1
|
Peng K, Xiao S, Xia S, Li C, Yu H, Yu Q. Butyrate Inhibits the HDAC8/NF-κB Pathway to Enhance Slc26a3 Expression and Improve the Intestinal Epithelial Barrier to Relieve Colitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24400-24416. [PMID: 39440960 DOI: 10.1021/acs.jafc.4c04456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Dietary fiber is known to promote the production of short-chain fatty acids (SCFAs) by gut bacteria, which can enhance intestinal epithelial barrier function and ameliorate intestinal inflammation in patients with inflammatory bowel disease (IBD). Interestingly, some IBD patients show reduced expression of solute carrier family member 3 (Slc26a3) in intestinal epithelial cells. The objective of this research was to investigate the interaction between SCFAs and Slc26a3 during colitis and assess how this interaction affects intestinal epithelial barrier function. We showed that butyrate alleviated colonic inflammation in a dose-dependent manner in a dextran sulfate sodium salt (DSS)-induced colitis model. Consistent with this, butyrate increased Slc26a3 and tight junction protein levels. In addition, butyrate inhibited histone deacetylase (HDAC) levels and significantly increased the expression of Slc26a3 by the acetylation of histones in Caco-2BBe cells. The utilization of a pan-HDAC inhibitor or inhibitors specific to certain classes of HDACs revealed that butyrate primarily suppressed HDAC8 to blunt the NF-κB pathways and enhance the expression of Slc26a3. Notably, we demonstrated that HDAC8 activation counteracted the beneficial effect of butyrate in DSS-induced colitis. Therefore, we concluded that butyrate improves the expression of Slc26a3 via inhibition of the HDAC8/NF-κB pathway, leading to increased intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Kaixin Peng
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Siqi Xiao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Congxin Li
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Hongbing Yu
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City 66160, Kansas, United States
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver V6H 3N1, British Columbia, Canada
| | - Qin Yu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
2
|
Wang X, Liu H, Yue M, Wang J, Zhang C, Qin L, Wang S, Hu L. Dietary nitrate maintains intestinal epithelia homeostasis in aged mice. Biogerontology 2024; 25:1171-1187. [PMID: 39162978 PMCID: PMC11486781 DOI: 10.1007/s10522-024-10127-5] [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: 06/04/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The intestinal tract, which is the primary site of digestion and absorption of nutrients, is one of the most vulnerable organs during aging. Dietary nitrate, which is mainly derived from the diet and absorbed in the intestinal tract, is a key messenger that connecting oral and general health. However, whether dietary nitrate regulates intestinal tract homeostasis remains unclear. Our data revealed that the serum and salivary nitrate levels decreased during mice aging. The functional proteins of the epithelial barrier (E-cadherin, Claudin-1 and Zonula Occludens-1) in the colon tissues decreased during the aging process. Long-term nitrate supplement in drinking water restored the serum and salivary nitrate levels and increased the functional proteins expression of the colon epithelial barrier. Dietary nitrates increase the relative abundance of some intestinal probiotics, particularly those associated with the production of short-chain fatty acids, such as Blautia, Alloprevotella, Butyricicoccus, and Ruminococcaceae, while promoting the butyric acid production in the colon. Moreover, the expression of Sialin (encoded by Slc17a5), which is a nitrate transporter, increased in the colon epithelial cells by nitrate supplementation. The epithelial cell-conditional Slc17a5-knockout mutant mice (K14-cre; Slc17a5fl/fl) revealed that the functional proteins expression of the colon epithelial barrier and the proliferation of PCNA-positive intestinal epithelial cells in the colon crypts was significantly decreased compared with those of the K14-cre; Slc17a5fl/+ mice. Taken together, our findings suggested that nitrate supplementations were associated with the increased expression of colonic epithelial barriers-related proteins and the increased Sialin expression. Nitrate may serve as a potential therapeutic approach in maintaining aged colonic homeostasis.
Collapse
Affiliation(s)
- Xue Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Huan Liu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Mingwei Yue
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Jinsong Wang
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Chunmei Zhang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Lizheng Qin
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
- Department of Oral and Maxillofacial & Head and Neck Oncology, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Songlin Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China.
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, You An Men Wai, Beijing, 100069, People's Republic of China.
| | - Lei Hu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
- Department of Prosthodontics, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China.
| |
Collapse
|
3
|
Tan X, Wang B, Zhou X, Liu C, Wang C, Bai J. Fecal fermentation behaviors of Konjac glucomannan and its impacts on human gut microbiota. Food Chem X 2024; 23:101610. [PMID: 39071938 PMCID: PMC11282934 DOI: 10.1016/j.fochx.2024.101610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Dietary fiber targets the regulation of the intestinal flora and thus affects host health, however, the complex relationship between these factors lacks direct evidence. In this study, the regulatory effects of Konjac glucomannan (KGM) on key metabolites of host intestinal flora were examined by using in vitro fermentation. The results showed that KGM could be utilized by the intestinal flora, which inhibited the relative abundance of Paeniclostridium, Lachnoclostridium, Phascolarctobacterium, and Bacteroides and enriched the relative abundance of Desulfovibrio, Sutterella, etc. Fermentation is accompanied by the production of short-chain acids, including acetic and propionic acids. Metabolomics revealed that KGM significantly promoted amino acid metabolism, lipid metabolism, and the biosynthesis of other secondary metabolites. Correlation analysis results showed that the increase of panose and N-(1-carboxy-3-carboxanilidopropyl) alanylproline content was positively correlated with the relative abundance of Megamonas. These results provide evidence that KGM affects host health by regulating gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Xiang Tan
- Citrus Research Institute, Southwest University, Chongqing, 400700, China
| | - Botao Wang
- Bloomage Biotechnology CO, LTD, Jinan, Shandong, 250000, China
| | - Xu Zhou
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Cuiping Liu
- Department of Radiology, Yuxi Children's Hospital, Yuxi, Yunnan 653100, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing, 400700, China
| |
Collapse
|
4
|
Roessler J, Zimmermann F, Heidecker B, Landmesser U, Haghikia A. Gut microbiota-related modulation of immune mechanisms in post-infarction remodelling and heart failure. ESC Heart Fail 2024. [PMID: 39385474 DOI: 10.1002/ehf2.14991] [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: 05/03/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 10/12/2024] Open
Abstract
The immune system has long been recognized as a key driver in the progression of heart failure (HF). However, clinical trials targeting immune effectors have consistently failed to improve patient outcome across different HF aetiologies. The activation of the immune system in HF is complex, involving a broad network of pro-inflammatory and immune-modulating components, which complicates the identification of specific immune pathways suitable for therapeutic targeting. Increasing attention has been devoted to identifying gut microbial pathways that affect cardiac remodelling and metabolism and, thereby impacting the development of HF. In particular, gut microbiota-derived metabolites, absorbed by the host and transported to the peripheral circulation, can act as signalling molecules, influencing metabolism and immune homeostasis. Recent reports suggest that the gut microbiota plays a crucial role in modulating immune processes involved in HF. Here, we summarize recent advances in understanding the contributory role of gut microbiota in (auto-)immune pathways that critically determine the progression or alleviation of HF. We also thoroughly discuss potential gut microbiota-based intervention strategies to treat or decelerate HF progression.
Collapse
Affiliation(s)
- Johann Roessler
- University Hospital St Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Friederike Zimmermann
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin, Berlin Institute of Health (BIH), Berlin, Germany
| | - Arash Haghikia
- University Hospital St Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin, Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
5
|
Islam MM, Mahbub NU, Hong ST, Chung HJ. Gut bacteria: an etiological agent in human pathological conditions. Front Cell Infect Microbiol 2024; 14:1291148. [PMID: 39439902 PMCID: PMC11493637 DOI: 10.3389/fcimb.2024.1291148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 08/12/2024] [Indexed: 10/25/2024] Open
Abstract
Through complex interactions with the host's immune and physiological systems, gut bacteria play a critical role as etiological agents in a variety of human diseases, having an impact that extends beyond their mere presence and affects the onset, progression, and severity of the disease. Gaining a comprehensive understanding of these microbial interactions is crucial to improving our understanding of disease pathogenesis and creating tailored treatment methods. Correcting microbial imbalances may open new avenues for disease prevention and treatment approaches, according to preliminary data. The gut microbiota exerts an integral part in the pathogenesis of numerous health conditions, including metabolic, neurological, renal, cardiovascular, and gastrointestinal problems as well as COVID-19, according to recent studies. The crucial significance of the microbiome in disease pathogenesis is highlighted by this role, which is comparable to that of hereditary variables. This review investigates the etiological contributions of the gut microbiome to human diseases, its interactions with the host, and the development of prospective therapeutic approaches. To fully harness the benefits of gut microbiome dynamics for improving human health, future research should address existing methodological challenges and deepen our knowledge of microbial interactions.
Collapse
Affiliation(s)
- Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
6
|
Wang T, Wang RX, Colgan SP. Physiologic hypoxia in the intestinal mucosa: a central role for short-chain fatty acids. Am J Physiol Cell Physiol 2024; 327:C1087-C1093. [PMID: 39159391 PMCID: PMC11482044 DOI: 10.1152/ajpcell.00472.2024] [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: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
The intestinal mucosa is a dynamic surface that facilitates interactions between the host and an outside world that includes trillions of microbes, collectively termed the microbiota. This fine balance is regulated by an energetically demanding physical and biochemical barrier that is formed by the intestinal epithelial cells. In addition, this homeostasis exists at an interface between the anaerobic colonic lumen and a highly oxygenated, vascularized lamina propria. The resultant oxygen gradient within the intestine establishes "physiologic hypoxia" as a central metabolic feature of the mucosa. Although oxygen is vital for energy production to meet cellular metabolism needs, the availability of oxygen has far-reaching influences beyond just energy provision. Recent studies have shown that the intestinal mucosa has purposefully adapted to use differential oxygen levels largely through the presence of short-chain fatty acids (SCFAs), particularly butyrate (BA). Intestinal epithelial cells use butyrate for a multitude of functions that promote mucosal homeostasis. In this review, we explore how the physiologic hypoxia profile interfaces with SCFAs to benefit host mucosal tissues.
Collapse
Affiliation(s)
- Timothy Wang
- Mucosal Inflammation Program, Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Healthcare Studies, University of Texas Dallas, Richardson, Texas, United States
| | - Ruth X Wang
- Mucosal Inflammation Program, Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Dermatology, University of California San Diego, San Diego, California, United States
| | - Sean P Colgan
- Mucosal Inflammation Program, Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, United States
| |
Collapse
|
7
|
Chen WL, Dong YZ, Zhang L, Liu ZS, He CF, Liu WB, Li XF. Xylooligosaccharides alleviate the carbohydrate-enriched diet-induced intestinal barrier dysfunction in carp Megalobrama amblycephala by promoting intestinal development, immunity and gut microbiota. Int J Biol Macromol 2024; 277:134346. [PMID: 39094883 DOI: 10.1016/j.ijbiomac.2024.134346] [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: 03/17/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
To date, although the high-carbohydrate (HC) feed has been extensively adopted in the aquaculture industry, its effects on the intestinal function and development of aquatic animals still remain unclear. In addition, the corresponding nutritional intervention is still barely reported. This study aimed to evaluate the influence of xylooligosaccharides (XOS) on the intestinal health of Megalobrama amblycephala subjected to a HC feeding. Fish (average weight: 44.55 ± 0.15 g) were randomly offered 3 diets, including a control one (29 % carbohydrate), a HC one (41 % carbohydrate), and a XOS supplemented one (HC + 1.0 % XOS, HCX) respectively for 12 weeks. The HC feeding caused morphological abnormalities of intestine, an increased intestinal permeability, and the intestinal immunosuppression, all of which were markedly reversed by XOS administration. In addition, compared with the HC group, HCX feeding remarkably promoted the intestinal activities of digestive and brush border enzymes, and the expressions of cell proliferation-related proteins (Wnt10b and Cyclin D1). The 16s rDNA sequencing also revealed that XOS administration increased the abundance of beneficial bacteria, and decreased that of pathogenic ones. In conclusion, dietary supplementation of XOS improved the intestinal histomorphology, barrier function, cell proliferation and bacterial communities of carbohydrate-overloaded fish Megalobrama amblycephala.
