88 vs. AUC 0.50), although no single microbial species emerged as a fully reliable biomarker.The evaluation of short chain fatty acid (SCFA) concentration in blood and stool revealed no correlation with responder status. Still, SCFA analysis showed a higher abundance of Akkermansia (rs = 0.51, P = 0.017) and Clostridia (rs = 0.52, P = 0.018), which correlated with increased levels of detectable fecal isobutyric acid. Higher levels of fecal Lactobacillus (rs = 0.49, P = 0.02) and Enterobacteriaceae (rs = 0.52, P < 0.03) correlated with increased fecal propionic acid.In conclusion, our study constitutes the first large-scale, multicenter assessment of GM composition, suggesting the potential for a complex microbial signature to predict patients more likely to respond to NAC based on multiple taxa. SIGNIFICANCE Our study highlights results that link the composition of the GM to the efficacy of NAC in MIBC. We discovered that patients with higher levels of Bacteroides experienced a worse response to NAC. This microbial signature shows promise as a superior predictor of treatment response over traditional clinical variables. Although preliminary, our findings advocate for larger, more detailed studies to validate these associations.
Collapse
Affiliation(s)
- Laura Bukavina
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Cleveland Clinic Glickman Urologic Institute, Cleveland, Ohio
- Case Western Reserve School of Medicine, Cleveland, Ohio
| | | | | | | | - Megan Prunty
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | | | | | - Adam Calaway
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Jason R. Brown
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Kirtishri Mishra
- Case Western Reserve School of Medicine, Cleveland, Ohio
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | | | | | | | | | - Lee Ponsky
- Case Western Reserve School of Medicine, Cleveland, Ohio
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Philip H. Abbosh
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Albert Einstein Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Song A, Cheng R, Jiang J, Qu H, Wu Z, Qian F, Shen S, Zhang L, Wang Z, Zhao W, Lou Y. Antidepressant-like effects of hyperoside on chronic stress-induced depressive-like behaviors in mice: Gut microbiota and short-chain fatty acids. J Affect Disord 2024; 354:356-367. [PMID: 38492650 DOI: 10.1016/j.jad.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/04/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND The antidepressant effect of hyperoside (HYP), which is the main component of Hypericum perforatum, is not established. This study aimed to determine the effects of HYP on depression. METHODS The antidepressant-like effect of HYP was studied in mice induced by chronic restraint stress (CRS). The effects of HYP on behavior, inflammation, neurotransmitters, gut microbiota, and short-chain fatty acids (SCFAs) were studied in CRS mice. RESULTS HYP improved depressive-like behavior in mice induced by CRS. Nissl staining analysis showed that HYP improved neuronal damage in CRS mice. Western blot (WB) analysis showed that HYP increased the expression levels of BDNF and PSD95 in the hippocampus of CRS mice. The results of ELISA showed that HYP down-regulated the expression levels of IL-6, IL-1β, TNF-α, and CORT in the hippocampus, blood, and intestinal tissues of mice and up-regulated the expression levels of 5-HT and BDNF. Hematoxylin and eosin (HE) staining results indicate that HYP can improve the intestinal histopathological injury of CRS mice. The results of 16S rRNA demonstrated that HYP attenuated the dysbiosis of the gut microbiota of depressed mice, along with altering the concentration of SCFAs. LIMITATIONS In the present study, direct evidence that HYP improves depressive behaviors via gut microbiota and SCFAs is lacking, and only female mice were evaluated, which limits the understanding of the effects of HYP on both sexes. CONCLUSIONS HYP can improve CRS-induced depressive-like behaviors in mice, which is associated with regulating the gut microbiota and SCFAs concentration.
Collapse
Affiliation(s)
- Aoqi Song
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Ru Cheng
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Jingjing Jiang
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Han Qu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenghua Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Shuyu Shen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Liwen Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyu Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjuan Zhao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China..
| | - Yuefen Lou
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai 200434, China.
| |
Collapse
|
25
|
Chen P, Lv H, Du M, Liu W, Che C, Zhao J, Liu H. Bacillus subtilis HW2 enhances growth performance and alleviates gut injury via attenuation of endoplasmic reticulum stress and regulation of gut microbiota in broilers under necrotic enteritis challenge. Poult Sci 2024; 103:103661. [PMID: 38547540 PMCID: PMC11000119 DOI: 10.1016/j.psj.2024.103661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
This study investigated the effects of Bacillus subtilis HW2 on the growth performance, immune response, endoplasmic reticulum (ER) stress, and intestinal health in broilers with necrotic enteritis. Three hundred 1-day-old male Cobb 500 broilers (33.88 ± 2.34 g) were randomly allocated to 5 groups including non-infected control (NC group), basal diet + necrotic enteritis challenge (NE group), basal diet + 1 × 106 CFU/g B. subtilis HW2 + necrotic enteritis challenge (L-Pro group), basal diet + 5 × 106 CFU/g B. subtilis HW2 + necrotic enteritis challenge (M-Pro group), and basal diet + 1 × 107 CFU/g B. subtilis HW2 + necrotic enteritis challenge (H-Pro group), with 6 replicates per group. All broilers except NC group were orally given with sporulated coccidian oocysts at day 14 and Clostridium perfringens from days 19 to 21. Results showed that L-Pro and M-Pro groups improved growth performance and intestinal morphology in necrotic enteritis-challenged broilers, and L-Pro, M-Pro, and H-Pro groups improved intestinal barrier function and immune response and decreased ER stress in necrotic enteritis-challenged broilers. Analysis of the gut microbiota revealed that L-Pro group increased the abundances of Alistipes, Coprobacter, Barnesiella, and Limosilactobacillus, decreased Erysipelatoclostridium abundance on day 42 in necrotic enteritis-challenged broilers. M-Pro group increased Turicibacter abundance on day 28 and the abundances of Alistipes, Barnesiella, and Limosilactobacillus on day 42 in necrotic enteritis-challenged broilers. H-Pro group decreased Romboutsia abundance on day 28 and unidentified_Clostridia abundance on day 42 in necrotic enteritis-challenged broilers. Analysis of short-chain fatty acids (SCFAs) revealed higher isobutyric acid and isovaleric acid levels in L-Pro and M-Pro groups than NE group. Correlation analysis revealed the correlations between the biochemical parameters and gut microbiota as well as SCFAs, especially Romboutsia, Barnesiella, Coprobacter, isobutyric acid, and isovaleric acid. Overall, our results indicated that B. subtilis HW2 supplementation could ameliorate necrotic enteritis infection-induced gut injury. The optimal dietary supplementation dosage of Bacillus subtilis HW2 was 5 × 106 CFU/g.
Collapse
Affiliation(s)
- Peng Chen
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Huimin Lv
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Mengmeng Du
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Weiyong Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chuanyan Che
- College of Animal Science and Technology, Anhui Science and Technology University, Fengyang, 233100, China
| | - Jinshan Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Huawei Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
26
|
Rätsep M, Kilk K, Zilmer M, Kuusik S, Kuus L, Vallas M, Gerulis O, Štšepetova J, Orav A, Songisepp E. Investigation of Effects of Novel Bifidobacterium longum ssp. longum on Gastrointestinal Microbiota and Blood Serum Parameters in a Conventional Mouse Model. Microorganisms 2024; 12:840. [PMID: 38674784 PMCID: PMC11052112 DOI: 10.3390/microorganisms12040840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Representatives of the genus Bifidobacterium are widely used as probiotics to modulate the gut microbiome and alleviate various health conditions. The action mechanisms of probiotics rely on their direct effect on the gut microbiota and the local and systemic effect of its metabolites. The main purpose of this animal experiment was to assess the biosafety of the Bifidobacterium longum strain BIOCC1719. Additional aims were to characterise the influence of the strain on the intestinal microbiota and the effect on several health parameters of the host during 15- and 30-day oral administration of the strain to mice. The strain altered the gut microbial community, thereby altering luminal short-chain fatty acid metabolism, resulting in a shift in the proportions of acetic, butyric, and propionic acids in the faeces and serum of the test group mice. Targeted metabolic profiling of serum revealed the possible ability of the strain to positively affect the hosts' amino acids and bile acids metabolism, as the cholic acid, deoxycholic acid, aspartate, and glutamate concentration were significantly higher in the test group. The tendency to increase anti-inflammatory polyamines (spermidine, putrescine) and neuroprotective 3-indolepropionic acid metabolism and to lower uremic toxins (P-cresol-SO4, indoxyl-SO4) was registered. Thus, B. longum BIOCC1719 may exert health-promoting effects on the host through modulation of the gut microbiome and the host metabolome via inducing the production of health-promoting bioactive compounds. The health effects of the strain need to be confirmed in clinical trials with human volunteers.
