1
|
Antoine D, Tao J, Singh S, Singh PK, Marin BG, Roy S. Neonatal exposure to morphine results in prolonged pain hypersensitivity during adolescence, driven by gut microbial dysbiosis and gut-brain axis-mediated inflammation. Brain Behav Immun 2025; 126:3-23. [PMID: 39900146 DOI: 10.1016/j.bbi.2025.01.021] [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: 10/18/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025] Open
Abstract
Opioids, such as morphine, are used in the Neonatal Intensive Care Unit (NICU) for pain relief in neonates. However, the available evidence concerning the benefits and harms of opioid therapy in neonates remains limited. While previous studies have reported that neonatal morphine exposure (NME) results in long-term heightened pain sensitivity, the underlying mechanisms are not well understood. This study proposes that dysbiosis of the gut microbiome contributes to pain hypersensitivity following NME. Using an adolescent female murine model, pain sensitivity was evaluated using the tail flick and hot plate assays for thermal pain and the Von Frey assay for mechanical pain. Gut microbiome composition was assessed using 16S rRNA sequencing, while transcriptomic changes in midbrain samples were investigated using bulk RNA sequencing. NME induced prolonged hypersensitivity to thermal and mechanical pain in adolescence, accompanied by persistent gut microbial dysbiosis and sustained systemic inflammation, characterized by elevated circulating cytokine levels (e.g., IL-1α, IL-12p70, IFN-γ, IL-10). Transplantation of the microbiome from NME adolescents recapitulated pain hypersensitivity in naïve adolescent mice, while neonatal probiotic intervention with Bifidobacterium infantis (B. infantis) reversed the pain hypersensitivity by preventing gut dysbiosis and associated systemic inflammation. Furthermore, transcriptomic analysis of midbrain tissues revealed that NME upregulated several genes and key signaling pathways, including those related to immune activation and excitatory signaling, which were notably mitigated with neonatal B. infantis administration. Together, these findings highlight the critical role of the gut-brain axis in modulating pain sensitivity and suggest that targeting the gut microbiome offers a promising therapeutic strategy for managing neurobiological disorders following early opioid exposure.
Collapse
Affiliation(s)
- Danielle Antoine
- Department of Surgery, University of Miami Miller School of Medicine Miami FL USA; Department of Neuroscience, University of Miami Miller School of Medicine Miami FL USA
| | - Junyi Tao
- Department of Surgery, University of Miami Miller School of Medicine Miami FL USA
| | - Salma Singh
- Department of Surgery, University of Miami Miller School of Medicine Miami FL USA
| | - Praveen Kumar Singh
- Department of Surgery, University of Miami Miller School of Medicine Miami FL USA
| | - Barbara G Marin
- Department of Surgery, University of Miami Miller School of Medicine Miami FL USA; Department of Neuroscience, University of Miami Miller School of Medicine Miami FL USA
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine Miami FL USA.
| |
Collapse
|
2
|
Liang Y, Han Y, Xiao L, Su Y, Bao T, Ji X, Jia L, Zhang J. Coenzyme Q10 modulates the immunity by enhancing mononuclear macrophage, NK cell activity, and regulating gut microbiota. Front Nutr 2025; 12:1504831. [PMID: 40165818 PMCID: PMC11955478 DOI: 10.3389/fnut.2025.1504831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Coenzyme Q10 (CoQ10), an important fat-soluble, bioactive molecule that predominantly found in the inner mitochondrial membrane, is widely used in functional food and health food raw materials, which has garnered considerable attention due to its potential role in immunoregulation. However, the intrinsic mechanism of CoQ10 on immunity, and the relationship to the gut microbiota have not been elucidated. Methods Here, we conducted a series of in vivo experiments with the aim of comprehensively exploring the effect of CoQ10 on both cellular and humoral immune functions, and on gut microbiota communities in mice. Results CoQ10 showed negligible impact on both mouse body weight fluctuations and tissue indices, but enhanced the mouse body immunity by elevating the carbon clearance ability and natural killer (NK) cellular viability. 16S rRNA gene sequencing revealed that administration of CoQ10 modulated the structure and composition of the gut microbiota in mice, notably by enhancing the abundance of Lactobacillus, Limosilactobacillus, and decreasing the abundance of Paramuribaculum species. Discussion This work makes a contribution to the application of CoQ10 as an immunomodulator in the biological, pharmaceutical and health care product industries.