Collapse
Affiliation(s)
- Wei-Liang Chen
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China
| | - Yan-Zou Dong
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China
| | - Ling Zhang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China
| | - Zi-Shang Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China
| | - Chao-Fan He
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China
| | - Wen-Bin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China
| | - Xiang-Fei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing 210095, PR China.
| |
Collapse
|
8
|
Guryanova SV. Bacteria and Allergic Diseases. Int J Mol Sci 2024; 25:10298. [PMID: 39408628 PMCID: PMC11477026 DOI: 10.3390/ijms251910298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Microorganisms colonize all barrier tissues and are present on the skin and all mucous membranes from birth. Bacteria have many ways of influencing the host organism, including activation of innate immunity receptors by pathogen-associated molecular patterns and synthesis of various chemical compounds, such as vitamins, short-chain fatty acids, bacteriocins, toxins. Bacteria, using extracellular vesicles, can also introduce high-molecular compounds, such as proteins and nucleic acids, into the cell, regulating the metabolic pathways of the host cells. Epithelial cells and immune cells recognize bacterial bioregulators and, depending on the microenvironment and context, determine the direction and intensity of the immune response. A large number of factors influence the maintenance of symbiotic microflora, the diversity of which protects hosts against pathogen colonization. Reduced bacterial diversity is associated with pathogen dominance and allergic diseases of the skin, gastrointestinal tract, and upper and lower respiratory tract, as seen in atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, food allergies, and asthma. Understanding the multifactorial influence of microflora on maintaining health and disease determines the effectiveness of therapy and disease prevention and changes our food preferences and lifestyle to maintain health and active longevity.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; ; Tel.: +7-(915)3150073
- Medical Institute, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| |
Collapse
|
9
|
Niu B, Feng Y, Cheng X, Xiao Y, Zhao J, Lu W, Tian F, Chen W. The alleviative effects of viable and inactive Lactobacillus paracasei CCFM1120 against alcoholic liver disease via modulation of gut microbiota and the Nrf2/HO-1 and TLR4/MyD88/NF-κB pathways. Food Funct 2024; 15:8797-8809. [PMID: 39114922 DOI: 10.1039/d4fo02592j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Probiotics can alleviate alcoholic liver disease. However, whether inactive counterparts can produce similar outcomes requires further investigation. We investigated the effects of viable (V) and dead (D) Lactobacillus paracasei CCFM1120 on alcohol-induced ALD mice. The results showed that CCFM1120V and D ameliorated the disease symptoms and intestinal injury. Specifically, these interventions strengthened the intestinal barrier, as evidenced by the increased expression of ZO-1 (zonula occludens 1), occludin, and claudin-1 in the colon and the restored ileal microstructure, including the villi and crypts. In addition, they enhanced the antioxidant capacity of the liver by reducing the production of malondialdehyde and increasing the levels of glutathione and superoxide dismutase. The activation of Nrf2 and HO-1 may be responsible for recovering the antioxidant capacity. Interventions can decrease mouse TNF-α, IL-6 and IL-1β content in serum, probably through the TLR4/MyD88/NF-κB pathway. Furthermore, they possess the ability to restore the quantities of bacteria responsible for producing butyric acid, such as Lactobacillus, Blautia, Bifidobacterium, Ruminococcaceae, Faecalibaculum and Lachnospiraceae. Taken together, CCFM1120V and D apparently can modify the composition of the gut microbiota, foster the gastrointestinal equilibrium, fortify the intestinal barrier, augment the antioxidant capacity of the liver, and effectively shield it from ethanol-induced injury.
Collapse
Affiliation(s)
- Ben Niu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yingxuan Feng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xu Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
10
|
Wang P, Wang S, Wang D, Li Y, Yip RCS, Chen H. Postbiotics-peptidoglycan, lipoteichoic acid, exopolysaccharides, surface layer protein and pili proteins-Structure, activity in wounds and their delivery systems. Int J Biol Macromol 2024; 274:133195. [PMID: 38885869 DOI: 10.1016/j.ijbiomac.2024.133195] [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: 03/20/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
Chronic wound healing is a pressing global public health concern. Abuse and drug resistance of antibiotics are the key problems in the treatment of chronic wounds at present. Postbiotics are a novel promising strategy. Previous studies have reported that postbiotics have a wide range of biological activities including antimicrobial, immunomodulatory, antioxidant and anti-inflammatory abilities. However, several aspects related to these postbiotic activities remain unexplored or poorly known. Therefore, this work aims to outline general aspects and emerging trends in the use of postbiotics for wound healing, such as the production, characterization, biological activities and delivery strategies of postbiotics. In this review, a comprehensive overview of the physiological activities and structures of postbiotic biomolecules that contribute to wound healing is provided, such as peptidoglycan, lipoteichoic acid, bacteriocins, exopolysaccharides, surface layer proteins, pili proteins, and secretory proteins (p40 and p75 proteins). Considering the presence of readily degradable components in postbiotics, potential natural polymer delivery materials and delivery systems are emphasized, followed by the potential applications and commercialization prospects of postbiotics. These findings suggest that the treatment of chronic wounds with postbiotic ingredients will help provide new insights into wound healing and better guidance for the development of postbiotic products.
Collapse
Affiliation(s)
- Pu Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Shuxin Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Donghui Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Yuanyuan Li
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Stocking Hall, 411 Tower Road, Ithaca, NY 14853, USA.
| | - Ryan Chak Sang Yip
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord St, Toronto, ON M5S 3G5, Canada.
| | - Hao Chen
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| |
Collapse
|
11
|
Wang N, Zhang C, Yang L, Min R, Wang X. In vitro fecal fermentation of acylated porous Canna edulis starch and corresponding stabilized Pickering emulsions. Int J Biol Macromol 2024; 274:133169. [PMID: 38885854 DOI: 10.1016/j.ijbiomac.2024.133169] [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: 03/18/2024] [Revised: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
In this study, acylated porous Canna edulis starch with varying degrees of substitution (DS) were prepared and employed for stabilizing Pickering emulsions. Subsequently, the fermentation characteristics of them were investigated. Enzymatically produced porous starch (PS) was esterified with acetic, propionic, butyric, or valeric anhydrides, yielding acetylated (PSA-0.116), propionylated (PSP-0.163), butyrylated (PSB-0.304), and valerylated PS (PSV-0.462) with different DS. Scanning electron microscopy revealed the presence of pores and surface micro-particles in the modified PS, confirming successful esterification through characteristic peaks in 1H NMR and a CO peak at 1736 cm-1 in the FT-IR spectrum. With increasing DS, starch exhibited reduced crystallinity (PSV, 26.61 %), elevated resistant starch content (PSV, 91.63 %), and a higher contact angle (PSV, 87.13°). Acylated PS particles effectively stabilized Pickering emulsions. Pickering emulsions stabilized by acylated PS with higher DS exhibited higher emulsification index and smaller droplet sizes. In vitro fermentation of acylated PS and corresponding stabilized Pickering emulsions fostered short-chain fatty acid production, boosted the relative abundance of beneficial bacteria (Bifidobacterium, Prevotella, etc.) while inhibited the growth of harmful bacteria (Escherichia-Shigella, Comamonas, etc.), maintaining the intestinal microbiota balance. These findings support the potential applications of acylated PS and corresponding stabilized Pickering emulsions in functional foods and drug delivery.
Collapse
Affiliation(s)
- Nan Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Chi Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Li Yang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Rongting Min
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China.
| |
Collapse
|
12
|
Yan L, Ye B, Yang M, Shan Y, Yan D, Fang D, Zhang K, Yu Y. Gut microbiota and metabolic changes in children with idiopathic short stature. BMC Pediatr 2024; 24:468. [PMID: 39039462 PMCID: PMC11265363 DOI: 10.1186/s12887-024-04944-3] [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: 10/14/2023] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Idiopathic short stature (ISS) is characterized by short stature with unknown causes. Recent studies showed different gut microbiota flora and reduced fecal short-chain fatty acids in ISS children. However, the roles of the microbiome and metabolites in the pathogenesis of ISS remains largely unknown. METHODS We recruited 51 Chinese subjects, comprising 26 ISS children and 25 normal-height control individuals. Untargeted metabolomics was performed to explore the fecal metabolic profiles between groups. A shotgun metagenomic sequencing approach was used to investigate the microbiome at the strains level. Mediation analyses were done to reveal correlations between the height standard deviation (SD) value, the gut microbiome and metabolites. RESULTS We detected marked differences in the composition of fecal metabolites in the ISS group, particularly a significant increase in erucic acid and a decrease in spermidine, adenosine and L-5-Hydroxytryptophan, when compared to those of controls. We further identified specific groups of bacterial strains to be associated with the different metabolic profile. Through mediation analysis, 50 linkages were established. KEGG pathway analysis of microbiota and metabolites indicated nutritional disturbances. 13 selected features were able to accurately distinguish the ISS children from the controls (AUC = 0.933 [95%CI, 79.9-100%]) by receiver operating characteristic (ROC) analysis. CONCLUSION Our study suggests that the microbiome and the microbial-derived metabolites play certain roles in children's growth. These findings provide a new research direction for better understanding the mechanism(s) underlying ISS.
Collapse
Affiliation(s)
- Luyan Yan
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Ye
- Department of Pediatric Internal Medicine, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| | - Min Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongsheng Shan
- Department of Pediatrics, Xiaoshan Hospital Affiliated to Hangzhou Normal University, Hangzhou, China
| | - Dan Yan
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - DanFeng Fang
- Department of Pediatric Internal Medicine, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| | - Kaichuang Zhang
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongguo Yu
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Ji H, Zhong Y, Zhang Z, Chen Y, Zhang Y, Bian S, Yin J, Hu J, Nie S. In vitro digestion and fermentation characteristics of eight kinds of pulses and suggestions for different populations. Food Funct 2024; 15:7314-7332. [PMID: 38898712 DOI: 10.1039/d4fo00551a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Pulse-based diets are attracting attention for their potential in combating diet-related non-communicable diseases. However, limited research studies have focused on the digestive and fermentative properties of pulses, which are crucial for exerting benefits. Here, we investigated the in vitro digestibility of starch/protein, along with the fermentation characteristics, of eight pulses and their pastes, including white kidney beans, adzuki beans, cowpeas, broad beans, mung beans, chickpeas, white lentils, and yellow peas. The findings indicated that pulse flours and pastes were low GL food (estimated GL < 10) and had a low degree of protein hydrolysis during simulated gastrointestinal digestion. During in vitro fermentation, pulses flours and pastes decreased the fermentation pH, increased the level of short-chain fatty acids (mainly consisting of valeric acid, followed by acetic acid, propionic acid, butyric acid, isobutyric acid, and isovaleric acid), and positively modulated the microbiota composition over time, specifically reducing the ratio of Firmicutes to Bacteroidetes. In addition, we found that boiling could affect the in vitro digestion and fermentation characteristics of pulses, possibly depending on their intrinsic nutrient characteristics. This research could provide a comprehensive summary of the nutrient content, digestibility, and fermentation of eight pulses and their pastes. Guided by factor analysis, for different individuals' consumption, pulses, cowpeas, broad beans, white lentils, and white kidney beans were preferred for diabetic individuals, yellow peas and white lentils were preferred for intestinal homeostasis disorders, and white lentils, broad beans, white kidney beans, and cowpeas were suitable for obese individuals, in which white lentils were considered healthier and suggested for healthy adults.
Collapse
Affiliation(s)
- Haihua Ji
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yadong Zhong
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Ziyi Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yu Chen
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yanli Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Shuigen Bian
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Junyi Yin
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Jielun Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
14
|
Kim S, Seo SU, Kweon MN. Gut microbiota-derived metabolites tune host homeostasis fate. Semin Immunopathol 2024; 46:2. [PMID: 38990345 PMCID: PMC11239740 DOI: 10.1007/s00281-024-01012-x] [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: 12/13/2023] [Accepted: 03/15/2024] [Indexed: 07/12/2024]
Abstract
The gut microbiota, housing trillions of microorganisms within the gastrointestinal tract, has emerged as a critical regulator of host health and homeostasis. Through complex metabolic interactions, these microorganisms produce a diverse range of metabolites that substantially impact various physiological processes within the host. This review aims to delve into the intricate relationships of gut microbiota-derived metabolites and their influence on the host homeostasis. We will explore how these metabolites affect crucial aspects of host physiology, including metabolism, mucosal integrity, and communication among gut tissues. Moreover, we will spotlight the potential therapeutic applications of targeting these metabolites to restore and sustain host equilibrium. Understanding the intricate interplay between gut microbiota and their metabolites is crucial for developing innovative strategies to promote wellbeing and improve outcomes of chronic diseases.
Collapse
Affiliation(s)
- Seungil Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine / Asan Medical Center, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine / Asan Medical Center, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Li Y, He P, Chen Y, Hu J, Deng B, Liu C, Yu B, Dong W. Microbial metabolite sodium butyrate enhances the anti-tumor efficacy of 5-fluorouracil against colorectal cancer by modulating PINK1/Parkin signaling and intestinal flora. Sci Rep 2024; 14:13063. [PMID: 38844824 PMCID: PMC11156851 DOI: 10.1038/s41598-024-63993-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: 03/04/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent global health issue, with 5-fluorouracil (5-FU) being a commonly used chemotherapeutic agent for its treatment. However, the efficacy of 5-FU is often hindered by drug tolerance. Sodium butyrate (NaB), a derivative of intestinal flora, has demonstrated anti-cancer properties both in vitro and in vivo through pro-apoptotic effects and has shown promise in improving outcomes when used in conjunction with traditional chemotherapy agents. This study seeks to evaluate the impact and potential mechanisms of NaB in combination with 5-FU on CRC. We employed a comprehensive set of assays, including CCK-8, EdU staining, Hoechst 33258 staining, flow cytometry, ROS assay, MMP assay, immunofluorescence, and mitophagy assay, to detect the effect of NaB on the biological function of CRC cells in vitro. Western blotting and immunohistochemistry were used to verify the above experimental results. The xenograft tumor model was established to evaluate the in vivo anti-CRC activity of NaB. Subsequently, 16S rRNA gene sequencing was used to analyze the intestinal flora. The findings of our study demonstrate that sodium butyrate (NaB) exerts inhibitory effects on tumor cell proliferation and promotes tumor cell apoptosis in vitro, while also impeding tumor progression in vivo through the enhancement of the mitophagy pathway. Furthermore, the combined treatment of NaB and 5-fluorouracil (5-FU) yielded superior therapeutic outcomes compared to monotherapy with either agent. Moreover, this combination therapy resulted in the specific enrichment of Bacteroides, LigiLactobacillus, butyric acid-producing bacteria, and acetic acid-producing bacteria in the intestinal microbiota. The improvement in the intestinal microbiota contributed to enhanced therapeutic outcomes and reduced the adverse effects of 5-FU. Taken together, these findings indicate that NaB, a histone acetylation inhibitor synthesized through intestinal flora fermentation, has the potential to significantly enhance the therapeutic efficacy of 5-FU in CRC treatment and improve the prognosis of CRC patients.