Collapse
Affiliation(s)
- Merle Rätsep
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Kalle Kilk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia; (K.K.)
| | - Mihkel Zilmer
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia; (K.K.)
| | - Sirje Kuusik
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Liina Kuus
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Mirjam Vallas
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Oksana Gerulis
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Jelena Štšepetova
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia
| | - Aivar Orav
- Tartu Health Care College, Nooruse St. 5, 50411 Tartu, Estonia
| | - Epp Songisepp
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| |
Collapse
|
27
|
Li J, Ye F, Zhou Y, Lei L, Chen J, Li S, Zhao G. Tailoring the composition, antioxidant activity, and prebiotic potential of apple peel by Aspergillus oryzae fermentation. Food Chem X 2024; 21:101134. [PMID: 38292687 PMCID: PMC10826609 DOI: 10.1016/j.fochx.2024.101134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Apple peel is a typical lignocellulosic food by-product rich in functional components. In this work, apple peel was solid-state fermented with Aspergillus oryzae with an aim to modulate its composition and bioactivity. The results showed that A. oryzae fermentation substantially tailored the composition, improved the antioxidant activity and prebiotic potential of apple peel. Upon the fermentation, 1) free phenolics increased and antioxidant activity improved; 2) the pectin substances degraded significantly, along with a decrease in soluble dietary fiber while an increase in insoluble dietary fiber; 3) the in vitro fermentability increased as indicated by the increase in total acid production. The gut microbiota was shaped with more health-promoting potentials, such as higher abundances of Lactobacillus, Bifidobacterium, Megamonas and Prevotella-9 as well as lower abundances of Enterobacter and Echerichia-Shigella. This work is conducive to the modification of apple peel as a potential ingredient in food formulations.
Collapse
Affiliation(s)
- Jianting Li
- College of Food Science, Southwest University, Chongqing 400715, People’s Republic of China
- Biomass Energy Technology Research Centre, Key Laboratory of Development and Application of Rural Renewable Energy (Ministry of Agriculture and Rural Affairs), Biogas Institute of Ministry of Agriculture and Rural Affairs, Chengdu 610041, People’s Republic of China
| | - Fayin Ye
- College of Food Science, Southwest University, Chongqing 400715, People’s Republic of China
| | - Yun Zhou
- College of Food Science, Southwest University, Chongqing 400715, People’s Republic of China
| | - Lin Lei
- College of Food Science, Southwest University, Chongqing 400715, People’s Republic of China
| | - Jia Chen
- College of Food Science, Southwest University, Chongqing 400715, People’s Republic of China
| | - Sheng Li
- Chongqing Academy of Chinese Materia Medica, Chongqing College of Traditional Chinese Medicine, Chongqing 402760, People’s Republic of China
| | - Guohua Zhao
- College of Food Science, Southwest University, Chongqing 400715, People’s Republic of China
- Chongqing Engineering Research Centre for Regional Foods, Chongqing 400715, People’s Republic of China
| |
Collapse
|
28
|
Bezan PN, Holland H, Vercesi BF, Ovídio PP, Simões LMC, Jordão AA. Fructooligosaccharides Supplementation: A Good Choice for the Prevention and Treatment of Non-Alcoholic Fatty Liver Disease? APPLIED BIOSCIENCES 2024; 3:123-136. [DOI: 10.3390/applbiosci3010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Background and objectives: Carbohydrates such as fructooligosaccharides (FOSs) are associated with improved gastrointestinal health and the prevention of excess body fat. We evaluated the long-term effects of high amounts of FOS on metabolic parameters, non-alcoholic fatty liver disease (NAFLD) and short-chain fatty acids (SCFAs). Methods: Sixty C57BL/6 mice received the following diets for four months: control (C), normolipid rich in fiber (F), normolipid supplemented with FOS (FOS), high fat (HL), high fat with high fiber (HLF) and high fat with FOS (HLFOS). We analyzed the animal weight; body composition; food intake; fasting blood glucose; serum and liver lipid profiles; liver and intestinal histologies; malondialdehyde (MDA), hepatic retinol and α-tocopherol; and SCFAs in the feces. Results: Supplementation with FOS in a high-fat diet promoted less body weight gain and reduced liver and retroperitoneal adipose tissue weights compared to HL and HF. FOS prevented NASH and decreased alanine aminotransferase and serum cholesterol levels in experimental animal models of obesity and metabolic syndrome (MS). There were statistical differences found in the dosages of the three main SCFAs in feces (acetic, isobutyric and isovaleric acids). Conclusions: Long-term supplementation with high doses of FOS was effective in reducing weight, adiposity, NAFLD and serum cholesterol in C57BL mice with obesity and MS induced by a high-fat diet.
Collapse
Affiliation(s)
- Priscila Nogueira Bezan
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Héric Holland
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Bárbara Ferreira Vercesi
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Paula Payão Ovídio
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Livia Maria Cordeiro Simões
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Alceu Afonso Jordão
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| |
Collapse
|
29
|
Ren D, Ding M, Su J, Ye J, He X, Zhang Y, Shang X. Stachyose in combination with L. rhamnosus GG ameliorates acute hypobaric hypoxia-induced intestinal barrier dysfunction through alleviating inflammatory response and oxidative stress. Free Radic Biol Med 2024; 212:505-519. [PMID: 38211833 DOI: 10.1016/j.freeradbiomed.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
High altitude is closely related to intestinal mucosal damage and intestinal microbiota imbalance, and there is currently no effective prevention and treatment measures. In this study, the effects of stachyose (STA), L. rhamnosus GG (LGG) and their combination on inflammatory response, oxidatve stress and intestinal barrier function in mice exposed to acute hypobaric hypoxia were investigated. Our results indicated the combination of STA and LGG could more effectively regulate intestinal microbiota disorders caused by hypobaric hypoxia than STA or LGG alone. When mice were administered with STA + LGG, the content of short chain fatty acids (SCFAs) especially butyric acid significantly increased, which helped intestinal cells to form tight connections, improve the level of anti-inflammatory cytokine (TGF-β) and antioxidant enzymes (SOD, CAT, GSH-Px), and decrease the expression of pro-inlammatory cytokines and hypoxia-inducing factors (IFN-γ, IL-1β, IL-6, TNF-α and HIF-1α), thereby enhance the strong intestinal barrier function. Furthermore, the synbiotics significantly reduced the ratio of Firmicutes to Bacteroidetes, while significantly increased the relative abundance of Rikenella, Bacteroides, Odoribacter, Ruminiclostridium_5 and Gordonibacter, which were correlated with production of SCFAs and anti-inflammatory role. Correlation analysis showed that the protective effect of synbiotics on intestinal barrier function was associated with its anti-inflammatory activity and antioxidant capacity. It provided a strong foundation for further research on the role of STA and LGG in maintaining normal intestinal function at high altitude. Our study has identified and demonstrated a new synbiotic that may be one of the ideal intervention measures for preventing and treating intestinal dysfunction at high altitude.
Collapse
Affiliation(s)
- Dingxin Ren
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Mengying Ding
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Junqing Su
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Jianzhou Ye
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Xiaoqin He
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Yafeng Zhang
- No. 889, Xi'an Institute for Food and Drug, Cangtai West Road, Chang'an District, Xi'an, Shaanxi, 710700, PR China
| | - Xiaoya Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| |
Collapse
|
30
|
Abdelfattah DSE, Fouad MA, Elmeshad AN, El-Nabarawi MA, Elhabal SF. Anti-Obesity Effect of Combining White Kidney Bean Extract, Propolis Ethanolic Extract and CrPi 3 on Sprague-Dawley Rats Fed a High-Fat Diet. Nutrients 2024; 16:310. [PMID: 38276548 PMCID: PMC10818276 DOI: 10.3390/nu16020310] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Obesity has been associated with the occurrence and prevalence of various chronic metabolic diseases. The management of obesity has evolved to focus not only on reducing weight, but also on preventing obesity-related complications. Studies have shown that bioactive components in natural products like white kidney bean extract (WKBE), propolis ethanolic extract (PEE), and chromium picolinate (CrPi3) showed anti-obesity properties. However, no studies have examined the outcomes of combining any of these nutraceutical supplements. We compared the effects of HFD supplemented with WKBE, WKBE+PEE, or WKBE+PEE+CrPi3 against control and obese groups using Sprague-Dawley rats fed a 45% high-fat diet as an in vivo model. Nutritional parameters, biochemical parameters, and biomarkers of cardiovascular disease, liver function, kidney function, and gut health were among the comparable effects. Our findings showed that combining the three nutraceutical supplements had a synergetic effect on reducing weight gain, food utilization rate, abdominal fat, serum lipids, arterial and hepatic lipids, risk of cardiovascular disease, and blood glucose level, in addition to improving renal function and gut microbiota. We attributed these effects to the α-amylase inhibitor action of WKBE, flavonoids, and polyphenol content of PEE, which were potentiated with CrPi3 resulting in a further reduction or normalization of certain parameters.