Collapse
Affiliation(s)
- Yajun Liang
- College of Pharmacy, Qilu Medical University, Zibo, China
| | - Yang Han
- College of Pharmacy, Qilu Medical University, Zibo, China
| | - Ling Xiao
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Yupeng Su
- College of Pharmacy, Qilu Medical University, Zibo, China
| | - Tongen Bao
- College of Pharmacy, Qilu Medical University, Zibo, China
| | - Xia Ji
- College of Pharmacy, Qilu Medical University, Zibo, China
| | - Longgang Jia
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Jing Zhang
- College of Pharmacy, Qilu Medical University, Zibo, China
| |
Collapse
|
3
|
Xu FH, Sun X, Zhu J, Kong LY, Chang Y, Li N, Hui WX, Zhang CP, Cheng YM, Han WX, Tian ZM, Qiao YN, Chen DF, Liu L, Feng DY, Han J. Significance of the gut tract in the therapeutic mechanisms of polydopamine for acute cerebral infarction: neuro-immune interaction through the gut-brain axis. Front Cell Infect Microbiol 2025; 14:1413018. [PMID: 40104260 PMCID: PMC11913817 DOI: 10.3389/fcimb.2024.1413018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/25/2024] [Indexed: 03/20/2025] Open
Abstract
Background Recent research has made significant progress in elucidating gastrointestinal complications following acute cerebral infarction (ACI), which includes disorders in intestinal motility and dysbiosis of the gut microbiota. Nevertheless, the role of the gut (which is acknowledged as being the largest immune organ) in the immunoreactive effects of polydopamine nanoparticles (PDA) on acute ischemic stroke remains inadequately understood. In addition to its function in nutrient absorption, the gut acts as a protective barrier against microbes. Systemic immune responses, which are triggered by the disruption of gut barrier integrity, are considered as one of the mechanisms underlying acute ischemic stroke, with the gut-brain axis (GBA) playing a pivotal role in this process. Methods In this study, we used a PDA intervention in an ACI model to investigate ACI-like behavior, intestinal barrier function, central and peripheral inflammation, and hippocampal neuron excitability, thus aiming to elucidate the mechanisms through which PDA improves ACI via the GBA. Results Our findings indicated that as ACI mice experienced dysbiosis of the gut microbiota and intestinal barrier damage, the levels of proinflammatory factors in the serum and brain significantly increased. Additionally, the activation of astrocytes in the hippocampal region and neuronal apoptosis were observed in ACI mice. Importantly, our study is the first to provide evidence demonstrating that PDA effectively suppresses the neuroimmune interactions of the gut-brain axis and significantly improves intestinal epithelial barrier integrity. Conclusion We hope that our discoveries will serve as a foundation for further explorations of the therapeutic mechanisms of PDA in ACI, particularly in elucidating the protective roles of gut microbiota and intestinal barrier function, as well as in the development of more targeted clinical interventions for ACI.