Collapse
Affiliation(s)
- Yangbo Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Pengzhan He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Ying Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Jiaming Hu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Chuan Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Baoping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
16
|
Han X, Hu X, Jin W, Liu G. Dietary nutrition, intestinal microbiota dysbiosis and post-weaning diarrhea in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:188-207. [PMID: 38800735 PMCID: PMC11126776 DOI: 10.1016/j.aninu.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 05/29/2024]
Abstract
Weaning is a critical transitional point in the life cycle of piglets. Early weaning can lead to post-weaning syndrome, destroy the intestinal barrier function and microbiota homeostasis, cause diarrhea and threaten the health of piglets. The nutritional components of milk and solid foods consumed by newborn animals can affect the diversity and structure of their intestinal microbiota, and regulate post-weaning diarrhea in piglets. Therefore, this paper reviews the effects and mechanisms of different nutrients, including protein, dietary fiber, dietary fatty acids and dietary electrolyte balance, on diarrhea and health of piglets by regulating intestinal function. Protein is an essential nutrient for the growth of piglets; however, excessive intake will cause many harmful effects, such as allergic reactions, intestinal barrier dysfunction and pathogenic growth, eventually aggravating piglet diarrhea. Dietary fiber is a nutrient that alleviates post-weaning diarrhea in piglets, which is related to its promotion of intestinal epithelial integrity, microbial homeostasis and the production of short-chain fatty acids. In addition, dietary fatty acids and dietary electrolyte balance can also facilitate the growth, function and health of piglets by regulating intestinal epithelial function, immune system and microbiota. Thus, a targeted control of dietary components to promote the establishment of a healthy bacterial community is a significant method for preventing nutritional diarrhea in weaned piglets.
Collapse
Affiliation(s)
- Xuebing Han
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410125, China
| | - Xiangdong Hu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China
| | - Wei Jin
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410125, China
| |
Collapse
|
17
|
Kean IRL, Clark JA, Zhang Z, Daubney E, White D, Ferrando-Vivas P, Milla G, Cuthbertson B, Pappachan J, Klein N, Mouncey P, Rowan K, Myburgh J, Gouliouris T, Baker S, Parkhill J, Pathan N, Arctic Research Team. Short-duration selective decontamination of the digestive tract infection control does not contribute to increased antimicrobial resistance burden in a pilot cluster randomised trial (the ARCTIC Study). Gut 2024; 73:910-921. [PMID: 38253478 PMCID: PMC11103307 DOI: 10.1136/gutjnl-2023-330851] [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: 08/04/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
OBJECTIVE Selective decontamination of the digestive tract (SDD) is a well-studied but hotly contested medical intervention of enhanced infection control. Here, we aim to characterise the changes to the microbiome and antimicrobial resistance (AMR) gene profiles in critically ill children treated with SDD-enhanced infection control compared with conventional infection control. DESIGN We conducted shotgun metagenomic microbiome and resistome analysis on serial oropharyngeal and faecal samples collected from critically ill, mechanically ventilated patients in a pilot multicentre cluster randomised trial of SDD. The microbiome and AMR profiles were compared for longitudinal and intergroup changes. Of consented patients, faecal microbiome baseline samples were obtained in 89 critically ill children. Additionally, samples collected during and after critical illness were collected in 17 children treated with SDD-enhanced infection control and 19 children who received standard care. RESULTS SDD affected the alpha and beta diversity of critically ill children to a greater degree than standard care. At cessation of treatment, the microbiome of SDD patients was dominated by Actinomycetota, specifically Bifidobacterium, at the end of mechanical ventilation. Altered gut microbiota was evident in a subset of SDD-treated children who returned late longitudinal samples compared with children receiving standard care. Clinically relevant AMR gene burden was unaffected by the administration of SDD-enhanced infection control compared with standard care. SDD did not affect the composition of the oral microbiome compared with standard treatment. CONCLUSION Short interventions of SDD caused a shift in the microbiome but not of the AMR gene pool in critically ill children at the end mechanical ventilation, compared with standard antimicrobial therapy.
Collapse
Affiliation(s)
| | - John A Clark
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Zhenguang Zhang
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Esther Daubney
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Deborah White
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | | | | | | | | | | | - John Myburgh
- The George Institute for Global Health, Newtown, New South Wales, Australia
| | | | - Stephen Baker
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Nazima Pathan
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
18
|
PRAMONO A, ARDIARIA M, LIMIJADI EKS, NOER ER, LESTARI ES, SISWANTO FM. Intermittent fasting modulates human gut microbiota diversity in a phenotype-dependent manner: a systematic review. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:170-182. [PMID: 38966051 PMCID: PMC11220331 DOI: 10.12938/bmfh.2023-111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/11/2024] [Indexed: 07/06/2024]
Abstract
Cumulative evidence suggests that intermittent fasting (IF) has beneficial effects on human metabolic health. It has been indicated that its impact on the gut microbiota may mediate these beneficial effects. As a result, we hypothesized that IF may impact the human gut microbiota. A systematic review was carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) protocol using the PubMed, Scopus, and CINAHL databases. We registered our systematic review protocol in PROSPERO under registration number CRD42021270050. Human intervention studies published until April 30, 2023, were included. The quality of the included studies was assessed using National Institutes of Health (NIH) quality assessment study tools for intervention studies. The search in the database returned 166 studies, of which 13 matched all criteria for the final qualitative analysis. The body of evidence suggests that IF modulates human gut microbiota alpha and beta diversity in lean (relatively healthy) and relatively healthy overweight/obese individuals but not in individuals with metabolic syndrome. Furthermore, IF also alters human gut microbiota composition in all phenotypes. Of interest, the gut microbiota taxa or microbial metabolites after an IF intervention are associated with metabolic markers. According to this review, IF influences the diversity and taxonomic levels of the human gut microbiota. Individual metabolic phenotypes may alter the effect of IF on the diversity and taxonomic levels of the gut microbiota.
Collapse
Affiliation(s)
- Adriyan PRAMONO
- Department of Nutrition Science, Faculty of Medicine,
Universitas Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak Pos 1269,
Indonesia
- Center of Nutrition Research (CENURE), Nutrition and
Metabolism Research Group, Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak
Pos 1269, Indonesia
| | - Martha ARDIARIA
- Department of Nutrition Science, Faculty of Medicine,
Universitas Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak Pos 1269,
Indonesia
- Center of Nutrition Research (CENURE), Nutrition and
Metabolism Research Group, Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak
Pos 1269, Indonesia
| | - Edward Kurnia Setiawan LIMIJADI
- Department of Clinical Pathology, Faculty of Medicine,
Universitas Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak Pos 1269,
Indonesia
| | - Etika Ratna NOER
- Department of Nutrition Science, Faculty of Medicine,
Universitas Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak Pos 1269,
Indonesia
- Center of Nutrition Research (CENURE), Nutrition and
Metabolism Research Group, Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak
Pos 1269, Indonesia
| | - Endang Sri LESTARI
- Department of Medical Microbiology, Faculty of Medicine,
Universitas Diponegoro, Jalan Prof. Soedarto, SH Tembalang, Semarang Kotak Pos 1269,
Indonesia
| | - Ferbian Milas SISWANTO
- Department of Chemistry and Biochemistry, School of Medicine
and Health Sciences, Atma Jaya Catholic University of Indonesia, Jl. Pluit Raya No.2
Jakarta Utara, Jakarta Utara, DKI Jakarta 14440, Indonesia
| |
Collapse
|
19
|
Ding F, Zhou N, Luo Y, Wang T, Li W, Qiao F, Du Z, Zhang M. Probiotic Pediococcus pentosaceus restored gossypol-induced intestinal barrier injury by increasing propionate content in Nile tilapia. J Anim Sci Biotechnol 2024; 15:54. [PMID: 38582865 PMCID: PMC10999087 DOI: 10.1186/s40104-024-01011-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/06/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Intestinal barrier is a dynamic interface between the body and the ingested food components, however, dietary components or xenobiotics could compromise intestinal integrity, causing health risks to the host. Gossypol, a toxic component in cottonseed meal (CSM), caused intestinal injury in fish or other monogastric animals. It has been demonstrated that probiotics administration benefits the intestinal barrier integrity, but the efficacy of probiotics in maintaining intestinal health when the host is exposed to gossypol remains unclear. Here, a strain (YC) affiliated to Pediococcus pentosaceus was isolated from the gut of Nile tilapia (Oreochromis niloticus) and its potential to repair gossypol-induced intestinal damage was evaluated. RESULTS A total of 270 Nile tilapia (2.20 ± 0.02 g) were allotted in 3 groups with 3 tanks each and fed with 3 diets including CON (control diet), GOS (control diet containing 300 mg/kg gossypol) and GP (control diet containing 300 mg/kg gossypol and 108 colony-forming unit (CFU)/g P. pentosaceus YC), respectively. After 10 weeks, addition of P. pentosaceus YC restored growth retardation and intestinal injury induced by gossypol in Nile tilapia. Transcriptome analysis and siRNA interference experiments demonstrated that NOD-like receptors (NLR) family caspase recruitment domain (CARD) domain containing 3 (Nlrc3) inhibition might promote intestinal stem cell (ISC) proliferation, as well as maintaining gut barrier integrity. 16S rRNA sequencing and gas chromatography-mass spectrometry (GC-MS) revealed that addition of P. pentosaceus YC altered the composition of gut microbiota and increased the content of propionate in fish gut. In vitro studies on propionate's function demonstrated that it suppressed nlrc3 expression and promoted wound healing in Caco-2 cell model. CONCLUSIONS The present study reveals that P. pentosaceus YC has the capacity to ameliorate intestinal barrier injury by modulating gut microbiota composition and elevating propionate level. This finding offers a promising strategy for the feed industry to incorporate cottonseed meal into fish feed formulations.
Collapse
Affiliation(s)
- Feifei Ding
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Nannan Zhou
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuan Luo
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Tong Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Weijie Li
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhenyu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Meiling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
20
|
Yang J, Qin K, Wang Q, Yang X. Deciphering the nutritional strategies for polysaccharides effects on intestinal barrier in broilers: Selectively promote microbial ecosystems. Int J Biol Macromol 2024; 264:130677. [PMID: 38458298 DOI: 10.1016/j.ijbiomac.2024.130677] [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: 09/07/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
The gut microbiota, a complex and dynamic microbial ecosystem, plays a crucial role in regulating the intestinal barrier. Polysaccharide foraging is specifically dedicated to establishing and maintaining microbial communities, contributing to the shaping of the intestinal ecosystem and ultimately enhancing the integrity of the intestinal barrier. The utilization and regulation of individual polysaccharides often rely on distinct gut-colonizing bacteria. The products of their metabolism not only benefit the formation of the ecosystem but also facilitate cross-feeding partnerships. In this review, we elucidate the mechanisms by which specific bacteria degrade polysaccharides, and how polysaccharide metabolism shapes the microbial ecosystem through cross-feeding. Furthermore, we explore how selectively promoting microbial ecosystems and their metabolites contributes to improvements in the integrity of the intestinal barrier.
Collapse
Affiliation(s)
- Jiantao Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Kailong Qin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qianggang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
21
|
Kim H, Jo JH, Lee HG, Park W, Lee HK, Park JE, Shin D. Inflammatory response in dairy cows caused by heat stress and biological mechanisms for maintaining homeostasis. PLoS One 2024; 19:e0300719. [PMID: 38527055 PMCID: PMC10962848 DOI: 10.1371/journal.pone.0300719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Climate change increases global temperatures, which is lethal to both livestock and humans. Heat stress is known as one of the various livestock stresses, and dairy cows react sensitively to high-temperature stress. We aimed to better understand the effects of heat stress on the health of dairy cows and observing biological changes. Individual cows were divided into normal (21-22 °C, 50-60% humidity) and high temperature (31-32 °C, 80-95% humidity), respectively, for 7-days. We performed metabolomic and transcriptome analyses of the blood and gut microbiomes of feces. In the high-temperature group, nine metabolites including linoleic acid and fructose were downregulated, and 154 upregulated and 72 downregulated DEGs (Differentially Expressed Genes) were identified, and eighteen microbes including Intestinimonas and Pseudoflavonifractor in genus level were significantly different from normal group. Linoleic acid and fructose have confirmed that associated with various stresses, and functional analysis of DEG and microorganisms showing significant differences confirmed that high-temperature stress is related to the inflammatory response, immune system, cellular energy mechanism, and microbial butyrate production. These biological changes were likely to withstand high-temperature stress. Immune and inflammatory responses are known to be induced by heat stress, which has been identified to maintain homeostasis through modulation at metabolome, transcriptome and microbiome levels. In these findings, heat stress condition can trigger alteration of immune system and cellular energy metabolism, which is shown as reduced metabolites, pathway enrichment and differential microbes. As results of this study did not include direct phenotypic data, we believe that additional validation is required in the future. In conclusion, high-temperature stress contributed to the reduction of metabolites, changes in gene expression patterns and composition of gut microbiota, which are thought to support dairy cows in withstanding high-temperature stress via modulating immune-related genes, and cellular energy metabolism to maintain homeostasis.