Collapse
Affiliation(s)
- Doaa Salah Eldin Abdelfattah
- National Nutrition Institute, Cairo 11435, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt
| | | | - Aliaa N Elmeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Technology, The Egyptian Chinese University, Cairo 11786, Egypt
| | - Mohamed A El-Nabarawi
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt
| | - Sammar Fathy Elhabal
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt
| |
Collapse
|
31
|
Li J, Xing H, Lin W, Yu H, Yang B, Jiang C, Zhang J, Wu R, Ding F, Pei M, Yang H. Specific gut microbiome and metabolome changes in patients with continuous ambulatory peritoneal dialysis and comparison between patients with different dialysis vintages. Front Med (Lausanne) 2024; 10:1302352. [PMID: 38249961 PMCID: PMC10797064 DOI: 10.3389/fmed.2023.1302352] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Background In recent years, the role of gut microbiota and derived metabolites in renal disease has attracted more attention. It has been established that the gut microbiota is a potential target for medical interventions in renal disease including chronic kidney disease (CKD), acute kidney injury (AKI) and renal calculus. Emerging evidence has related dialysis treatment to the microbial composition and function of the intestines, and there are many reports related to HD, but few studies have been related to PD. Previous studies have found that PD patients have intestinal flora disturbances, so we speculate that intestinal flora and its metabolites may be the regulatory factors in long-term therapy of PD. And as far as we know, there have been no studies characterized the gut microbiota in PD patients of different dialysis vintages. Methods It is a cross-sectional study based on clinical data and biological samples of 72 patients with CAPD, 13 patients with ESRD and 13 healthy volunteers. The intestinal microecological characteristics of CAPD patients were comprehensively evaluated by combining the intestinal microflora structure, enterotoxin and receptor (serum LPS and LBP), intestinal barrier function index (serum D-Lactate), intestinal uremic toxin (serum IS, PCS, TMAO), fecal SCFAs and other multi-dimensional and multi-omics studies. Furthermore, the changes of intestinal microecology in CAPD patients of different dialysis vintages (≥ 3 and < 12 months, ≥ 12 and < 24 months, ≥ 24 and < 60 months, ≥ 60 months) were further explored, and the correlations between intestinal microecology indicators and some clinical indicators were analyzed. Fecal and serum samples were collected from PD patients (PD group, n = 72), ESRD patients (ESRD group, n = 13) and healthy volunteers (Normal group, n = 13). Fecal samples were subjected to microbiome (16S rDNA) and SCFA (GC-MS) analyses. Serum samples were subjected to LPS, LBP, D-lactate, IS, PCS, and TMAO (ELISA) analyses. Results The diversity and richness of intestinal flora in CAPD patients were lower than those in healthy people and ESRD patients, and the microflora structure was different. Anaerobes of Blautia and facultative anaerobes and aerobic bacteria with Bacilli and Lactobacillales those in Firmicutes are the main intestinal flora in CAPD patients. The abundance of Bacteroidaceae, Bacteroides, Faecalibacterium and other dominant bacteria in the intestinal tract of CAPD patients decreased. Proteobacteria, Enterobacteriaceae and Escherichia-Shigella increased their colonization (LDA > 4). In CAPD patients of different dialysis vintages, there was no significant change in the diversity and richness of microflora, and the microflora structure of PDC group was significantly different from that of PDD, which the abnormal expansion of enterobacter group was more prominent in PDC and the abundance of Bacteroides group was relatively higher in PDD. Intestinal barrier damage, intestinal uremic toxin accumulation and short-chain fatty acid reduction were observed in CAPD patients, such as the serum level of D-Lactate, PCS and TMAO were significantly higher than that in the Normal group (P < 0.05),and the fecal levels of BA and CA were significantly lower (P < 0.05). The intestinal microecological disorder of PDC group, while that of PDD group showed a better trend. Such as the PDC group had a significantly higher serum level of LPS, D-Lactate and TMAO (P < 0.01), and significantly lower serum level of LBP (P < 0.01), and lower fecal levels of AA and BA (P > 0.05) than the PDD group. Conclusion The intestinal microecology and metabolic system of CAPD patients had changes compared with healthy people and ESRD non-dialysis patients, and there were differences in CAPD patients with different dialysis vintages. PD patients on dialysis for more than 60 months showed a better trend in the intestinal microecology than patients with 24∼36 months, which suggested that the intestinal microecology of PD patients had a certain ability of self-regulation and remodeling under the management of standardized system and it is necessary to strengthen the monitoring of the intestinal status and the occurrence of related complications in PD patients on dialysis of 24∼36 months of dialysis vintage. It is initially considered that the mechanism of intestinal microecology is a potential target for intervention in the diagnosis and treatment of CAPD and incorporating intestinal microecosystem monitoring into the long-term management of CAPD patients is a new strategy.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haitao Xing
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Lin
- Department of Nephrology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, China
| | - Hangxing Yu
- Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Bo Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chen Jiang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jin Zhang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruoxi Wu
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fengmei Ding
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ming Pei
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongtao Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
32
|
Mattos Rocha Olivieri C, Aparecida Manólio Soares Freitas R, Alfredo Gomes Arêas J. Jatobá-do-cerrado (Hymenaea stigonocarpa Mart.) pulp positively affects plasma and hepatic lipids and increases short-chain fatty acid production in hamsters fed a hypercholesterolemic diet. Food Res Int 2024; 175:113766. [PMID: 38129058 DOI: 10.1016/j.foodres.2023.113766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/03/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
This study aimed to assess the impact of jatobá pulp, in its fresh (FJ) and extruded (EJ) forms, on lipid metabolism and intestinal fermentation parameters in hamsters. In a 21-day experiment, we determined the parameters of the animal lipid metabolism and colonic production of short chain fatty acids in four different groups. Control (C), fresh pulp (FJ) and extruded pulp (EJ) were fed using hypercholesterolemic diets, and the reference (R) was fed using AIN93 meal. R and C diets contained cellulose, FJ and EJ were added by jatobá pulp as a fiber source. The results showed that FJ and EJ exhibited lower levels of triglycerides, total cholesterol, LDL-c, non-HDL-c serum levels, liver lipids, and liver weight compared to C. The EJ had higher bile acid excretion in stool than the C. EJ and FJ exhibited lower excreted fiber compared to R and C, implying greater fermentation. Furthermore, the production of short-chain fatty acids (SCFA) in the cecum of FJ and EJ animals exceeded that of the C. Acetic and propionic acids were more abundant in the FJ and EJ diets, with FJ producing more butyric acid than the other groups.In conclusion, jatobá pulp maintained at normal levels of total cholesterol, LDL and HDL-associated cholesterol, non-HDL cholesterol, and serum triglycerides, while also reducing the accumulation of hepatic lipids. Jatobá also promoted SCFA formation and fermentation, making it a valuable ingredient for preventing chronic diseases.
Collapse
Affiliation(s)
- Camila Mattos Rocha Olivieri
- Department of Nutrition, School of Public Health, University of São Paulo, Av. Dr. Arnaldo, 715, São Paulo, SP 01246-904, Brazil.
| | | | - José Alfredo Gomes Arêas
- Department of Nutrition, School of Public Health, University of São Paulo, Av. Dr. Arnaldo, 715, São Paulo, SP 01246-904, Brazil.
| |
Collapse
|
33
|
Marsiglia R, Marangelo C, Vernocchi P, Scanu M, Pane S, Russo A, Guanziroli E, Del Chierico F, Valeriani M, Molteni F, Putignani L. Gut Microbiota Ecological and Functional Modulation in Post-Stroke Recovery Patients: An Italian Study. Microorganisms 2023; 12:37. [PMID: 38257864 PMCID: PMC10819831 DOI: 10.3390/microorganisms12010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Ischemic stroke (IS) can be caused by perturbations of the gut-brain axis. An imbalance in the gut microbiota (GM), or dysbiosis, may be linked to several IS risk factors and can influence the brain through the production of different metabolites, such as short-chain fatty acids (SCFAs), indole and derivatives. This study examines ecological changes in the GM and its metabolic activities after stroke. Fecal samples of 10 IS patients were compared to 21 healthy controls (CTRLs). GM ecological profiles were generated via 16S rRNA taxonomy as functional profiles using metabolomics analysis performed with a gas chromatograph coupled to a mass spectrometer (GC-MS). Additionally fecal zonulin, a marker of gut permeability, was measured using an enzyme-linked immuno assay (ELISA). Data were analyzed using univariate and multivariate statistical analyses and correlated with clinical features and biochemical variables using correlation and nonparametric tests. Metabolomic analyses, carried out on a subject subgroup, revealed a high concentration of fecal metabolites, such as SCFAs, in the GM of IS patients, which was corroborated by the enrichment of SCFA-producing bacterial genera such as Bacteroides, Christensellaceae, Alistipes and Akkermansia. Conversely, indole and 3-methyl indole (skatole) decreased compared to a subset of six CTRLs. This study illustrates how IS might affect the gut microbial milieu and may suggest potential microbial and metabolic biomarkers of IS. Expanded populations of Akkermansia and enrichment of acetic acid could be considered potential disease phenotype signatures.