Collapse
Affiliation(s)
- Feng-Hua Xu
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
| | - Xiao Sun
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Jun Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
| | - Ling-Yang Kong
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Yuan Chang
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Ning Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wen-Xiang Hui
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Cong-Peng Zhang
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Yi-Ming Cheng
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
- College of life sciences, Shaanxi Normal University, Xi’an, China
| | - Wen-Xin Han
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Zhi-Min Tian
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Yan-Ning Qiao
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| | - Dong-feng Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lei Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Da-Yun Feng
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
4
|
Zhong X, Wang X, Xu L, Zhang J, Yu W, Ji L, Huang J, Zhong X, Zhang J, Long L. Alterations in gut microbiota in Rheumatoid arthritis patients with interstitial lung Disease: A Comparative study. Hum Immunol 2025; 86:111239. [PMID: 39983663 DOI: 10.1016/j.humimm.2025.111239] [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: 08/07/2024] [Revised: 11/06/2024] [Accepted: 01/11/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is considered one of the most severe complications of rheumatoid arthritis. The etiology of RA-ILD is complex, involving genetic and environmental factors. Recent studies suggest that the gut microbiota, a critical component of the immune system, may influence the pathogenesis of RA and other autoimmune disorders. However, specific data on the gut microbiota in patients with RA-ILD remain limited. OBJECTIVE This study aimed to investigate alterations in the gut microbiota of RA-ILD patients and compare these profiles with those of RA patients without ILD and health controls. METHODS We included three groups: RA-ILD patients (n = 30), RA patients without ILD (n = 31), and health controls (n = 30). Fresh fecal samples were collected and subjected to 16S rRNA gene sequencing to analyze microbial diversity. Statistical analyses involved α-diversity and β-diversity assessments, principal coordinates analysis (PCoA), and differential abundance testing with LEfSe and PICRUSt2. RESULTS Significant differences in gut microbiota composition were observed between RA-ILD patients and the other groups. Notably, g_Prevotella showed differential abundance, particularly in RA-ILD patients. KEGG pathway analysis revealed upregulation in several metabolic pathways in RA-ILD compared to RA and health controls, suggesting a distinct microbial metabolic activity associated with RA-ILD. CONCLUSION Our findings indicate that RA-ILD patients have a markedly different gut microbiota profile compared to RA patients without ILD and health controls. The observed microbial alterations may contribute to RA-ILD pathogenesis and could serve as potential biomarkers or therapeutic targets. Further studies are needed to explore these findings' clinical implications and validate the role of gut microbiota in RA-ILD progression.
Collapse
Affiliation(s)
- Xue Zhong
- Department of Gerontology, Chongqing General Hospital, Chongqing University, PR China.
| | - Xiaohong Wang
- Department of Gerontology, Chongqing General Hospital, Chongqing University, PR China
| | - Lulu Xu
- Department of Gerontology, Chongqing General Hospital, Chongqing University, PR China
| | - Jie Zhang
- Department of Gerontology, Chongqing General Hospital, Chongqing University, PR China
| | - Wei Yu
- Department of Nephrology, Chongqing General Hospital, Chongqing University, PR China
| | - Lei Ji
- Department of Nephrology, Chongqing General Hospital, Chongqing University, PR China
| | - Jing Huang
- Department of Respiratory Medicine, Chongqing General Hospital, Chongqing University, PR China
| | - Xun Zhong
- Department of Medical Affairs Office, Chongqing General Hospital, Chongqing University, PR China
| | - Jie Zhang
- Department of Rheumatology and Immunology, Deyang people's hospital, Deyang, PR China
| | - Li Long
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China.
| |
Collapse
|
5
|
Li R, Ding X, Lei M, Li P, Giannenas I, Wang J, Zhu W. The impact of combined thymol and rosmarinic acid on the intestinal microbiota and barrier function of the piglets challenged by Escherichia coli K88. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:131-144. [PMID: 39967693 PMCID: PMC11834115 DOI: 10.1016/j.aninu.2024.11.008] [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: 10/09/2023] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 02/20/2025]
Abstract
It has been found that thymol (Thy) and rosmarinic acid (Ros-A) improve the growth performance of piglets and relieve intestinal inflammation in animals. The effects of Thy and Ros-A separately or in combination (Thy × Ros-A) on the intestinal function and health of piglets challenged with Escherichia coli K88 (E. coli K88) were investigated. A total of 30 piglets aged 21 d were assigned to 5 groups (n = 6). The control (Con) and K88 groups piglets received a basal diet, while the Thy, Ros-A, and Thy × Ros-A groups were fed a basal diet supplemented with 500 mg/kg Thy, 500 mg/kg Ros-A, and 250 mg/kg Thy + 250 mg/kg Ros-A, respectively. On the 19th and 20th day, piglets in the K88, Thy, Ros-A, and Thy × Ros-A groups were orally administered 10 mL of phosphate-buffered saline (PBS) containing approximately 1 × 109 CFU/mL of E. coli K88, while the Con group received an equal volume of PBS. The results showed that the Thy × Ros-A treatment reduced the damage to ileal villi induced by the E. coli K88 challenge, leading to longer villi in the ileum (P < 0.05). Thy and Ros-A modulated the composition of the ileal microbiota. Compared to the K88 group, the Thy × Ros-A group had a higher abundance of Lactobacillus and Romboutsia, while Escherichia-Shigella and Desulforvibrio were lower (P < 0.05). Additionally, the Thy × Ros-A group showed elevated levels of gene and protein expressions for zonula occludens-1, occludin, and claudin-1 compared to the K88 group (P < 0.05). In conclusion, combining Thy and Ros-A reduced ileal damage and relieved the inflammation in weaned piglets challenged with E. coli K88 by regulating intestinal microflora and improving barrier function.