Collapse
Affiliation(s)
- Hana Kim
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Jang-Hoon Jo
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Hong-Gu Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Woncheoul Park
- Division of Animal Genomics and Bioinformatics, National Institute of Animal Science, Rural Development Administration, Wanju, Jeollabuk-do, Republic of Korea
| | - Hak-Kyo Lee
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Jong-Eun Park
- Department of Animal Biotechnology, College of Applied Life Science, Jeju National University, Jeju, Jeju-do, Republic of Korea
| | - Donghyun Shin
- Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| |
Collapse
|
22
|
Trasciatti S, Grizzi F. Vitamin D and celiac disease. ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 109:249-270. [PMID: 38777415 DOI: 10.1016/bs.afnr.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Celiac disease (CD) is an immune-mediated condition affecting the small intestine. Its reported global prevalence falls within the range of 0.7% to 1.4%. Notably, historically, higher rates, reaching 1% in Western Ireland, have been documented. Recent research has even revealed prevalence rates as elevated as 2% in northern Europe. These findings underscore the urgency for swift and cost-effective diagnosis, especially in individuals identified through screening efforts. At present, the diagnosis of CD relies on a multifaceted approach involving positive serological markers such as IgA anti-tissue transglutaminase (anti-TTG) and anti-endomysial antibodies (anti-EMA). These serological findings are assessed in conjunction with classical histological alterations, as outlined in the Marsh classification. CD is an inflammatory condition triggered by the consumption of gluten, resulting from intricate interactions between genetic, immunological, and environmental factors. CD is linked to malabsorption, leading to nutritional deficiencies. Individuals with CD are required to adhere to a gluten-free diet, which itself can lead to nutrient deficiencies. One such deficiency includes vitamin D, and there is substantial experimental evidence supporting the notion of a bidirectional relationship between CD and vitamin D status. A low level of vitamin D has a detrimental impact on the clinical course of the disease. Here we summarize the key characteristics of CD and explore the prominent roles of vitamin D in individuals with CD.
Collapse
Affiliation(s)
| | - Fabio Grizzi
- Head Histology Core, Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
23
|
Dong Z, Liu Z, Xu Y, Tan B, Sun W, Ai Q, Yang Z, Zeng J. Potential for the development of Taraxacum mongolicum aqueous extract as a phytogenic feed additive for poultry. Front Immunol 2024; 15:1354040. [PMID: 38529273 PMCID: PMC10961442 DOI: 10.3389/fimmu.2024.1354040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Taraxacum mongolicum (TM) is a kind of medicinal and edible homologous plant which is included in the catalogue of feed raw materials in China. It is rich in polyphenols, flavonoids, polysaccharides and other active substances, and shows many benefits to livestock, poultry and aquatic products. The study aimed to assess the potential of TM aqueous extract (TMAE) as a substitute for poultry AGPs. Methods A total of 240 one-day-old Arbor Acker broilers were randomly assigned to four groups and fed a basal diet (Con) supplemented with 500, 1000, and 2000 mg/kg TMAE (Low, Medium, and High groups). The growth performance of the broilers was measured on day 21 and day 42. At the end of the trial, the researchers measured slaughter performance and collected serum, liver, spleen, ileum, and intestinal contents to investigate the effects of TMAE on serum biochemistry, antioxidant capacity, immune function, organ coefficient, intestinal morphology, flora composition, and short-chain fatty acids (SCFAs). Results The results showed that broilers treated with TMAE had a significantly higher average daily gain from 22 to 42 days old compared to the Con group. Various doses of TMAE resulted in different levels of improvement in serum chemistry. High doses increased serum alkaline phosphatase and decreased creatinine. TMAE also increased the antioxidant capacity of serum, liver, and ileum in broilers. Additionally, middle and high doses of TMAE enhanced the innate immune function of the liver (IL-10) and ileum (Occludin) in broilers. Compared to the control group, the TMAE treatment group exhibited an increase in the ratio of villi length to villi crypt in the duodenum. TMAE increased the abundance of beneficial bacteria, such as Alistipes and Lactobacillus, while reducing the accumulation of harmful bacteria, such as Colidextracter and Sellimonas. The cecum's SCFAs content increased with a medium dose of TMAE. Supplementing broiler diets with TMAE at varying doses enhanced growth performance and overall health. The most significant benefits were observed at a dose of 1000 mg/kg, including improved serum biochemical parameters, intestinal morphology, antioxidant capacity of the liver and ileum, immune function of the liver and ileum, and increased SCFAs content. Lactobacillus aviarius, norank_f_norank_o__Clostridia_UCG-014, and Flavonifractor are potentially dominant members of the intestinal microflora. Conclusion In conclusion, TMAE is a promising poultry feed additive and 1000 mg/kg is an effective reference dose.
Collapse
Affiliation(s)
- Zhen Dong
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Zhiqin Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Yufeng Xu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Bin Tan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Wenqing Sun
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Qin Ai
- DHN Business Division, Wens Foodstuff Group Co., Ltd., Zhaoqing, China
| | - Zihui Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Jianguo Zeng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Province Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| |
Collapse
|
24
|
Schreiber F, Balas I, Robinson MJ, Bakdash G. Border Control: The Role of the Microbiome in Regulating Epithelial Barrier Function. Cells 2024; 13:477. [PMID: 38534321 DOI: 10.3390/cells13060477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/28/2024] Open
Abstract
The gut mucosal epithelium is one of the largest organs in the body and plays a critical role in regulating the crosstalk between the resident microbiome and the host. To this effect, the tight control of what is permitted through this barrier is of high importance. There should be restricted passage of harmful microorganisms and antigens while at the same time allowing the absorption of nutrients and water. An increased gut permeability, or "leaky gut", has been associated with a variety of diseases ranging from infections, metabolic diseases, and inflammatory and autoimmune diseases to neurological conditions. Several factors can affect gut permeability, including cytokines, dietary components, and the gut microbiome. Here, we discuss how the gut microbiome impacts the permeability of the gut epithelial barrier and how this can be harnessed for therapeutic purposes.
Collapse
|
25
|
Castro C, Niknafs S, Gonzalez-Ortiz G, Tan X, Bedford MR, Roura E. Dietary xylo-oligosaccharides and arabinoxylans improved growth efficiency by reducing gut epithelial cell turnover in broiler chickens. J Anim Sci Biotechnol 2024; 15:35. [PMID: 38433214 PMCID: PMC10910751 DOI: 10.1186/s40104-024-00991-z] [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: 09/14/2023] [Accepted: 01/02/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND One of the main roles of the intestinal mucosa is to protect against environmental hazards. Supplementation of xylo-oligosaccharides (XOS) is known to selectively stimulate the growth of beneficial intestinal bacteria and improve gut health and function in chickens. XOS may have an impact on the integrity of the intestinal epithelia where cell turnover is critical to maintain the compatibility between the digestive and barrier functions. The aim of the study was to evaluate the effect of XOS and an arabinoxylan-rich fraction (AXRF) supplementation on gut function and epithelial integrity in broiler chickens. METHODS A total of 128 broiler chickens (Ross 308) were assigned into one of two different dietary treatments for a period of 42 d: 1) control diet consisting of a corn/soybean meal-based diet; or 2) a control diet supplemented with 0.5% XOS and 1% AXRF. Each treatment was randomly distributed across 8 pens (n = 8) with 8 chickens each. Feed intake and body weight were recorded weekly. On d 42, one male chicken per pen was selected based on average weight and euthanized, jejunum samples were collected for proteomics analysis. RESULTS Dietary XOS/AXRF supplementation improved feed efficiency (P < 0.05) from d 1 to 42 compared to the control group. Proteomic analysis was used to understand the mechanism of improved efficiency uncovering 346 differentially abundant proteins (DAP) (Padj < 0.00001) in supplemented chickens compared to the non-supplemented group. In the jejunum, the DAP translated into decreased ATP production indicating lower energy expenditure by the tissue (e.g., inhibition of glycolysis and tricarboxylic acid cycle pathways). In addition, DAP were associated with decreased epithelial cell differentiation, and migration by reducing the actin polymerization pathway. Putting the two main pathways together, XOS/AXRF supplementation may decrease around 19% the energy required for the maintenance of the gastrointestinal tract. CONCLUSIONS Dietary XOS/AXRF supplementation improved growth efficiency by reducing epithelial cell migration and differentiation (hence, turnover), actin polymerization, and consequently energy requirement for maintenance of the jejunum of broiler chickens.
Collapse
Affiliation(s)
- Carla Castro
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Shahram Niknafs
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | | | - Xinle Tan
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | | | - Eugeni Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
26
|
Stepanova M, Aherne CM. Adenosine in Intestinal Epithelial Barrier Function. Cells 2024; 13:381. [PMID: 38474346 DOI: 10.3390/cells13050381] [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: 12/23/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
At the intestinal front, several lines of defense are in place to resist infection and injury, the mucus layer, gut microbiome and strong epithelial junctions, to name a few. Their collaboration creates a resilient barrier. In intestinal disorders, such as inflammatory bowel disease (IBD), barrier function is compromised, which results in rampant inflammation and tissue injury. In response to the destruction, the intestinal epithelium releases adenosine, a small but powerful nucleoside that functions as an alarm signal. Amidst the chaos of inflammation, adenosine aims to restore order. Within the scope of its effects is the ability to regulate intestinal epithelial barrier integrity. This review aims to define the contributions of adenosine to mucus production, microbiome-dependent barrier protection, tight junction dynamics, chloride secretion and acid-base balance to reinforce its importance in the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Mariya Stepanova
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Carol M Aherne
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
27
|
Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW, Khaw KY. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 2024; 9:37. [PMID: 38360862 PMCID: PMC10869798 DOI: 10.1038/s41392-024-01743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 02/17/2024] Open
Abstract
The human gastrointestinal tract is populated with a diverse microbial community. The vast genetic and metabolic potential of the gut microbiome underpins its ubiquity in nearly every aspect of human biology, including health maintenance, development, aging, and disease. The advent of new sequencing technologies and culture-independent methods has allowed researchers to move beyond correlative studies toward mechanistic explorations to shed light on microbiome-host interactions. Evidence has unveiled the bidirectional communication between the gut microbiome and the central nervous system, referred to as the "microbiota-gut-brain axis". The microbiota-gut-brain axis represents an important regulator of glial functions, making it an actionable target to ameliorate the development and progression of neurodegenerative diseases. In this review, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases. As the gut microbiome provides essential cues to microglia, astrocytes, and oligodendrocytes, we examine the communications between gut microbiota and these glial cells during healthy states and neurodegenerative diseases. Subsequently, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases using a metabolite-centric approach, while also examining the role of gut microbiota-related neurotransmitters and gut hormones. Next, we examine the potential of targeting the intestinal barrier, blood-brain barrier, meninges, and peripheral immune system to counteract glial dysfunction in neurodegeneration. Finally, we conclude by assessing the pre-clinical and clinical evidence of probiotics, prebiotics, and fecal microbiota transplantation in neurodegenerative diseases. A thorough comprehension of the microbiota-gut-brain axis will foster the development of effective therapeutic interventions for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Wen Qi Mak
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Chu Xin Ng
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Hong Hao Chan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Shiau Hueh Yeow
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
28
|
Zhao XC, Ju B, Xiu NN, Sun XY, Meng FJ. When inflammatory stressors dramatically change, disease phenotypes may transform between autoimmune hematopoietic failure and myeloid neoplasms. Front Immunol 2024; 15:1339971. [PMID: 38426096 PMCID: PMC10902444 DOI: 10.3389/fimmu.2024.1339971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Aplastic anemia (AA) and hypoplastic myelodysplastic syndrome are paradigms of autoimmune hematopoietic failure (AHF). Myelodysplastic syndrome and acute myeloid leukemia are unequivocal myeloid neoplasms (MNs). Currently, AA is also known to be a clonal hematological disease. Genetic aberrations typically observed in MNs are detected in approximately one-third of AA patients. In AA patients harboring MN-related genetic aberrations, a poor response to immunosuppressive therapy (IST) and an increased risk of transformation to MNs occurring either naturally or after IST are predicted. Approximately 10%-15% of patients with severe AA transform the disease phenotype to MNs following IST, and in some patients, leukemic transformation emerges during or shortly after IST. Phenotypic transformations between AHF and MNs can occur reciprocally. A fraction of advanced MN patients experience an aplastic crisis during which leukemic blasts are repressed. The switch that shapes the disease phenotype is a change in the strength of extramedullary inflammation. Both AHF and MNs have an immune-active bone marrow (BM) environment (BME). In AHF patients, an inflamed BME can be evoked by infiltrated immune cells targeting neoplastic molecules, which contributes to the BM-specific autoimmune impairment. Autoimmune responses in AHF may represent an antileukemic mechanism, and inflammatory stressors strengthen antileukemic immunity, at least in a significant proportion of patients who have MN-related genetic aberrations. During active inflammatory episodes, normal and leukemic hematopoieses are suppressed, which leads to the occurrence of aplastic cytopenia and leukemic cell regression. The successful treatment of underlying infections mitigates inflammatory stress-related antileukemic activities and promotes the penetration of leukemic hematopoiesis. The effect of IST is similar to that of treating underlying infections. Investigating inflammatory stress-powered antileukemic immunity is highly important in theoretical studies and clinical practice, especially given the wide application of immune-activating agents and immune checkpoint inhibitors in the treatment of hematological neoplasms.