Collapse
Affiliation(s)
- Riccardo Marsiglia
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (C.M.); (P.V.); (M.S.); (F.D.C.)
| | - Chiara Marangelo
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (C.M.); (P.V.); (M.S.); (F.D.C.)
| | - Pamela Vernocchi
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (C.M.); (P.V.); (M.S.); (F.D.C.)
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (C.M.); (P.V.); (M.S.); (F.D.C.)
| | - Stefania Pane
- Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (A.R.)
| | - Alessandra Russo
- Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (A.R.)
| | - Eleonora Guanziroli
- Villa Beretta Rehabilitation Center, Valduce Hospital Como, 23845 Costa Masnaga, Italy; (E.G.); (F.M.)
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (C.M.); (P.V.); (M.S.); (F.D.C.)
| | - Massimiliano Valeriani
- Developmental Neurology, Bambino Gesù Children Hospital, IRCCS, 00165 Rome, Italy;
- Center for Sensory Motor Interaction, Aalborg University, 9220 Aalborg, Denmark
| | - Franco Molteni
- Villa Beretta Rehabilitation Center, Valduce Hospital Como, 23845 Costa Masnaga, Italy; (E.G.); (F.M.)
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
34
|
Tilves C, Zhao HJ, Differding MK, Zhang M, Liu T, Hoyo C, Østbye T, Benjamin-Neelon SE, Mueller NT. Associations of Plastic Bottle Exposure with Infant Growth, Fecal Microbiota, and Short-Chain Fatty Acids. Microorganisms 2023; 11:2924. [PMID: 38138068 PMCID: PMC10745781 DOI: 10.3390/microorganisms11122924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND/OBJECTIVES Murine models show that plastics, via their chemical constituents (e.g., phthalates), influence microbiota, metabolism, and growth. However, research on plastics in humans is lacking. Here, we examine how the frequency of plastic bottle exposure is associated with fecal microbiota, short-chain fatty acids (SCFAs), and anthropometry in the first year of life. SUBJECTS/METHODS In 442 infants from the prospective Nurture birth cohort, we examined the association of frequency of plastic bottle feeding at 3 months with anthropometric outcomes (skinfolds, length-for-age, and weight-for-length) at 12 months of age and growth trajectories between 3 and 12 months. Furthermore, in a subset of infants (n = 70) that contributed fecal samples at 3 months and 12 months of age, we examined plastic bottle frequency in relation to fecal microbiota composition and diversity (measured by 16S rRNA gene sequencing of V4 region), and fecal SCFA concentrations (quantified using gas chromatography mass spectrometry). RESULTS At 3 months, 67.6% of infants were plastic bottle fed at every feeding, 15.4% were exclusively breast milk fed, and 48.9% were exclusively formula fed. After adjustment for potential confounders, infants who were plastic bottle fed less than every feeding compared to those who were plastic bottle fed at every feeding at 3 months did not show differences in anthropometry over the first 12 months of life, save for lower length-for-age z-score at 12 months (adjusted β = -0.45, 95% CI: -0.76, -0.13). Infants who were plastic bottle fed less than every feeding versus every feeding had lower fecal microbiota alpha diversity at 3 months (mean difference for Shannon index: -0.59, 95% CI: -0.99, -0.20) and lower isovaleric acid concentration at 3 months (mean difference: -2.12 μmol/g, 95% CI: -3.64, -0.60), but these results were attenuated following adjustment for infant diet. Plastic bottle frequency was not strongly associated with microbiota diversity or SCFAs at 12 months after multivariable adjustment. Frequency of plastic bottle use was associated with differential abundance of some bacterial taxa, however, significance was not consistent between statistical approaches. CONCLUSIONS Plastic bottle frequency at 3 months was not strongly associated with measures of adiposity or growth (save for length-for-age) over the first year of life, and while plastic bottle use was associated with some features of fecal microbiota and SCFAs in the first year, these findings were attenuated in multivariable models with infant diet. Future research is needed to assess health effects of exposure to other plastic-based products and objective measures of microplastics and plastic constituents like phthalates.
Collapse
Affiliation(s)
- Curtis Tilves
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.T.); (H.J.Z.); (M.K.D.); (M.Z.); (T.L.)
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Heather Jianbo Zhao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.T.); (H.J.Z.); (M.K.D.); (M.Z.); (T.L.)
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Moira K. Differding
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.T.); (H.J.Z.); (M.K.D.); (M.Z.); (T.L.)
| | - Mingyu Zhang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.T.); (H.J.Z.); (M.K.D.); (M.Z.); (T.L.)
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Tiange Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.T.); (H.J.Z.); (M.K.D.); (M.Z.); (T.L.)
| | - Cathrine Hoyo
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA;
| | - Truls Østbye
- Department of Family Medicine and Community Health, Duke University, Durham, NC 27708, USA;
| | - Sara E. Benjamin-Neelon
- Department of Health, Behavior and Society, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Noel T. Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.T.); (H.J.Z.); (M.K.D.); (M.Z.); (T.L.)
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
35
|
Ismail HM, Perera D, Mandal R, DiMeglio LA, Evans-Molina C, Hannon T, Petrosino J, Javornick CreGreen S, Schmidt NW. Gut microbial changes associated with obesity in youth with type 1 diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.01.23299251. [PMID: 38076970 PMCID: PMC10705628 DOI: 10.1101/2023.12.01.23299251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Obesity is increasingly prevalent in type 1 diabetes (T1D) and is associated with management problems and higher risk for diabetes complications. Gut microbiome changes have been described separately in each of T1D and obesity, however, it is unknown to what extent gut microbiome changes are seen when obesity and T1D concomitantly occur. OBJECTIVE To describe the gut microbiome and microbial metabolite changes associated with obesity in T1D. We hypothesized significant gut microbial and metabolite differences between T1D youth who are lean (BMI: 5-<85%) vs. those with obesity (BMI: ≥95%). METHODS We analyzed stool samples for gut microbial (using metagenomic shotgun sequencing) and short-chain fatty acid (SCFA) metabolite differences in lean (n=27) and obese (n=21) T1D youth. The mean±SD age was 15.3±2.2yrs, A1c 7.8±1.3%, diabetes duration 5.1±4.4yrs, 42.0% females, and 94.0% were White. Linear discriminant analysis (LDA) effect size (LEfSe) was used to identify taxa that best discriminated between the BMI groups. RESULTS Bacterial community composition showed differences in species type (β-diversity) by BMI group (p=0.013). At the genus level, there was a higher ratio of Prevotella to Bacteroides in the obese group (p=0.0058). LEfSe analysis showed a differential distribution of significantly abundant taxa in either the lean or obese groups, including increased relative abundance of Prevotella copri , among other taxa in the obese group. Functional profiling showed that pathways associated with decreased insulin sensitivity were upregulated in the obese group. Stool SCFAs (acetate, propionate and butyrate) were higher in the obese compared to the lean group (p<0.05 for all). CONCLUSIONS Our findings identify gut microbiome, microbial metabolite and functional pathways differences associated with obesity in T1D. These findings could be helpful in identifying gut microbiome targeted therapies to manage obesity in T1D.