Collapse
Affiliation(s)
- Xi-Chen Zhao
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Bo Ju
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Nuan-Nuan Xiu
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Xiao-Yun Sun
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Fan-Jun Meng
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
29
|
Ikeda E, Yamaguchi M, Kawabata S. Gut Microbiota-mediated Alleviation of Dextran Sulfate Sodium-induced Colitis in Mice. GASTRO HEP ADVANCES 2024; 3:461-470. [PMID: 39131720 PMCID: PMC11308119 DOI: 10.1016/j.gastha.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/29/2024] [Indexed: 08/13/2024]
Abstract
Background and Aims Gut dysbiosis characterized by an imbalanced microbiota is closely involved in the pathogenesis of a widespread gastrointestinal inflammatory disorder, inflammatory bowel disease. However, it is unclear how the complex intestinal microbiota affects development or resistant of mucosal inflammation. Our aim was to investigate the impact of the gut microbiota on susceptibility in a mouse model of ulcerative colitis. Methods We compared the susceptibility to dextran sulfate sodium (DSS)-induced colitis of inbred BALB/c mice obtained from the 3 main distributors of laboratory animals in Japan. Clinical symptoms of the colitis and the faecal microbiota were assessed. Cohousing approach was used to identify whether the gut microbiota is a primary factor determining disease susceptibility. Results Here, we showed differences in the susceptibility of BALB/c mice from the vendors to DSS colitis. Analysis of the gut microbiota using 16S ribosomal RNA sequencing revealed clear separation of the gut microbial composition among mice from the vendors. Notably, the abundance of the phylum Actinobacteriota was strongly associated with disease activity. We also observed the expansion of butyrate-producing Roseburia species in mice with decreased susceptibility of the disease. Further cohousing experiments showed that variation in clinical outcomes was more correlated with the gut microbiota than genetic variants among substrains from different suppliers. Conclusion A BALB/c substrain that was resistant to DSS-induced colitis was observed, and the severity of DSS-induced colitis was mainly influenced by the gut microbiota. Targeting butyrate-producing bacteria could have therapeutic potential for ulcerative colitis.
Collapse
Affiliation(s)
- Eri Ikeda
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Suita, Osaka, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Suita, Osaka, Japan
- Bioinformatics Research Unit, Graduates School of Dentistry, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
- Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Graduates School of Dentistry, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
30
|
Chen S, Cai X, Lao L, Wang Y, Su H, Sun H. Brain-Gut-Microbiota Axis in Amyotrophic Lateral Sclerosis: A Historical Overview and Future Directions. Aging Dis 2024; 15:74-95. [PMID: 37307822 PMCID: PMC10796086 DOI: 10.14336/ad.2023.0524] [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: 04/11/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease which is strongly associated with age. The incidence of ALS increases from the age of 40 and peaks between the ages of 65 and 70. Most patients die of respiratory muscle paralysis or lung infections within three to five years of the appearance of symptoms, dealing a huge blow to patients and their families. With aging populations, improved diagnostic methods and changes in reporting criteria, the incidence of ALS is likely to show an upward trend in the coming decades. Despite extensive researches have been done, the cause and pathogenesis of ALS remains unclear. In recent decades, large quantities of studies focusing on gut microbiota have shown that gut microbiota and its metabolites seem to change the evolvement of ALS through the brain-gut-microbiota axis, and in turn, the progression of ALS will exacerbate the imbalance of gut microbiota, thereby forming a vicious cycle. This suggests that further exploration and identification of the function of gut microbiota in ALS may be crucial to break the bottleneck in the diagnosis and treatment of this disease. Hence, the current review summarizes and discusses the latest research advancement and future directions of ALS and brain-gut-microbiota axis, so as to help relevant researchers gain correlative information instantly.
Collapse
Affiliation(s)
- Shilan Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Xinhong Cai
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Lin Lao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Yuxuan Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Huanxing Su
- Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau.
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Paiano L, Mastronardi M, Campisciano G, Rosso N, Casagranda B, Comar M, de Manzini N, Palmisano S. Liver Bacterial Colonization in Patients with Obesity and Gut Dysbiosis. Obes Surg 2024; 34:402-408. [PMID: 38102371 DOI: 10.1007/s11695-023-06989-2] [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: 10/01/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
PURPOSE Recently, the link between gut microbiota, liver inflammation, and obesity has become an interesting focus of research. The aim of this study is to show the possible relation between gut microbiota dysbiosis in patients with obesity and the presence of bacterial genomes in their liver biopsies. MATERIALS AND METHODS A prospective study on patients undergoing bariatric surgery was carried out. Anthropometric and metabolic data, comorbidities, stool samples, and hepatic biopsies were collected and analyzed at the time of surgery. The V3-16S rRNA region was sequenced using the Ion Torrent new-generation sequencing platform. RESULTS In each of the 23 patients enrolled, the bacterial population was analyzed both in the stools and liver. In eight patients (34.7%), Prevotella (62.5%), Bacteroides (50%), Streptococcus (12.5%), and Dalister (12.5%) were found in both samples, simultaneously; in 15 cases, the liver was free from colonization. The statistically significant difference between groups was a Roseburia intestinalis reduction in fecal samples of patients with liver biopsies colonized by bacteria (1% vs 3%; p = 0.0339). CONCLUSION To the best of our knowledge, this is the first study reporting the presence of bacterial genome in a liver biopsy on bariatric patients, instead of the microbe-associated molecular patterns. Notably, in literature, the presence of Roseburia intestinalis in stool samples has been shown to prevent intestinal inflammation playing its role in the gut barrier integrity. In our population, the Roseburia reduction was associated with the presence of bacterial genome in the liver, probably related to a greater permeability of the gut and vascular barriers.
Collapse
Affiliation(s)
- Lucia Paiano
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Manuela Mastronardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy.
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy.
| | - Giuseppina Campisciano
- Institute for Maternal and Child Health IRCCS, Burlo Garofolo, Via dell' Istria 65/1, 34149, Trieste, Italy
| | - Natalia Rosso
- Fondazione Italiana Fegato, Centro Studi Fegato, Area Science Park Basovizza Bldg.Q SS14 Km, 163.5, 34149, Trieste, Italy
| | - Biagio Casagranda
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Manola Comar
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Institute for Maternal and Child Health IRCCS, Burlo Garofolo, Via dell' Istria 65/1, 34149, Trieste, Italy
| | - Nicolò de Manzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Silvia Palmisano
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| |
Collapse
|
32
|
Xu H, Wen Q, Hu H, Yang S, Lu L, Hu X, Li H, Huang X, Li N. Electroacupuncture at ST36 modulates the intestinal microecology and may help repair the intestinal barrier in the rat model of severe acute pancreatitis. Microb Biotechnol 2024; 17:e14401. [PMID: 38381095 PMCID: PMC10880739 DOI: 10.1111/1751-7915.14401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024] Open
Abstract
Severe acute pancreatitis (SAP) onset and development are closely associated with intestinal barrier injury. Evidence from clinical practice and research has shown that electroacupuncture (EA) at the Zusanli (ST36) acupoint can improve intestinal barrier function and abdominal symptoms in patients with SAP; however, the specific mechanisms of action remain unclear. This study aimed to observe the changes in the intestinal microbiota and metabolites in SAP rats and to explore the effect of EA at ST36 on intestinal barrier injury in SAP rats. 16S rRNA gene sequencing combined with microbial diversity analysis, short-chain fatty acids (SCFAs)-targeted metabolomics, immunohistochemistry, immunofluorescence, western blotting, and other techniques were used to explore the mechanism of EA at bilateral ST36 acupoints on SAP-related intestinal barrier injury. Our results showed that EA at ST36 could repair intestinal barrier injury by modulating intestinal microecology, thereby reducing intestinal inflammation, restoring intestinal function, and ultimately alleviating the prognosis of SAP. Our study provides new insights into the mechanisms and treatment of intestinal barrier injury in patients with SAP from the perspectives of microbiota and SCFAs regulation.
Collapse
Affiliation(s)
- Huimin Xu
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Qian Wen
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Hangqi Hu
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Sihao Yang
- Department of Integrated Traditional Chinese and Western MedicineYibin Second People's HospitalYibinSichuanChina
| | - Lingyun Lu
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xiangyun Hu
- Chengdu University of Traditional Chinese MedicineChengduSichuanChina
| | - Hao Li
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xianhao Huang
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ning Li
- Institute of Integrated Traditional Chinese and Western MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
33
|
Fan X, Zhang Z, Gao W, Pan Q, Luo K, He B, Pu Y. An Engineered Butyrate-Derived Polymer Nanoplatform as a Mucosa-Healing Enhancer Potentiates the Therapeutic Effect of Magnolol in Inflammatory Bowel Disease. ACS NANO 2024; 18:229-244. [PMID: 38112525 DOI: 10.1021/acsnano.3c05732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Colonic epithelial damage and dysregulated immune response are crucial factors in the progression and exacerbation of inflammatory bowel disease (IBD). Nanoenabled targeted drug delivery to the inflamed intestinal mucosa has shown promise in inducing and maintaining colitis remission, while minimizing side effects. Inspired by the excellent antioxidative and anti-inflammatory efficacy of naturally derived magnolol (Mag) and gut homeostasis regulation of microbiota-derived butyrate, we developed a pH/redox dual-responsive butyrate-rich polymer nanoparticle (PSBA) as an oral Mag delivery system for combinational therapy of IBD. PSBA showed a high butyrate content of 22% and effectively encapsulated Mag. The Mag-loaded nanoparticles (PSBA@Mag) demonstrated colonic pH and reduction-responsive drug release, ensuring efficient retention and adhesion in the colon of colitis mice. PSBA@Mag not only normalized the level of reactive oxygen species and inflammatory effectors in inflamed colonic mucosa but also restored the epithelial barrier function in both ulcerative colitis and Crohn's disease mouse models. Importantly, PSBA promoted the migration and healing ability of intestinal epithelial cells in vitro and in vivo, sensitizing the therapeutic efficacy of Mag in animal models. Moreover, transcriptomics and metabolism analyses revealed that PSBA@Mag mitigated inflammation by suppressing the production of pro-inflammatory cytokines and chemokines and restoring the lipid metabolism. Additionally, this nanomedicine modulated the gut microbiota by inhibiting pathogenic Proteus and Escherichia-Shigella and promoting the proliferation of beneficial probiotics, including Lachnoclostridium, Lachnospiraceae_NK4A136_group and norank_f_Ruminococcaceae. Overall, our findings highlight the potential of butyrate-functionalized polymethacrylates as versatile and effective nanoplatforms for colonic drug delivery and mucosa repair in combating IBD and other gastrointestinal disorders.
Collapse
Affiliation(s)
- Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhuangzhuang Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
34
|
Song B, Sun P, Kong L, Xiao C, Pan X, Song Z. The improvement of immunity and activation of TLR2/NF-κB signaling pathway by Romboutsia ilealis in broilers. J Anim Sci 2024; 102:skae286. [PMID: 39305205 DOI: 10.1093/jas/skae286] [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: 07/09/2024] [Accepted: 09/19/2024] [Indexed: 11/09/2024] Open
Abstract
This study was conducted to investigate the effects of Romboutsia ilealis on the immune function of broilers and the underlying mechanisms. A total of 48 one-day-old Arbor Acres broilers were allocated to 4 groups as follows: broilers treated daily with 1 mL live R. ilealis in general anaerobic medium broth media (0, 1 × 104, 1 × 106, and 1 × 108 CFU/mL) from days 1 to 7. Samples were collected on days 8 and 14. The results showed that R. ilealis had no negative effect on the body weight of broilers (P > 0.05). R. ilealis significantly increased the levels of lysozyme, IFN-γ, IFN-γ/IL-4, and IgG in the serum (P < 0.05). R. ilealis significantly increased the levels of IL-4, IFN-γ, sIgA, lysozyme, and iNOS in the ileal mucosa (P < 0.05). R. ilealis significantly increased the mRNA levels of TLR2, TLR4, NF-κB, IL-1β, TNF-α, IFN-γ, IgA, pIgR, iNOS, and MHC-II in the ileum (P < 0.05). R. ilealis significantly increased the relative abundance of Enterococcus and Paracoccus in the jejunum and ileum, ileal Candidatus Arthromitus, and cecal Romboutsia and Intestinimonas (P < 0.05). Correlation analysis showed that Enterococcus, Paracoccus, Romboutsia, and Intestinimonas were significantly positively correlated with humoral immune function (P < 0.05). In conclusion, R. ilealis boosted the immune system, activated the intestinal TLR2/NF-κB signaling pathway, and improved the gut microbiota in broilers.
Collapse
Affiliation(s)
- Bochen Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| | - Peng Sun
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Linglian Kong
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
- Department of Biological and Chemical Engineering, Jining Polytechnic, Jining, Shandong, China
| | - Chuanpi Xiao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
35
|
Zhou R, Huang Y, Feng X, Zhou R, Wang L, Xie G, Xiao Y, Zhou H. Decreased YB-1 expression denervates brown adipose tissue and contributes to age-related metabolic dysfunction. Cell Prolif 2024; 57:e13520. [PMID: 37321837 PMCID: PMC10771110 DOI: 10.1111/cpr.13520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
Thermogenesis in brown adipose tissue (BAT) declines with aging, however, the underlying mechanism remains unclear. Here, we show that the expression of Y-box binding protein 1 (YB-1), a critical DNA/RNA binding protein, decreased in the BAT of aged mice due to the reduction of microbial metabolite butyrate. Genetic ablation of YB-1 in the BAT accelerated diet-induced obesity and BAT thermogenic dysfunction. In contrast, overexpression of YB-1 in the BAT of aged mice was sufficient to promote BAT thermogenesis, thus alleviating diet-induced obesity and insulin resistance. Interestingly, YB-1 had no direct effect on adipose UCP1 expression. Instead, YB-1 promoted axon guidance of BAT via regulating the expression of Slit2, thus potentiating sympathetic innervation and thermogenesis. Moreover, we have identified that a natural compound Sciadopitysin, which promotes YB-1 protein stability and nuclear translocation, alleviated BAT aging and metabolic disorders. Together, we reveal a novel fat-sympathetic nerve unit in regulating BAT senescence and provide a promising strategy against age-related metabolic disorders.
Collapse
Affiliation(s)
- Ruoyu Zhou
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Liwen Wang
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Genqing Xie
- Department of EndocrinologyThe First People's Hospital of Xiangtan cityXiangtanChina
| | - Yuan Xiao
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaChina
| | - Haiyan Zhou
- Department of Endocrinology, Endocrinology Research CenterXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaChina
| |
Collapse
|
36
|
Hu X, He Z, Zhao C, He Y, Qiu M, Xiang K, Zhang N, Fu Y. Gut/rumen-mammary gland axis in mastitis: Gut/rumen microbiota-mediated "gastroenterogenic mastitis". J Adv Res 2024; 55:159-171. [PMID: 36822391 PMCID: PMC10770137 DOI: 10.1016/j.jare.2023.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/25/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Mastitis is an inflammatory response in the mammary gland that results in huge economic losses in the breeding industry. The aetiology of mastitis is complex, and the pathogenesis has not been fully elucidated. It is commonly believed that mastitis is induced by pathogen infection of the mammary gland and induces a local inflammatory response. However, in the clinic, mastitis is often comorbid or secondary to gastric disease, and local control effects targeting the mammary gland are limited. In addition, recent studies have found that the gut/rumen microbiota contributes to the development of mastitis and proposed the gut/rumen-mammary gland axis. Combined with studies indicating that gut/rumen microbiota disturbance can damage the gut mucosa barrier, gut/rumen bacteria and their metabolites can migrate to distal extraintestinal organs. It is believed that the occurrence of mastitis is related not only to the infection of the mammary gland by external pathogenic microorganisms but also to a gastroenterogennic pathogenic pathway. AIM OF REVIEW We propose the pathological concept of "gastroenterogennic mastitis" and believe that the gut/rumen-mammary gland axis-mediated pathway is the pathological mechanism of "gastroenterogennic mastitis". KEY SCIENTIFIC CONCEPTS OF REVIEW To clarify the concept of "gastroenterogennic mastitis" by summarizing reports on the effect of the gut/rumen microbiota on mastitis and the gut/rumen-mammary gland axis-mediated pathway to provide a research basis and direction for further understanding and solving the pathogenesis and difficulties encountered in the prevention of mastitis.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Zhaoqi He
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yuhong He
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Min Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Kaihe Xiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China.
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China.
| |
Collapse
|
37
|
Thiele Orberg E, Meedt E, Hiergeist A, Xue J, Heinrich P, Ru J, Ghimire S, Miltiadous O, Lindner S, Tiefgraber M, Göldel S, Eismann T, Schwarz A, Göttert S, Jarosch S, Steiger K, Schulz C, Gigl M, Fischer JC, Janssen KP, Quante M, Heidegger S, Herhaus P, Verbeek M, Ruland J, van den Brink MRM, Weber D, Edinger M, Wolff D, Busch DH, Kleigrewe K, Herr W, Bassermann F, Gessner A, Deng L, Holler E, Poeck H. Bacteria and bacteriophage consortia are associated with protective intestinal metabolites in patients receiving stem cell transplantation. NATURE CANCER 2024; 5:187-208. [PMID: 38172339 DOI: 10.1038/s43018-023-00669-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 10/13/2023] [Indexed: 01/05/2024]
Abstract
The microbiome is a predictor of clinical outcome in patients receiving allogeneic hematopoietic stem cell transplantation (allo-SCT). Microbiota-derived metabolites can modulate these outcomes. How bacteria, fungi and viruses contribute to the production of intestinal metabolites is still unclear. We combined amplicon sequencing, viral metagenomics and targeted metabolomics from stool samples of patients receiving allo-SCT (n = 78) and uncovered a microbiome signature of Lachnospiraceae and Oscillospiraceae and their associated bacteriophages, correlating with the production of immunomodulatory metabolites (IMMs). Moreover, we established the IMM risk index (IMM-RI), which was associated with improved survival and reduced relapse. A high abundance of short-chain fatty acid-biosynthesis pathways, specifically butyric acid via butyryl-coenzyme A (CoA):acetate CoA-transferase (BCoAT, which catalyzes EC 2.8.3.8) was detected in IMM-RI low-risk patients, and virome genome assembly identified two bacteriophages encoding BCoAT as an auxiliary metabolic gene. In conclusion, our study identifies a microbiome signature associated with protective IMMs and provides a rationale for considering metabolite-producing consortia and metabolite formulations as microbiome-based therapies.
Collapse
Affiliation(s)
- Erik Thiele Orberg
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.
- German Cancer Consortium (DKTK), partner-site Munich, a partnership between DKFZ and Klinikum rechts der Isar, Munich, Germany.
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.
| | - Elisabeth Meedt
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Medical Center, Regensburg, Germany
| | - Jinling Xue
- Institute of Virology, Helmholtz Zentrum Munich, Munich, Germany
- Chair of Prevention for Microbial Infectious Disease, Central Institute of Disease Prevention and School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Paul Heinrich
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Jinlong Ru
- Institute of Virology, Helmholtz Zentrum Munich, Munich, Germany
- Chair of Prevention for Microbial Infectious Disease, Central Institute of Disease Prevention and School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Sakhila Ghimire
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Oriana Miltiadous
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah Lindner
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Melanie Tiefgraber
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Sophia Göldel
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Tina Eismann
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Alix Schwarz
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Sascha Göttert
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- German Cancer Consortium (DKTK), partner-site Munich, a partnership between DKFZ and Klinikum rechts der Isar, Munich, Germany
- Comparative Experimental Pathology, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christian Schulz
- Department of Internal Medicine II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Michael Gigl
- Bavarian Center for Biomolecular Mass Spectrometry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Julius C Fischer
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar TUM, Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar TUM, Munich, Germany
| | - Michael Quante
- Department of Internal Medicine II, University Medical Center, Freiburg, Germany
| | - Simon Heidegger
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Peter Herhaus
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Mareike Verbeek
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Jürgen Ruland
- German Cancer Consortium (DKTK), partner-site Munich, a partnership between DKFZ and Klinikum rechts der Isar, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Marcel R M van den Brink
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Daniela Weber
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Daniel Wolff
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Florian Bassermann
- Department of Internal Medicine III, School of Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
- German Cancer Consortium (DKTK), partner-site Munich, a partnership between DKFZ and Klinikum rechts der Isar, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Medical Center, Regensburg, Germany
| | - Li Deng
- Institute of Virology, Helmholtz Zentrum Munich, Munich, Germany
- Chair of Prevention for Microbial Infectious Disease, Central Institute of Disease Prevention and School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Ernst Holler
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany.
- Leibniz Institute for Immunotherapy, Regensburg, Germany.
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany.
| |
Collapse
|
38
|
Liu L, Kaur GI, Kumar A, Kanwal A, Singh SP. The Role of Gut Microbiota and Associated Compounds in Cardiovascular Health and its Therapeutic Implications. Cardiovasc Hematol Agents Med Chem 2024; 22:375-389. [PMID: 38275032 DOI: 10.2174/0118715257273506231208045308] [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: 08/05/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 01/27/2024]
Abstract
It is possible that gut bacteria may have a beneficial effect on cardiovascular health in humans. It may play a major role in the progression of a variety of cardiovascular diseases, including Heart Failure (HF), Atherosclerosis, Coronary Arterial Disease (CAD), Ischemic Heart Disease (IHD), and Others. Dysbiosis of the gut microbiota, along with its direct and indirect impact on gut health, may induce cardiovascular disorders. Although advanced studies have demonstrated the relationship of various metabolites to cardiovascular diseases (CVD) in animals, translating their functional capacity to humans remains a significant area of research. This paper simplifies the demonstration of some compounds, pathways, and components like Trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and butyrate production. It demonstrates how a change in eating habits causes TMAO and how the impact of different drugs on gut microbiota species and high consumption of Westernized food causes several heartrelated problems, such as atherosclerosis and inflammation that can even become the cause of heart failure. Modulation of the gut microbiome, on the other hand, is a novel therapeutic measure because it can be easily altered through diet and other lifestyle changes. It could then be used to lower the risk of several CVDs.
Collapse
Affiliation(s)
- Lu Liu
- Endoscopic Diagnosis and Treatment Center, Baoding First Central Hospital, Baoding, China
| | - Guneet Inderjeet Kaur
- Department of Sports Psychology, Central University of Rajasthan, Ajmer, 305817, India
| | - Avinash Kumar
- Department of Sports Biosciences, Central University of Rajasthan, Ajmer, 305817, India
| | | | | |
Collapse
|
39
|
Yuan J, Li Y, Sun S, Wu J, Zhou J, He S. Response of growth performance and cecum microbial community to cyclic heat stress in broilers. Trop Anim Health Prod 2023; 56:9. [PMID: 38085433 DOI: 10.1007/s11250-023-03849-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
Heat stress (HS) can affect growth performance through alterations in specific gut microbiota, which greatly threatens poultry production. How HS affects the mechanisms of microbial changes in the poultry cecum and the complex interactions between cecal microbial changes and growth performance have not yet been well evaluated. This study was conducted to examine the changes in growth performance and cecal microbiotal community in cyclic heat stress (CHS)-treated broilers. A total of 200 twenty-eight-day-old female Arbor Acres (AA) broilers were equally allotted into neutral ambient temperature group (TN group, 24 ± 1°C, 24 h/day) and CHS group (33 ± 1°C, 8 h/day) with five replicates of 10 broilers each, respectively. Growth performance, cecum microbial diversity, flora composition, and community structure were analyzed on days 35 and 42. The decreased average daily feed intake (ADFI), average daily gain (ADG), and the increased feed/gain ratio (F:G) were observed in heat-stressed broilers on days 35 and 42. The alpha and beta diversity index had no significant changes at the two experimental periods (P > 0.05). At the genus level, CHS significantly increased the relative abundance of Enterococcus at 42 days (P < 0.05). Based on the analysis of linear effect size feature selection, CHS made an enriched Reyranella and a reduced Romboutsia and Ruminiclostridium at 35 days of age (P < 0.05). CHS made an enriched Weissella and Enterococcus at 42 days of age (P < 0.05). The present study revealed that CHS reduces broiler growth performance and alters the microbial community of the cecum microbiota and the abundance of species. These findings are of critical importance to alleviate the negative effects of CHS on broiler chickens' growth performance by maintaining gut microbial balance.
Collapse
Affiliation(s)
- Junjun Yuan
- College of Animal Science, Anhui Science and Technology University, Fengyang, Anhui, 233100, China
| | - Yan Li
- College of Animal Science, Anhui Science and Technology University, Fengyang, Anhui, 233100, China
| | - Shiang Sun
- College of Animal Science, Anhui Science and Technology University, Fengyang, Anhui, 233100, China
| | - Jiaying Wu
- College of Animal Science, Anhui Science and Technology University, Fengyang, Anhui, 233100, China
| | - Jin Zhou
- College of Animal Science, Anhui Science and Technology University, Fengyang, Anhui, 233100, China
| | - Shaojun He
- College of Animal Science, Anhui Science and Technology University, Fengyang, Anhui, 233100, China.
| |
Collapse
|
40
|
Ornelas A, Welch N, Countess JA, Zhou L, Wang RX, Dowdell AS, Colgan SP. Mimicry of microbially-derived butyrate reveals templates for potent intestinal epithelial HIF stabilizers. Gut Microbes 2023; 15:2267706. [PMID: 37822087 PMCID: PMC10572066 DOI: 10.1080/19490976.2023.2267706] [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: 06/24/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
Microbiota-derived short-chain fatty acids, including butyrate (BA), have multiple beneficial health effects. In the colon, BA concentrations range from 10 to 20 mM and up to 95% is utilized as energy by the mucosa. BA plays a key role in epithelial-barrier regulation and anti-inflammation, and regulates cell growth and differentiation, at least in part, due to its direct influence on stabilization of the transcription factor hypoxia-inducible factor (HIF). It remains unclear whether BA is the optimal metabolite for such a response. In this study, we explored metabolite mimicry as an attractive strategy for the biological response to HIF. We discovered that 4-mercapto butyrate (MBA) stabilizes HIF more potently and has a longer biological half-life than BA in intestinal epithelial cells (IECs). We validated the MBA-mediated HIF transcriptional activity through the induction of classic HIF gene targets in IECs and enhanced epithelial barrier formation in vitro. In-vivo studies with MBA revealed systemic HIF stabilization in mice, which was more potent than its parent BA metabolite. Mechanistically, we found that MBA enhances oxygen consumption and that the sulfhydryl group is essential for HIF stabilization, but exclusively as a four-carbon SCFA. These findings reveal a combined biochemical mechanism for HIF stabilization and provide a foundation for the discovery of potent metabolite-like scaffolds.