Collapse
|
36
|
Li B, Hsieh YR, Lai WD, Tung TH, Chen YX, Yang CH, Fang YC, Huang SY. Melatonin Ameliorates Neuropsychiatric Behaviors, Gut Microbiome, and Microbiota-Derived Metabolites in Rats with Chronic Sleep Deprivation. Int J Mol Sci 2023; 24:16820. [PMID: 38069141 PMCID: PMC10706682 DOI: 10.3390/ijms242316820] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
With the increasing prevalence of sleep deprivation (SD)-related disorders, the effective treatment of sleep disorders has become a critical health research topic. Thus, we hypothesized and investigated the effectiveness of a 3-week melatonin intervention on neuropsychiatric behavioral responses mediated throughout melatonin receptors, gut microbiota, and lipid metabolites in rats with chronic SD. Eighteen 6-week-old Wistar rats were used and divided into the control grup (C, n = 6), SD group (n = 6), and melatonin-supplemented group (SDM, n = 6). During weeks 0 to 6, animals were provided with the AIN-93M diet and free access to water. Four-week chronic SD was conducted from weeks 7 to 10. Exogenous melatonin administration (10 mg/kg BW) was injected intraperitoneally 1 h before the daily administration of SD for 3 weeks in the SDM group. SD rats exhibited anxiety-like behavior, depression-like behavior, and cognitive impairment. Exogenous melatonin administration ameliorated neuropsychiatric behaviors induced by chronic SD. Analysis of fecal metabolites indicated that melatonin may influence brain messaging through the microbiota-gut-brain axis by increasing the production of short-chain fatty acids (SCFA) and decreasing the production of secondary bile acids (SBA). Four-week SD reduced the cerebral cortex expression of MT1, but not in the colon. Chronic SD led to anxiety and depression-like behaviors and cognitive decline, as well as the reduced intestinal level of SCFAs and the enhanced intestinal level of SBAs in rats. In this work, we confirmed our hypothesis that a 3-week melatonin intervention on neuropsychiatric behavioral response mediated throughout melatonin receptors, gut microbiota, and lipid metabolites in rats with chronic SD.
Collapse
Affiliation(s)
- Bingcong Li
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Yin-Ru Hsieh
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Wen-De Lai
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Te-Hsuan Tung
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Yu-Xuan Chen
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Chia-Hui Yang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Yu-Chiao Fang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan; (B.L.); (Y.-R.H.)
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
37
|
Smith M, Polite L, Christy A, Edirisinghe I, Burton-Freeman B, Sandhu A. An Improved Validated Method for the Determination of Short-Chain Fatty Acids in Human Fecal Samples by Gas Chromatography with Flame Ionization Detection (GC-FID). Metabolites 2023; 13:1106. [PMID: 37999203 PMCID: PMC10673161 DOI: 10.3390/metabo13111106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/04/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are metabolites produced by the gut microbiota through the fermentation of non-digestible carbohydrates. Recent studies suggest that the gut microbiota composition, diet and metabolic status play an important role in the production of SCFAs. The primary objective of this study was to develop a simplified method for SCFA analysis in human fecal samples by gas chromatography with flame ionization detection (GC-FID). The secondary objective was to apply the method to fecal samples collected from a clinical trial. The developed GC-FID method showed excellent linearity (R2 > 0.99994), with a limit of detection (LOD) ranging from 0.02 to 0.23 µg/mL and a limit of quantification (LOQ) ranging from 0.08 to 0.78 µg/mL. Recovery for the method ranged between 54.24 ± 1.17% and 140.94 ± 2.10%. Intra- and inter-day repeatability ranged from 0.56 to 1.03 and from 0.10 to 4.76% RSD, respectively. Nine SCFAs were identified and quantified (acetic, propionic, iso-butyric, butyric, iso-valeric, valeric, 4-methyl valeric, hexanoic and heptanoic acids) in freeze-dried fecal samples. The clinical trial compared participants with prediabetes mellitus and insulin resistance (IR-group, n = 20) to metabolically healthy participants (reference group, R-group, n = 9) following a 4-week intervention of a daily red raspberry smoothie (RRB, 1 cup fresh-weight equivalent) with or without fructo-oligosaccharide (RRB + FOS, 1 cup RRB + 8 g FOS). The statistical analysis (Student's t-test, ANCOVA) was performed on PC-SAS 9.4 (SAS Institute). Acetic acid was higher in the R-group compared to the IR-group at baseline/week 0 (p = 0.14). No significant changes in fecal SCFA content were observed after 4 weeks of either RRB or RRB + FOS.
Collapse
Affiliation(s)
- Morganne Smith
- Department of Food Science and Nutrition and Center for Nutrition Research, Illinois Institute of Technology, Chicago, IL 60616, USA; (M.S.); (I.E.); (B.B.-F.)
| | - Lee Polite
- Axion Analytical Labs Inc., Chicago, IL 60607, USA; (L.P.); (A.C.)
| | - Andreas Christy
- Axion Analytical Labs Inc., Chicago, IL 60607, USA; (L.P.); (A.C.)
| | - Indika Edirisinghe
- Department of Food Science and Nutrition and Center for Nutrition Research, Illinois Institute of Technology, Chicago, IL 60616, USA; (M.S.); (I.E.); (B.B.-F.)
| | - Britt Burton-Freeman
- Department of Food Science and Nutrition and Center for Nutrition Research, Illinois Institute of Technology, Chicago, IL 60616, USA; (M.S.); (I.E.); (B.B.-F.)
| | - Amandeep Sandhu
- Department of Food Science and Nutrition and Center for Nutrition Research, Illinois Institute of Technology, Chicago, IL 60616, USA; (M.S.); (I.E.); (B.B.-F.)
| |
Collapse
|
38
|
Zhang Y, Shen Y, Liufu N, Liu L, Li W, Shi Z, Zheng H, Mei X, Chen CY, Jiang Z, Abtahi S, Dong Y, Liang F, Shi Y, Cheng LL, Yang G, Kang JX, Wilkinson JE, Xie Z. Transmission of Alzheimer's disease-associated microbiota dysbiosis and its impact on cognitive function: evidence from mice and patients. Mol Psychiatry 2023; 28:4421-4437. [PMID: 37604976 PMCID: PMC11733706 DOI: 10.1038/s41380-023-02216-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/26/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023]
Abstract
Spouses of Alzheimer's disease (AD) patients are at a higher risk of developing incidental dementia. However, the causes and underlying mechanism of this clinical observation remain largely unknown. One possible explanation is linked to microbiota dysbiosis, a condition that has been associated with AD. However, it remains unclear whether gut microbiota dysbiosis can be transmitted from AD individuals to non-AD individuals and contribute to the development of AD pathogenesis and cognitive impairment. We, therefore, set out to perform both animal studies and clinical investigation by co-housing wild-type mice and AD transgenic mice, analyzing microbiota via 16S rRNA gene sequencing, measuring short-chain fatty acid amounts, and employing behavioral test, mass spectrometry, site-mutations and other methods. The present study revealed that co-housing between wild-type mice and AD transgenic mice or administrating feces of AD transgenic mice to wild-type mice resulted in AD-associated gut microbiota dysbiosis, Tau phosphorylation, and cognitive impairment in the wild-type mice. Gavage with Lactobacillus and Bifidobacterium restored these changes in the wild-type mice. The oral and gut microbiota of AD patient partners resembled that of AD patients but differed from healthy controls, indicating the transmission of microbiota. The underlying mechanism of these findings includes that the butyric acid-mediated acetylation of GSK3β at lysine 15 regulated its phosphorylation at serine 9, consequently impacting Tau phosphorylation. Pending confirmative studies, these results provide insight into a potential link between the transmission of AD-associated microbiota dysbiosis and development of cognitive impairment, which underscore the need for further research in this area.
Collapse
Affiliation(s)
- Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Yuan Shen
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
- Mental Health Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Ning Liufu
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, 510120, PR China
| | - Ling Liu
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, 510120, PR China
| | - Wei Li
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Zhongyong Shi
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
- Mental Health Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Hailin Zheng
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Xinchun Mei
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
- Mental Health Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Zengliang Jiang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, PR China
| | - Shabnamsadat Abtahi
- Biostatistics Department and Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Feng Liang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yujiang Shi
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Leo L Cheng
- Departments of Radiology and Pathology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jeremy E Wilkinson
- Biostatistics Department and Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
39
|
Giridharan VV, Catumbela CSG, Catalão CHR, Lee J, Ganesh BP, Petronilho F, Dal-Pizzol F, Morales R, Barichello T. Sepsis exacerbates Alzheimer's disease pathophysiology, modulates the gut microbiome, increases neuroinflammation and amyloid burden. Mol Psychiatry 2023; 28:4463-4473. [PMID: 37452088 PMCID: PMC10926876 DOI: 10.1038/s41380-023-02172-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 05/25/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
While our understanding of the molecular biology of Alzheimer's disease (AD) has grown, the etiology of the disease, especially the involvement of peripheral infection, remains a challenge. In this study, we hypothesize that peripheral infection represents a risk factor for AD pathology. To test our hypothesis, APP/PS1 mice underwent cecal ligation and puncture (CLP) surgery to develop a polymicrobial infection or non-CLP surgery. Mice were euthanized at 3, 30, and 120 days after surgery to evaluate the inflammatory mediators, glial cell markers, amyloid burden, gut microbiome, gut morphology, and short-chain fatty acids (SCFAs) levels. The novel object recognition (NOR) task was performed 30 and 120 days after the surgery, and sepsis accelerated the cognitive decline in APP/PS1 mice at both time points. At 120 days, the insoluble Aβ increased in the sepsis group, and sepsis modulated the cytokines/chemokines, decreasing the cytokines associated with brain homeostasis IL-10 and IL-13 and increasing the eotaxin known to influence cognitive function. At 120 days, we found an increased density of IBA-1-positive microglia in the vicinity of Aβ dense-core plaques, compared with the control group confirming the predictable clustering of reactive glia around dense-core plaques within 15 μm near Aβ deposits in the brain. In the gut, sepsis negatively modulated the α- and β-diversity indices evaluated by 16S rRNA sequencing, decreased the levels of SCFAs, and significantly affected ileum and colon morphology in CLP mice. Our data suggest that sepsis-induced peripheral infection accelerates cognitive decline and AD pathology in the AD mouse model.