Collapse
Affiliation(s)
- Alfredo Ornelas
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
| | - Nichole Welch
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| | - Jacob A. Countess
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
| | - Liheng Zhou
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
| | - Ruth X. Wang
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
| | - Alexander S. Dowdell
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program and Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| |
Collapse
|
41
|
Chandrasekaran P, Han Y, Zerbe CS, Heller T, DeRavin SS, Kreuzberg SA, Marciano BE, Siu Y, Jones DR, Abraham RS, Stephens MC, Tsou AM, Snapper S, Conlan S, Subramanian P, Quinones M, Grou C, Calderon V, Deming C, Leiding JW, Arnold DE, Logan BR, Griffith LM, Petrovic A, Mousallem TI, Kapoor N, Heimall JR, Barnum JL, Kapadia M, Wright N, Rayes A, Chandra S, Broglie LA, Chellapandian D, Deal CL, Grunebaum E, Lim SS, Mallhi K, Marsh RA, Murguia-Favela L, Parikh S, Touzot F, Cowan MJ, Dvorak CC, Haddad E, Kohn DB, Notarangelo LD, Pai SY, Puck JM, Pulsipher MA, Torgerson TR, Kang EM, Malech HL, Segre JA, Bryant CE, Holland SM, Falcone EL. Intestinal microbiome and metabolome signatures in patients with chronic granulomatous disease. J Allergy Clin Immunol 2023; 152:1619-1633.e11. [PMID: 37659505 PMCID: PMC11279821 DOI: 10.1016/j.jaci.2023.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Chronic granulomatous disease (CGD) is caused by defects in any 1 of the 6 subunits forming the nicotinamide adenine dinucleotide phosphate oxidase complex 2 (NOX2), leading to severely reduced or absent phagocyte-derived reactive oxygen species production. Almost 50% of patients with CGD have inflammatory bowel disease (CGD-IBD). While conventional IBD therapies can treat CGD-IBD, their benefits must be weighed against the risk of infection. Understanding the impact of NOX2 defects on the intestinal microbiota may lead to the identification of novel CGD-IBD treatments. OBJECTIVE We sought to identify microbiome and metabolome signatures that can distinguish individuals with CGD and CGD-IBD. METHODS We conducted a cross-sectional observational study of 79 patients with CGD, 8 pathogenic variant carriers, and 19 healthy controls followed at the National Institutes of Health Clinical Center. We profiled the intestinal microbiome (amplicon sequencing) and stool metabolome, and validated our findings in a second cohort of 36 patients with CGD recruited through the Primary Immune Deficiency Treatment Consortium. RESULTS We identified distinct intestinal microbiome and metabolome profiles in patients with CGD compared to healthy individuals. We observed enrichment for Erysipelatoclostridium spp, Sellimonas spp, and Lachnoclostridium spp in CGD stool samples. Despite differences in bacterial alpha and beta diversity between the 2 cohorts, several taxa correlated significantly between both cohorts. We further demonstrated that patients with CGD-IBD have a distinct microbiome and metabolome profile compared to patients without CGD-IBD. CONCLUSION Intestinal microbiome and metabolome signatures distinguished patients with CGD and CGD-IBD, and identified potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Yu Han
- Division of Molecular Genetics and Pathology, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Md; Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Christa S Zerbe
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Theo Heller
- Translational Hepatology Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Md
| | - Suk See DeRavin
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Samantha A Kreuzberg
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Beatriz E Marciano
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Yik Siu
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY
| | - Roshini S Abraham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn; Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | | | - Amy M Tsou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, NY
| | - Scott Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Sean Conlan
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Md
| | - Mariam Quinones
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Md
| | - Caroline Grou
- Bioinformatics Core, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
| | - Virginie Calderon
- Bioinformatics Core, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
| | - Clayton Deming
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, Md
| | - Danielle E Arnold
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Md
| | - Brent R Logan
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wis
| | - Linda M Griffith
- Division of Allergy, Immunology, and Transplantation, NIAID, NIH, Bethesda, Md
| | - Aleksandra Petrovic
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital and Research Center, Seattle, Wash
| | - Talal I Mousallem
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Neena Kapoor
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, Calif
| | - Jennifer R Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jessie L Barnum
- Division of Blood and Marrow Transplantation and Cellular Therapies, University of Pittsburgh Medical Center (UPMC) and Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Malika Kapadia
- Department of Pediatrics, Harvard University Medical School, Boston, Mass
| | - Nicola Wright
- Section of Hematology/Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Ahmad Rayes
- Intermountain Primary Children's Hospital, University of Utah, Salt Lake City, Utah
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Larisa A Broglie
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis
| | - Deepak Chellapandian
- Center for Cell and Gene Therapy for Non-Malignant Conditions, Johns Hopkins All Children's Hospital, St Petersburg, Fla
| | - Christin L Deal
- Division of Allergy and Immunology, UPMC, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Eyal Grunebaum
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Si Lim
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, Hawaii; University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, Hawaii
| | | | - Rebecca A Marsh
- Cincinnati Children's Hospital Medical Center, and University of Cincinnati, Cincinnati, Ohio
| | - Luis Murguia-Favela
- Section of Hematology/Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Suhag Parikh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Fabien Touzot
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Morton J Cowan
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Christopher C Dvorak
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Elie Haddad
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Donald B Kohn
- Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Calif
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Sung-Yun Pai
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Md
| | - Jennifer M Puck
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Michael A Pulsipher
- Division of Pediatric Hematology and Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute at the University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, Utah
| | | | - Elizabeth M Kang
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Harry L Malech
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Julia A Segre
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Clare E Bryant
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Emilia Liana Falcone
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada; Center for Immunity, Inflammation and Infectious Diseases, IRCM, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
42
|
Sereti I, Verburgh ML, Gifford J, Lo A, Boyd A, Verheij E, Verhoeven A, Wit FWNM, Schim van der Loeff MF, Giera M, Kootstra NA, Reiss P, Vujkovic-Cvijin I. Impaired gut microbiota-mediated short-chain fatty acid production precedes morbidity and mortality in people with HIV. Cell Rep 2023; 42:113336. [PMID: 37918403 PMCID: PMC10872975 DOI: 10.1016/j.celrep.2023.113336] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Antiretroviral therapy (ART) has dramatically lengthened lifespan among people with HIV (PWH), but this population experiences heightened rates of inflammation-related comorbidities. HIV-associated inflammation is linked with an altered microbiome; whether such alterations precede inflammation-related comorbidities or occur as their consequence remains unknown. We find that ART-treated PWH exhibit depletion of gut-resident bacteria that produce short-chain fatty acids (SCFAs)-crucial microbial metabolites with anti-inflammatory properties. Prior reports establish that fecal SCFA concentrations are not depleted in PWH. We find that gut-microbiota-mediated SCFA production capacity is better reflected in serum than in feces and that PWH exhibit reduced serum SCFA, which associates with inflammatory markers. Leveraging stool and serum samples collected prior to comorbidity onset, we find that HIV-specific microbiome alterations precede morbidity and mortality in ART-treated PWH. Among these microbiome alterations, reduced microbiome-mediated conversion of lactate to propionate precedes mortality in PWH. Thus, gut microbial fiber/lactate conversion to SCFAs may modulate HIV-associated comorbidity risk.
Collapse
Affiliation(s)
- Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, NIAID/NIH, Rockville, MD, USA; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Myrthe L Verburgh
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Jacob Gifford
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alice Lo
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anders Boyd
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; HIV Monitoring Foundation, Amsterdam, the Netherlands; Public Health Service of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
| | - Eveline Verheij
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Aswin Verhoeven
- Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, the Netherlands
| | - Ferdinand W N M Wit
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; HIV Monitoring Foundation, Amsterdam, the Netherlands
| | - Maarten F Schim van der Loeff
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Public Health Service of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, the Netherlands
| | - Neeltje A Kootstra
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Experimental Immunology, Amsterdam, the Netherlands
| | - Peter Reiss
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands; Amsterdam University Medical Centers, University of Amsterdam, Department of Global Health, Meibergdreef 9, Amsterdam, the Netherlands
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Liao Y, Chen Z, Yang Y, Shen D, Chai S, Ma Y, Ge R, Wang X, Wang S, Liu S. Antibiotic intervention exacerbated oxidative stress and inflammatory responses in SD rats under hypobaric hypoxia exposure. Free Radic Biol Med 2023; 209:70-83. [PMID: 37806597 DOI: 10.1016/j.freeradbiomed.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
The gut microbiota plays a crucial role in maintaining host nutrition, metabolism, and immune homeostasis, particularly in extreme environmental conditions. However, the regulatory mechanisms of the gut microbiota in animal organisms hypobaric hypoxia exposure require further study. We conducted a research by comparing SD rats treated with an antibiotic (ABX) cocktail and untreated SD rats that were housed in a low-pressure oxygen chamber (simulating low pressure and hypoxic environment at 6000 m altitude) for 30 days. After the experiment, blood, feces, and lung tissues from SD rats were collected for analysis of blood, 16S rRNA amplicon sequencing, and non-targeted metabolomics. The results demonstrated that the antibiotic cocktail-treated SD rats exhibited elevated counts of neutrophil (Neu) and monocyte (Mon) cells, an enrichment of sulfate-reducing bacteria (SBC), reduced levels of glutathione, and accumulated phospholipid compounds. Notably, the accumulation of phospholipid compounds, particularly lysophosphatidic acid (LPA), lipopolysaccharide (LPS), and lysophosphatidylcholine (LPC), along with the aforementioned changes, contributed to heightened oxidative stress and inflammation in the organism. In addition, we explored the resistance mechanisms of SD rats in low-oxygen and low-pressure environments and found that increasing the quantity of the Prevotellaceae and related beneficial bacteria (especially Lactobacillus) could reduce oxidative stress and inflammation. These findings offer valuable insights into enhancing the adaptability of low-altitude animals under hypobaric hypoxia exposure.
Collapse
Affiliation(s)
- Yang Liao
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Zheng Chen
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Yingkui Yang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Di Shen
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Shatuo Chai
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Yan Ma
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, China
| | - Rili Ge
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, China
| | - Xun Wang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Shuxiang Wang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China.
| | - Shujie Liu
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China.
| |
Collapse
|
44
|
Alula KM, Dowdell AS, LeBere B, Lee JS, Levens CL, Kuhn KA, Kaipparettu BA, Thompson WE, Blumberg RS, Colgan SP, Theiss AL. Interplay of gut microbiota and host epithelial mitochondrial dysfunction is necessary for the development of spontaneous intestinal inflammation in mice. MICROBIOME 2023; 11:256. [PMID: 37978573 PMCID: PMC10655390 DOI: 10.1186/s40168-023-01686-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/30/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Intestinal epithelial cell (IEC) mitochondrial dysfunction involvement in inflammatory bowel diseases (IBD), including Crohn's disease affecting the small intestine, is emerging in recent studies. As the interface between the self and the gut microbiota, IECs serve as hubs of bidirectional cross-talk between host and luminal microbiota. However, the role of mitochondrial-microbiota interaction in the ileum is largely unexplored. Prohibitin 1 (PHB1), a chaperone protein of the inner mitochondrial membrane required for optimal electron transport chain function, is decreased during IBD. We previously demonstrated that mice deficient in PHB1 specifically in IECs (Phb1i∆IEC) exhibited mitochondrial impairment, Paneth cell defects, gut microbiota dysbiosis, and spontaneous inflammation in the ileum (ileitis). Mice deficient in PHB1 in Paneth cells (epithelial secretory cells of the small intestine; Phb1∆PC) also exhibited mitochondrial impairment, Paneth cell defects, and spontaneous ileitis. Here, we determined whether this phenotype is driven by Phb1 deficiency-associated ileal microbiota alterations or direct effects of loss of PHB1 in host IECs. RESULTS Depletion of gut microbiota by broad-spectrum antibiotic treatment in Phb1∆PC or Phb1i∆IEC mice revealed a necessary role of microbiota to cause ileitis. Using germ-free mice colonized with ileal microbiota from Phb1-deficient mice, we show that this microbiota could not independently induce ileitis without host mitochondrial dysfunction. The luminal microbiota phenotype of Phb1i∆IEC mice included a loss of the short-chain fatty acid butyrate. Supplementation of butyrate in Phb1-deficient mice ameliorated Paneth cell abnormalities and ileitis. Phb1-deficient ileal enteroid models suggest deleterious epithelial-intrinsic responses to ileal microbiota that were protected by butyrate. CONCLUSIONS These results suggest a mutual and essential reinforcing interplay of gut microbiota and host IEC, including Paneth cell, mitochondrial health in influencing ileitis. Restoration of butyrate is a potential therapeutic option in Crohn's disease patients harboring epithelial cell mitochondrial dysfunction. Video Abstract.
Collapse
Affiliation(s)
- Kibrom M Alula
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz Medical Campus, 12700 East 19Th Avenue, RC2 Campus Box BB158 HSC, Aurora, CO, 80045, USA
| | - Alexander S Dowdell
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz Medical Campus, 12700 East 19Th Avenue, RC2 Campus Box BB158 HSC, Aurora, CO, 80045, USA
| | - Brittany LeBere
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz Medical Campus, 12700 East 19Th Avenue, RC2 Campus Box BB158 HSC, Aurora, CO, 80045, USA
| | - J Scott Lee
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz Medical Campus, 12700 East 19Th Avenue, RC2 Campus Box BB158 HSC, Aurora, CO, 80045, USA
| | - Cassandra L Levens
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Benny A Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Winston E Thompson
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean P Colgan
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz Medical Campus, 12700 East 19Th Avenue, RC2 Campus Box BB158 HSC, Aurora, CO, 80045, USA
| | - Arianne L Theiss
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz Medical Campus, 12700 East 19Th Avenue, RC2 Campus Box BB158 HSC, Aurora, CO, 80045, USA.
| |
Collapse
|
45
|
Chen L, Zhao R, Kang Z, Cao Z, Liu N, Shen J, Wang C, Pan F, Zhou X, Liu Z, Yang Y, Chen Q. Delivery of short chain fatty acid butyrate to overcome Fusobacterium nucleatum-induced chemoresistance. J Control Release 2023; 363:43-56. [PMID: 37734673 DOI: 10.1016/j.jconrel.2023.09.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/10/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
The gut microbiota is closely associated with the progression of colorectal cancer (CRC) in which Fusobacterium nucleatum (F. nucleatum) was found to induce cancer resistance to chemotherapeutics. To relieve F. nucleatum-induced drug resistance, herein, we found that short-chain fatty acid butyrate can inhibit the growth, enrichment and adhesion of F. nucleatum in colorectal cancer tissues by downregulating the expression of adhesion-associated outer membrane proteins, including RadD, FomA, and FadA, to reduce the colonization and invasion of F. nucleatum and relieve the chemoresistance induced by F. nucleatum. Leveraging the killing effect of butyrate on F. nucleatum, sodium butyrate (NaBu) was encapsulated in liposomes or prepared as NaBu tablets with Eudragit S100 coating and administered by intravenous injection or oral administration, respectively. Interestingly, both intravenous administration of NaBu liposomes and oral delivery of NaBu tablets could effectively inhibit the proliferation of F. nucleatum and significantly improve the therapeutic efficacy of oxaliplatin in mice with subcutaneous colorectal tumors, orthotopic colorectal tumors and even spontaneously formed colorectal tumors. Thus, our work provides a simple but effective formulation of NaBu to relieve F. nucleatum-induced chemoresistance, exhibiting ideal clinical application prospects.