Collapse
Affiliation(s)
- Vijayasree V Giridharan
- Faillace Department of Psychiatry and Behavioural Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Celso S G Catumbela
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Carlos Henrique R Catalão
- Faillace Department of Psychiatry and Behavioural Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabricia Petronilho
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Rodrigo Morales
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Tatiana Barichello
- Faillace Department of Psychiatry and Behavioural Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
40
|
Meiller L, Sauvinet V, Breyton AE, Ranaivo H, Machon C, Mialon A, Meynier A, Bischoff SC, Walter J, Neyrinck AM, Laville M, Delzenne NM, Vinoy S, Nazare JA. Metabolic signature of 13C-labeled wheat bran consumption related to gut fermentation in humans: a pilot study. Eur J Nutr 2023; 62:2633-2648. [PMID: 37222787 DOI: 10.1007/s00394-023-03161-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/18/2023] [Indexed: 05/25/2023]
Abstract
PURPOSE The aim of this pilot study was to analyze concomitantly the kinetics of production of 13C-labeled gut-derived metabolites from 13C-labeled wheat bran in three biological matrices (breath, plasma, stools), in order to assess differential fermentation profiles among subjects. METHODS Six healthy women consumed a controlled breakfast containing 13C-labeled wheat bran biscuits. H2, CH4 and 13CO2, 13CH4 24 h-concentrations in breath were measured, respectively, by gas chromatography (GC) and GC-isotope ratio mass spectrometry (GC-IRMS). Plasma and fecal concentrations of 13C-short-chain fatty acids (linear SCFAs: acetate, propionate, butyrate, valerate; branched SCFAs: isobutyrate, isovalerate) were quantified using GC-combustion-IRMS. Gut microbiota composition was assessed by16S rRNA gene sequencing analysis. RESULTS H2 and CH4 24 h-kinetics distinguished two groups in terms of fermentation-related gas excretion: high-CH4 producers vs low-CH4 producers (fasting concentrations: 45.3 ± 13.6 ppm vs 6.5 ± 3.6 ppm). Expired 13CH4 was enhanced and prolonged in high-CH4 producers compared to low-CH4 producers. The proportion of plasma and stool 13C-butyrate tended to be higher in low-CH4 producers, and inversely for 13C-acetate. Plasma branched SCFAs revealed different kinetics of apparition compared to linear SCFAs. CONCLUSION This pilot study allowed to consider novel procedures for the development of biomarkers revealing dietary fiber-gut microbiota interactions. The non-invasive assessment of exhaled gas following 13C-labeled fibers ingestion enabled to decipher distinct fermentation profiles: high-CH4 producers vs low-CH4 producers. The isotope labeling permits a specific in vivo characterisation of the dietary fiber impact consumption on microbiota metabolite production. CLINICAL TRIAL REGISTRATION The study has been registered under the number NCT03717311 at ClinicalTrials.gov on October 24, 2018.
Collapse
Affiliation(s)
- Laure Meiller
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Valérie Sauvinet
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Anne-Esther Breyton
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France
| | - Harimalala Ranaivo
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France
| | - Christelle Machon
- Service de Biochimie et Biologie Moléculaire, Hospices Civils de Lyon, Pierre Bénite, France
| | - Anne Mialon
- Service de Biochimie et Biologie Moléculaire, Hospices Civils de Lyon, Pierre Bénite, France
| | | | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, UCLouvain, Brussels, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France
- Service d'Endocrinologie Diabète Nutrition, Hospices Civils de Lyon, Pierre Bénite, France
| | | | - Sophie Vinoy
- Nutrition Research, Mondelez International, Saclay, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France.
| |
Collapse
|
41
|
Yi X, Cai R, Shaoyong W, Wang G, Yan W, He Z, Li R, Chao M, Zhao T, Deng L, Yang G, Pang W. Melatonin promotes gut anti-oxidative status in perinatal rat by remodeling the gut microbiome. Redox Biol 2023; 65:102829. [PMID: 37527604 PMCID: PMC10407234 DOI: 10.1016/j.redox.2023.102829] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
Gut health is important for nutrition absorption, reproduction, and lactation in perinatal and early weaned mammals. Although melatonin functions in maintaining circadian rhythms and preventing obesity, neurodegenerative diseases, and viral infections, its impact on the gut microbiome and its function in mediating gut health through gut microbiota remain largely unexplored. In the present study, the microbiome of rats was monitoring after fecal microbiota transplantation (FMT) and foster care (FC). The results showed that FMT and FC increased intestinal villus height/crypt depth in perinatal rats. Mechanistically, the melatonin-mediated remodeling of gut microbiota inhibited oxidative stress, which led to attenuation of autophagy and inflammation. In addition, FMT and FC encouraged the growth of more beneficial intestinal bacteria, such as Allobaculum, Bifidobacterium, and Faecalibaculum, which produce more short-chain fatty acids to strengthen intestinal anti-oxidation. These findings suggest that melatonin-treated gut microbiota increase the production of SCFAs, which improve gut health by reducing oxidative stress, autophagy and inflammation. The transfer of melatonin-treated gut microbiota may be a new and effective method by which to ameliorate gut health in perinatal and weaned mammals.
Collapse
Affiliation(s)
- Xudong Yi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Rui Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Weike Shaoyong
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Guoyan Wang
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Wenyong Yan
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhaozhao He
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Ri Li
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Mingkun Chao
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Tiantian Zhao
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lu Deng
- Innovative Research Team of Animal Nutrition & Healthy Feeding, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Weijun Pang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
42
|
Jiang K, Wang D, Su L, Liu X, Yue Q, Zhang S, Zhao L. Tamarind Seed Polysaccharide Hydrolysate Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis via Regulating the Gut Microbiota. Pharmaceuticals (Basel) 2023; 16:1133. [PMID: 37631047 PMCID: PMC10459238 DOI: 10.3390/ph16081133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Ulcerative colitis (UC) is a disease caused by noninfectious chronic inflammation characterized by varying degrees of inflammation affecting the colon or its entire mucosal surface. Current therapeutic strategies rely on the suppression of the immune response, which is effective, but can have detrimental effects. Recently, different plant polysaccharides and their degradation products have received increasing attention due to their prominent biological activities. The aim of this research was to evaluate the mitigation of inflammation exhibited by tamarind seed polysaccharide hydrolysate (TSPH) ingestion in colitis mice. (2) Methods: TSPH was obtained from the hydrolysis of tamarind seed polysaccharide (TSP) by trifluoroacetic acid (TFA). The structure and physical properties of TSPH were characterized by ultraviolet spectroscopy (UV), thin-layer chromatography (TLC), fourier transform infrared spectroscopy (FT-IR), and High-Performance Liquid Chromatography and Electrospray Ionization Mass Spectrometry (HPLC-ESI/MS) analysis. Then, the alleviative effects of the action of TSPH on 2.5% dextran sodium sulfate (DSS)-induced colitis mice were investigated. (3) Results: TSPH restored pathological lesions in the colon and inhibited the over-secretion of pro-inflammatory cytokines in UC mice. The relative expression level of mRNA for colonic tight junction proteins was increased. These findings suggested that TSPH could reduce inflammation in the colon. Additionally, the structure of the gut microbiota was also altered, with beneficial bacteria, including Prevotella and Blautia, significantly enriched by TSPH. Moreover, the richness of Blautia was positively correlated with acetic acid. (4) Conclusions: In conclusion, TSPH suppressed colonic inflammation, alleviated imbalances in the intestinal flora and regulated bacterial metabolites. Thus, this also implies that TSPH has the potential to be a functional food against colitis.
Collapse
Affiliation(s)
- Kangjia Jiang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Duo Wang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Xinli Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Qiulin Yue
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Song Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
- Shandong Chenzhang Biotechnology Co., Ltd., Jinan 250353, China
| |
Collapse
|
43
|
Khan A, Li S, Han H, Jin WL, Ling Z, Ji J, Iram S, Liu P, Xiao S, Salama ES, Li X. A gluten degrading probiotic Bacillus subtilis LZU-GM relieve adverse effect of gluten additive food and balances gut microbiota in mice. Food Res Int 2023; 170:112960. [PMID: 37316006 DOI: 10.1016/j.foodres.2023.112960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/24/2023] [Accepted: 05/10/2023] [Indexed: 06/16/2023]
Abstract
Gluten accumulation damages the proximal small intestine and causes celiac disease (CeD) which has not been effectively treated except by using a gluten-free diet. In this study, strain Bacillus subtilis LZU-GM was isolated from Pakistani traditional fermented sourdough and could degrade 73.7% of gluten in 24 h in vitro. Strain LZU-GM was employed for practical application to investigate gluten degradation in mice models. The results showed that strain LZU-GM was colonized in mice and the survival rate was around 0.95 % (P < 0.0001). The gluten degradation was 3-fold higher in the small intestine of the strain LZU-GM treated mice group remaining 1511.96 ng/mL of gluten peptides than the untreated mice group (6500.38 ng/mL). Immunochemical analysis showed that gluten-treated mice established positive antigliadin antibodies (AGA) in serum (IgA, IgG, and anti-TG2 antibodies) as compared to the strain LZU-GM treatment group. Furthermore, the number of IFN-γ, TNF-α, IL-10, and COX-2 cells decrease in the lamina propria of the strain LZU-GM treatment group (P < 0.0001). Microbial community bar plot analysis showed that Lactobacillus, Dubosiella, and Enterococcus genera were restored and stabilized in the LZU-GM treatment group while Blautia and Ruminococcus were found lower. The oral gavage of probiotic strain LZU-GM might be useful for gluten metabolism in the intestine during digestion and would be a long-term dietary treatment for CeD management.
Collapse
Affiliation(s)
- Aman Khan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, Gansu Province 730000, PR China; State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Shiqing Li
- State Key Laboratory of Soil Erosion and Dryland Farming on the Loess Plateau, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Huawen Han
- State Key Laboratory of Grassland Agro-ecosystems, Center for Grassland Microbiome, and College of Pastoral Agricultural Science and Technology, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Zhenmin Ling
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Jing Ji
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, Gansu Province 730000, PR China; State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Shazia Iram
- Department of Environmental Sciences, Fatima Jinnah Women University, Rawalpindi 46000, Pakistan
| | - Pu Liu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Sa Xiao
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - El-Sayed Salama
- Department of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, PR China
| | - Xiangkai Li
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, Gansu Province 730000, PR China.
| |
Collapse
|
44
|
Wang S, Cai Q, Xu L, Sun Y, Wang M, Wang Y, Zhang L, Li K, Ni Z. Isoalantolactone relieves depression-like behaviors in mice after chronic social defeat stress via the gut-brain axis. Psychopharmacology (Berl) 2023; 240:1775-1787. [PMID: 37400661 PMCID: PMC10349788 DOI: 10.1007/s00213-023-06413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023]
Abstract
RATIONALE The management of depression continues to be challenging despite the variety of available antidepressants. Herbal medicines are used in many cultures but lack stringent testing to understand their efficacy and mechanism of action. Isoalantolactone (LAT) from Elecampane (Inula helenium) improved the chronic social defeat stress (CSDS)-induced anhedonia-like phenotype in mice comparable to fluoxetine, a selective serotonin reuptake inhibitor (SSRI). OBJECTIVES Compare the effects of LAT and fluoxetine on depression-like behaviors in mice exposed to CSDS. RESULT The CSDS-induced decrease in protein expression of postsynaptic density (PSD95), brain derived neurotrophic factor (BDNF), and glutamate receptor subunit-1 (GluA1) in the prefrontal cortex was restored by LAT. LAT showed robust anti-inflammatory activity and can lessen the increase in IL-6 and TNF-α caused by CSDS. CSDS altered the gut microbiota at the taxonomic level, resulting in significant changes in α- and β-diversity. LAT treatment reestablished the bacterial abundance and diversity and increased the production of butyric acid in the gut that was inhibited by CSDS. The levels of butyric acid were negatively correlated with the abundance of Bacteroidetes, and positively correlated with those of Proteobacteria and Firmicutes across all treatment groups. CONCLUSIONS The current data suggest that, similar to fluoxetine, LAT show antidepressant-like effects in mice exposed to CSDS through the modulation of the gut-brain axis.
Collapse
Affiliation(s)
- Siming Wang
- School of Basic Medical Science, Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Qihan Cai
- School of Basic Medical Science, Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Lu Xu
- School of Basic Medical Science, Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Yanan Sun
- College of Traditional Chinese Medicine, Hebei University, Baoding, 071000, China
| | - Mengmeng Wang
- School of Basic Medical Science, Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Yu Wang
- School of Basic Medical Science, Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Lili Zhang
- Hebei Provincial Mental Health Center, Baoding, 071000, Hebei Province, China
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Baoding, 071000, China
| | - Keqing Li
- Hebei Provincial Mental Health Center, Baoding, 071000, Hebei Province, China.
- Hebei Key Laboratory of Major Mental and Behavioral Disorders, Baoding, 071000, China.
- , Baoding, China.
| | - Zhiyu Ni
- Affiliated Hospital of Hebei University, Baoding, 071000, China.
- Clinical Medical College, Hebei University, Baoding, 071000, Hebei Province, People's Republic of China.
- Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, 071000, China.
| |
Collapse
|
45
|
Littlejohn PT, Bar-Yoseph H, Edwards K, Li H, Ramirez-Contreras CY, Holani R, Metcalfe-Roach A, Fan YM, Yang TMS, Radisavljevic N, Hu X, Johnson JD, Finlay BB. Multiple micronutrient deficiencies alter energy metabolism in host and gut microbiome in an early-life murine model. Front Nutr 2023; 10:1151670. [PMID: 37497061 PMCID: PMC10365968 DOI: 10.3389/fnut.2023.1151670] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
Introduction Micronutrients perform a wide range of physiological functions essential for growth and development. However, most people still need to meet the estimated average requirement worldwide. Globally, 2 billion people suffer from micronutrient deficiency, most of which are co-occurring deficiencies in children under age five. Despite decades of research, animal models studying multiple micronutrient deficiencies within the early-life period are lacking, which hinders our complete understanding of the long-term health implications and may contribute to the inefficacy of some nutritional interventions. Evidence supporting the Developmental Origins of Health and Disease (DOHaD) theory demonstrates that early-life nutritional deficiencies carry life-long consequences mediated through various mechanisms such as abnormal metabolic programming, stunting, altered body composition, and the gut microbiome. However, this is largely unexplored in the multiple micronutrient deficient host. Methods we developed a preclinical model to examine undernutrition's metabolic and functional impact on the host and gut microbiome early in life. Three-week-old weanling C57BL/6N male mice were fed a low-micronutrient diet deficient in zinc, folate, iron, vitamin A, and vitamin B12 or a control diet for 4-weeks. Results Our results showed that early-life multiple micronutrient deficiencies induced stunting, altered body composition, impaired glucose and insulin tolerance, and altered the levels of other micronutrients not depleted in the diet within the host. In addition, functional metagenomics profiling and a carbohydrate fermentation assay showed an increased microbial preference for simple sugars rather than complex ones, suggestive of a less developed microbiome in the low-micronutrient-fed mice. Moreover, we found that a zinc-only deficient diet was not sufficient to induce these phenotypes, further supporting the importance of studying co-occurring deficiencies. Discussion Together, these findings highlight a previously unappreciated role of early-life multiple micronutrient deficiencies in shaping the metabolic phenome of the host and gut microbiome through altered glucose energy metabolism, which may have implications for metabolic disease later in life in micronutrient-deficient survivors.