Collapse
Affiliation(s)
- Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Rui Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Zheyu Kang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Zhiqin Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Nanhui Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Jingjing Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Cheng Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Feng Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Xiao Zhou
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
46
|
Shin Y, Han S, Kwon J, Ju S, Choi TG, Kang I, Kim SS. Roles of Short-Chain Fatty Acids in Inflammatory Bowel Disease. Nutrients 2023; 15:4466. [PMID: 37892541 PMCID: PMC10609902 DOI: 10.3390/nu15204466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The gut microbiome is a diverse bacterial community in the human gastrointestinal tract that plays important roles in a variety of biological processes. Short-chain fatty acids (SCFA) are produced through fermentation of dietary fiber. Certain microbes in the gut are responsible for producing SCFAs such as acetate, propionate and butyrate. An imbalance in gut microbiome diversity can lead to metabolic disorders and inflammation-related diseases. Changes in SCFA levels and associated microbiota were observed in IBD, suggesting an association between SCFAs and disease. The gut microbiota and SCFAs affect reactive oxygen species (ROS) associated with IBD. Gut microbes and SCFAs are closely related to IBD, and it is important to study them further.
Collapse
Affiliation(s)
- Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhui Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
47
|
Chen C, Sang Z, Xie Q, Xue W. Effects of hazelnut protein isolate-induced food allergy on the gut microenvironment in a BALB/c mouse model. Food Funct 2023; 14:8761-8774. [PMID: 37718731 DOI: 10.1039/d3fo02324a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Hazelnuts are reported as among the nuts that cause severe allergic reactions. However, few systematic studies exist on the changes in the gut microenvironment following hazelnut allergy. This study focused on the effects of hazelnut allergy on the duodenum, jejunum, ileum and colon microenvironment in vivo. We established a hazelnut protein isolate (HPI)-allergic mouse model, which was distinguished by the visible allergy symptoms, dropped temperatures and enhanced allergic inflammatory factor levels in serum, such as HPI-specific immunoglobulin E (sIgE), sIgG2a, interleukin-4, histamine, mouse mast cell protease-1, TNF-α, monocyte chemotactic protein-1 and lipopolysaccharide. For HPI sensitized mice, aggravated mast cell degranulation, severe morphologic damage and inflammatory cell infiltration were observed in the duodenum, jejunum, ileum, and colon, while goblet cell numbers were reduced in the duodenum, jejunum and ileum. Secretory IgA of the jejunum and tight junctions of the duodenum and jejunum were decreased significantly after HPI sensitization. There was no remarkable difference in the pH values of small intestinal contents, but the pH values of colonic contents were elevated, which was due to the decreased short-chain fatty acids (mainly acetate, propionate and butyrate) in the colon. The antioxidant capacity of both large and small intestinal contents declined after HPI sensitization, as evidenced by the increased malondialdehyde and decreased superoxide dismutase activity. HPI sensitization induced gut microbiota dysbiosis with decreased α diversity and altered β diversity in colonic contents. Spearman correlation analysis indicated that the increased characteristic genera, namely Bacteroides, Lactobacillus, Alloprevotella, Erysipelatoclostridium, Parabacteroides, and Helicobacter, played potentially synergistic roles in promoting allergy and gut microenvironment dysregulation.
Collapse
Affiliation(s)
- Chen Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Ziqing Sang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Qiang Xie
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Wentong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
48
|
Ke W, Flay KJ, Huang X, Hu X, Chen F, Li C, Yang DA. Polysaccharides from Platycodon grandiflorus attenuates high-fat diet induced obesity in mice through targeting gut microbiota. Biomed Pharmacother 2023; 166:115318. [PMID: 37572640 DOI: 10.1016/j.biopha.2023.115318] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/14/2023] Open
Abstract
The root of Platycodon grandiflorus (PG), abundant in soluble polysaccharides, has a long history in traditional Asian diets and herbal medicine due to its anti-inflammatory activity and anti-obesity effects. Our previous study was the first to establish a link between the beneficial effects of PG and changes in the gut microbiota, and suggested potential roles that the polysaccharide components play. However, more evidence was needed to understand the anti-obesity functions of polysaccharides from PG (PS) and their relationship with the regulation of the gut microbiota. In this study, we first performed an experiment to explore the anti-obesity activities of PS: Male C57BL/6 mice (six-weeks-old) were fed either a standard control diet (CON), or a high-fat diet (HFD) to induce obesity, or a HFD supplemented with PS (HFPS) for 8 weeks. Body weight and food intake were monitored throughout. Lipid metabolism were determined and related gene expression changes in adipose tissues were analyzed by RNA-seq. Amplicon sequencing of the bacterial 16 S rRNA gene was used to explore gut microbiota structure in fecal samples. Then, we performed the second experiment to explore whether the anti-obesity activities of PS were dependent on the regulation of the gut microbiota: Male C57BL/6 mice (six-weeks-old), treated with an antibiotic cocktail to reduce the gut microbial load, were fed either a HFD (A-HFD) or a HFPS (A-HFPS) diet for 8 weeks. Finally, we used in vitro fermentation experiments to verify the effects of PS on the growth and metabolic activities of the gut microbes. We found that PS significantly reduced HFD-induced weight gain and excessive fat accumulation, changed the expression of key genes involved in lipid metabolism, and attenuated HFD-induced changes in the gut microbiota. However, PS did not affect fat accumulation or lipid metabolism in the gut microbiota depleted mice. Overall, our results show that PS has significant effects on the gut microbiota in the mouse model, and the anti-obesity effects of PS are mediated via changes in the gut microbiota composition and metabolic activity.
Collapse
Affiliation(s)
- Weixin Ke
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center of Meat Quality and Safety Control, Nanjing 210095, China; National Key Laboratory of Meat Quality Control and New Resource, Nanjing Agricultural University, Nanjing 210095, China
| | - Kate Jade Flay
- Department of Veterinary Clinical Sciences, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of China
| | - Xiaoning Huang
- Department of bioengineering, University of Illinois at Urbana, Champaign 61801, USA
| | - Xiaosong Hu
- National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Fang Chen
- National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chunbao Li
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center of Meat Quality and Safety Control, Nanjing 210095, China; National Key Laboratory of Meat Quality Control and New Resource, Nanjing Agricultural University, Nanjing 210095, China
| | - Dan Aaron Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
49
|
YAMASAKI M, MIYAMOTO Y, OGAWA K, NISHIYAMA K, TSEND-AYUSH C, LI Y, MATSUSAKI T, NAKANO T, TAKESHITA M, ARIMA Y. Lactiplantibacillus plantarum 06CC2 upregulates intestinal ZO-1 protein and bile acid metabolism in Balb/c mice fed high-fat diet. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 43:13-22. [PMID: 38188659 PMCID: PMC10767321 DOI: 10.12938/bmfh.2023-002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/11/2023] [Indexed: 01/09/2024]
Abstract
The effects of Lactiplantibacillus plantarum 06CC2 (LP06CC2), which was isolated from a Mongolian dairy product, on lipid metabolism and intestinal tight junction-related proteins in Balb/c mice fed a high-fat diet (HFD) were evaluated. The mice were fed the HFD for eight weeks, and the plasma and hepatic lipid parameters, as well as the intestinal tight junction-related factors, were evaluated. LP06CC2 slightly reduced the adipose tissue mass. Further, it dose-dependently decreased plasma total cholesterol (TC). The HFD tended to increase the plasma level of endotoxin and suppressed intestinal ZO-1 expression, whereas a low LP06CC2 dose increased ZO-1 expression and tended to reduce the plasma lipopolysaccharide level. Furthermore, a low LP06CC2 dose facilitated a moderate accumulation of Lactobacillales, a significant decrease in Clostridium cluster IV, and an increase in Clostridium cluster XVIII. The results obtained from analyzing the bile acids (BAs) in feces and cecum contents exhibited a decreasing trend for secondary and conjugated BAs in the low LP06CC2-dose group. Moreover, a high LP06CC2 dose caused excess accumulation of Lactobacillales and failed to increase intestinal ZO-1 and occludin expression, while the fecal butyrate level increased dose dependently in the LP06CC2-fed mice. Finally, an appropriate LP06CC2 dose protected the intestinal barrier function from the HFD and modulated BA metabolism.
Collapse
Affiliation(s)
- Masao YAMASAKI
- Graduate School of Agriculture, University of Miyazaki, 1-1
Gakuen Kibanadai-nishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Yuko MIYAMOTO
- Graduate School of Agriculture, University of Miyazaki, 1-1
Gakuen Kibanadai-nishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Kenjirou OGAWA
- Organization for Promotion of Tenure Track, University of
Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Kazuo NISHIYAMA
- Graduate School of Agriculture, University of Miyazaki, 1-1
Gakuen Kibanadai-nishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Chuluunbat TSEND-AYUSH
- School of Industrial Technology, Mongolian University of
Science and Technology, P.O. Box-46/520, Baga Toiruu, Sukhbaatar district, Ulaanbaatar-46,
Mongolia
| | - Yiran LI
- Research and Development Division, Minami Nihon Rakuno Kyodo
Co., Ltd., 5282 Takagi, Miyakonojo, Miyazaki 885-0003, Japan
| | - Tatsuya MATSUSAKI
- Research and Development Division, Minami Nihon Rakuno Kyodo
Co., Ltd., 5282 Takagi, Miyakonojo, Miyazaki 885-0003, Japan
| | - Tomoki NAKANO
- Research and Development Division, Minami Nihon Rakuno Kyodo
Co., Ltd., 5282 Takagi, Miyakonojo, Miyazaki 885-0003, Japan
| | - Masahiko TAKESHITA
- Research and Development Division, Minami Nihon Rakuno Kyodo
Co., Ltd., 5282 Takagi, Miyakonojo, Miyazaki 885-0003, Japan
| | - Yuo ARIMA
- Research and Development Division, Minami Nihon Rakuno Kyodo
Co., Ltd., 5282 Takagi, Miyakonojo, Miyazaki 885-0003, Japan
| |
Collapse
|
50
|
Martoni CJ, Srivastava S, Damholt A, Leyer GJ. Efficacy and dose response of Lactiplantibacillus plantarum in diarrhea-predominant irritable bowel syndrome. World J Gastroenterol 2023; 29:4451-4465. [PMID: 37576702 PMCID: PMC10415969 DOI: 10.3748/wjg.v29.i28.4451] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Probiotics have shown promise in alleviating symptoms of diarrhea-predominant irritable bowel syndrome (IBS-D); however, the certainty of evidence is low. Well-powered randomized controlled dose-ranging trials are warranted on promising single-strain candidates. AIM To investigate the clinical efficacy of Lactiplantibacillus plantarum (L. plantarum) Lpla33 (DSM34428) in adults with IBS-D. METHODS This is a randomized, double-blind, placebo-controlled, multi-center, and dose-ranging study. Three hundred and seven adults, 18-70 years of age, with IBS-D, according to Rome IV criteria, were allocated (1:1:1) to receive placebo or L. plantarum Lpla33 at 1 × 109 (1B) or 1 × 1010 (10B) colony-forming units/d over an 8-wk intervention period. The primary outcome was the change in IBS severity scoring system (IBS-SSS) total score after 8 wk, while secondary and exploratory outcomes included abdominal pain severity, IBS related quality of life, stool and microbial profile, and perceived stress. RESULTS IBS-SSS was significantly reduced, after 8 wk, in participants receiving L. plantarum 1B (-128.45 ± 83.30; P < 0.001) and L. plantarum 10B (-156.77 ± 99.06; P < 0.001), compared to placebo (-58.82 ± 74.75). Further, a dose-ranging effect was observed, with a greater absolute reduction in the L. plantarum 10B group (P < 0.05). A reduction in sub-scores related to abdominal pain, abdominal distension, bowel habits, and quality of life was observed in both L. plantarum groups compared to placebo (P < 0.001). Further, 62.5% and 88.4% of participants administered L. plantarum 1B and 10B, respectively, were classified as stool consistency responders based on a reduction in diarrheal stool form, as compared to 26.3% in the placebo group (P < 0.001). In contrast, no significant shifts were observed in microbial diversity. CONCLUSION L. plantarum Lpla33 (DSM34428) is well tolerated and improves IBS symptom severity with a dose-ranging effect and a corresponding normalization of bowel habits in adults with IBS-D.
Collapse
Affiliation(s)
| | | | - Anders Damholt
- Clinical Development, Human Health, Chr. Hansen A/S, Hoersholm 2970, Denmark
| | - Gregory J Leyer
- Scientific Affairs,Human Health, Chr. Hansen A/S, Hoersholm 2970, Denmark
| |
Collapse
|