Collapse
Affiliation(s)
- Paula T. Littlejohn
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Haggai Bar-Yoseph
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Karlie Edwards
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Hong Li
- Life Sciences Institute and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Ravi Holani
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Avril Metcalfe-Roach
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Yiyun M. Fan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Tom Min-Shih Yang
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Nina Radisavljevic
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Xiaoke Hu
- Life Sciences Institute and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - James D. Johnson
- Life Sciences Institute and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Weninger SN, Ding A, Browne EN, Frost ML, Schiro G, Laubitz D, Duca FA. Longitudinal Characterization of the Gut Microbiota in the Diabetic ZDSD Rat Model and Therapeutic Potential of Oligofructose. Metabolites 2023; 13:660. [PMID: 37233701 PMCID: PMC10220957 DOI: 10.3390/metabo13050660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
The complex development of type 2 diabetes (T2D) creates challenges for studying the progression and treatment of the disease in animal models. A newly developed rat model of diabetes, the Zucker Diabetic Sprague Dawley (ZDSD) rat, closely parallels the progression of T2D in humans. Here, we examine the progression of T2D and associated changes in the gut microbiota in male ZDSD rats and test whether the model can be used to examine the efficacy of potential therapeutics such as prebiotics, specifically oligofructose, that target the gut microbiota. Bodyweight, adiposity, and fed/fasting blood glucose and insulin were recorded over the course of the study. Glucose and insulin tolerance tests were performed, and feces collected at 8, 16, and 24 weeks of age for short-chain fatty acids and microbiota analysis using 16s rRNA gene sequencing. At the end of 24 weeks of age, half of the rats were supplemented with 10% oligofructose and tests were repeated. We observed a transition from healthy/nondiabetic to prediabetic and overtly diabetic states, via worsened insulin and glucose tolerance and significant increases in fed/fasted glucose, followed by a significant decrease in circulating insulin. Acetate and propionate levels were significantly increased in the overt diabetic state compared to healthy and prediabetic. Microbiota analysis demonstrated alterations in the gut microbiota with shifts in alpha and beta diversity as well as alterations in specific bacterial genera in healthy compared to prediabetic and diabetic states. Oligofructose treatment improved glucose tolerance and shifted the cecal microbiota of the ZDSD rats during late-stage diabetes. These findings underscore the translational potential of ZDSD rats as a model of T2D and highlight potential gut bacteria that could impact the development of the disease or serve as a biomarker for T2D. Additionally, oligofructose treatment was able to moderately improve glucose homeostasis.
Collapse
Affiliation(s)
- Savanna N. Weninger
- Department of Physiological Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Angela Ding
- Department of Physiological Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Elizabeth N. Browne
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Morgan L. Frost
- School of Animal and Comparative Biomedical Sciences, College of Agricultural and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Gabriele Schiro
- The PANDA Core for Genomics and Microbiome Research, Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | - Daniel Laubitz
- The PANDA Core for Genomics and Microbiome Research, Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | - Frank A. Duca
- School of Animal and Comparative Biomedical Sciences, College of Agricultural and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
47
|
Forsman TT, Paulson AE, Larson EA, Looft T, Lee YJ. On-tissue chemical derivatization of volatile metabolites for matrix-assisted laser desorption/ionization mass spectrometry imaging. JOURNAL OF MASS SPECTROMETRY : JMS 2023; 58:e4918. [PMID: 37045444 DOI: 10.1002/jms.4918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Mass spectrometry imaging (MSI) of volatile metabolites is challenging, especially in matrix-assisted laser desorption/ionization (MALDI). Most MALDI ion sources operate in vacuum, which leads to the vaporization of volatile metabolites during analysis. In addition, tissue samples are often dried during sample preparation, leading to the loss of volatile metabolites even for other MSI techniques. On-tissue chemical derivatization can dramatically reduce the volatility of analytes. Herein, a derivatization method is proposed utilizing N,N,N-trimethyl-2-(piperazin-1-yl)ethan-1-aminium iodide to chemically modify short-chain fatty acids in chicken cecum, ileum, and jejunum tissue sections before sample preparation for MSI visualization.
Collapse
Affiliation(s)
- Trevor T Forsman
- Department of Chemistry, Iowa State University, Ames, Iowa, 50011, USA
| | - Andrew E Paulson
- Department of Chemistry, Iowa State University, Ames, Iowa, 50011, USA
| | - Evan A Larson
- Department of Chemistry, Iowa State University, Ames, Iowa, 50011, USA
| | - Torey Looft
- National Animal Disease Center, Agricultural Research Services, United States Department of Agriculture, Ames, Iowa, 50010, USA
| | - Young Jin Lee
- Department of Chemistry, Iowa State University, Ames, Iowa, 50011, USA
| |
Collapse
|
48
|
Li M, Lu P, Wu H, de Souza TSP, Suleria HAR. In vitro digestion and colonic fermentation of phenolic compounds and their bioaccessibility from raw and roasted nut kernels. Food Funct 2023; 14:2727-2739. [PMID: 36852611 DOI: 10.1039/d2fo03392e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Roasting and digestion affect nut kernel phenolic compounds' bioaccessibility and bioactivity. In this study, three types of raw and commercially roasted nut kernels (almonds, cashews, and walnuts) were treated by in vitro digestion and colonic fermentation. The objective was to analyze the effect of roasting on their phenolic content, associated antioxidant potential, bioaccessibility, and short chain fatty acid (SCFA) synthesis altering. Among these, raw and roasted walnuts performed best, with significantly higher total phenolic content (TPC), total flavonoid content (TFC), free radical scavenging (2,2'-diphenyl-1-picrylhydrazyl (DPPH) assay) values, and ferric reducing antioxidant power (FRAP) values after completing gastrointestinal digestion. With the exception of cashews, roasting had no significant effect on antioxidant capacity during digestion from oral to small intestinal phase. Almonds showed the highest DPPH values after 16-hour colonic fermentation, reaching above 7.60 mg TE per g. Roasting had a positive effect on the free radical savagery capacity of walnuts within 16-24 hours of fecal fermentation. Significant differences were found in the bioaccessibility of individual compounds in raw and roasted nuts. As for almond and walnut, roasting increases the release and breakdown of phenolic compounds during colonic fermentation and have a positive impact on the bioaccessibility of specific phenolic compounds. The colonic bioaccessibility of most phenolic compounds was the highest. Due to heat polysaccharide breakdown, the total SCFAs produced were limited up to 0.03 mM. Raw almonds produced the most SCFAs at 16-hour fermentation and illustrated more benefits to gut health.
Collapse
Affiliation(s)
- Minhao Li
- School of Agriculture and Food, Faculty of Science, The University of Melbourne, Parkville 3010, VIC, Australia.
| | - Peiyao Lu
- School of Agriculture and Food, Faculty of Science, The University of Melbourne, Parkville 3010, VIC, Australia.
- Wuxi Food Safety Inspection and Test Center, 35 South Changjiang Road, Wuxi, Jiangsu Province, 214000, China
| | - Hanjing Wu
- School of Agriculture and Food, Faculty of Science, The University of Melbourne, Parkville 3010, VIC, Australia.
| | - Thaiza S P de Souza
- School of Agriculture and Food, Faculty of Science, The University of Melbourne, Parkville 3010, VIC, Australia.
| | - Hafiz A R Suleria
- School of Agriculture and Food, Faculty of Science, The University of Melbourne, Parkville 3010, VIC, Australia.
| |
Collapse
|
49
|
Luo S, Hou Y, Xie L, Zhang H, Liu C, Chen T. Effects of microwave on the potential microbiota modulating effects of agro-industrial by-product fibers among different individuals. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
50
|
Shum TF, Wang L, Chiou J. Impact of Plasticizer on the Intestinal Epithelial Integrity and Tissue-Repairing Ability within Cells in the Proximity of the Human Gut Microbiome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2152. [PMID: 36767519 PMCID: PMC9915929 DOI: 10.3390/ijerph20032152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/22/2023] [Accepted: 01/22/2023] [Indexed: 06/18/2023]
Abstract
Toxicological research into the impact of plasticizer on different organs has been reported in the past few decades, while their effects on shifting the gut microbiota and immune cells homeostasis in zebrafish were only studied recently. However, studies on the impact of plasticizer on human gut microbiota are scarce. In this study, we co-incubated healthy human fecal microbiota with different concentrations of Di(2-ethylhexyl) phthalate (DEHP) and di-iso-nonyl phthalate (DINP), analyzed microbial composition by 16S rDNA sequencing, and compared the influence of their derived microbiomes on the human enterocyte (HT-29) and murine macrophage (RAW264.7) cell lines. Microbial diversity is reduced by DEHP treatment in a dose-dependent manner. DEHP treatment reduced the phyla Firmicutes/Bacteroidetes ratio, while DINP treatment promoted Proteobacteria. Expressions of tight/adherens junction genes in HT-29 and anti-inflammatory genes in RAW264.7 were down-regulated by plasticizer-co-incubated microbiota derived metabolites. Overall, it is observed that selected plasticizers at high dosages can induce compositional changes in human microbiota. Metabolites from such altered microbiota could affect the tight junction integrity of the intestinal epithelium and upset macrophage differentiation homeostasis in proximity. Chronic exposure to these plasticizers may promote risks of dysbiosis, leaky gut or the exacerbation of intestinal inflammation.
Collapse
Affiliation(s)
- Tim-Fat Shum
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Liwen Wang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Jiachi Chiou
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|