Collapse
Affiliation(s)
- Runlin Li
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuedong Ding
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingkang Lei
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Panpan Li
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ilias Giannenas
- Aristotle University of Thessaloniki, University Campus, Thessaloniki 54124, Greece
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
6
|
Zhang Q, Zhao W, Luo J, Shi S, Niu X, He J, Wang Y, Zeng Z, Jiang Q, Fang B, Chen J, Li Y, Wang F, He J, Guo J, Zhang M, Zhang L, Ge S, Hung WL, Wang R. Synergistic defecation effects of Bifidobacterium animalis subsp. lactis BL-99 and fructooligosaccharide by modulating gut microbiota. Front Immunol 2025; 15:1520296. [PMID: 39850898 PMCID: PMC11754280 DOI: 10.3389/fimmu.2024.1520296] [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: 10/31/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction Synbiotics have revealed the possibility of improving constipation through gut microbiota. The synergistic efficacy of Bifidobacterium animalis subsp. lactis BL-99 (BL-99) and fructooligosaccharide (FOS) on constipation have not been investigated. Methods Loperamide-induced constipated mice model was established to explore the effect of BL-99, FOS, and BL-99+FOS on changes of defecation-related parameters, gut microbiota and metabolites. Results and discussion The results showed that BL-99, FOS, and BL-99+FOS each alleviated constipation, with the synbiotic showing significant efficacy in the first black stool defecation time, fecal number, fecal weight, and the gastrointestinal transit rate (P < 0.05). Additionally, significant increased in serum 5-HT and IL-10 were observed in the BL-99+FOS group, alongside an increased relative abundance of Lachnospiraceae_NK4A136_group, Blautia, and Clostridium sensu stricto 1, while significantly reducing the relative abundance of Alistipes and Bacteroides. These changes facilitated alterations in short-chain fatty acids (SCFAs) metabolism, and were closely associated with the expression of genes related to the 5-HT pathway and the modulation of serum inflammatory factors. This study provides a theoretical basis for BL-99 and FOS synbiotics to improve constipation by regulating the gut microbiota and metabolites.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Wen Zhao
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jie Luo
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Shaoqi Shi
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xiaokang Niu
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jian He
- Probiotics R&D Department, Inner Mongolia National Center of Technology Innovation for Dairy Co. Ltd., Hohhot, China
| | - Yicheng Wang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Zhaozhong Zeng
- Probiotics R&D Department, Inner Mongolia National Center of Technology Innovation for Dairy Co. Ltd., Hohhot, China
| | - Qiuyue Jiang
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co. Ltd., Hohhot, China
| | - Bing Fang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Juan Chen
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fuqing Wang
- Department of Food Science, Tibet Tianhong Science and Technology Co., Ltd., Lhasa, China
| | - Jingjing He
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jie Guo
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ming Zhang
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China
| | - Liwei Zhang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Shaoyang Ge
- Probiotics R&D Department, Hebei Engineering Research Center of Animal Product, Sanhe, China
| | - Wei-Lian Hung
- Probiotics R&D Department, Inner Mongolia National Center of Technology Innovation for Dairy Co. Ltd., Hohhot, China
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
da Silva DR, Sharjeel AB, Beliakoff R, Teixeira LD, Kima PE, Jones MK, Gonzalez CF, Lorca GL. The Sdp-SH3b2 domain contained in Lactobacillus johnsonii N6.2-derived extracellular vesicles inhibit murine norovirus replication. Front Immunol 2024; 15:1490755. [PMID: 39712028 PMCID: PMC11659762 DOI: 10.3389/fimmu.2024.1490755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
The internalization of Lactobacillus johnsonii N6.2 extracellular vesicles (EVs) by cells results in a significant induction of the 2',5'-oligoadenylate synthetase (OAS) pathway. It also induces expression of IFI44L, MX1, MX2 and DDX60. In this work, we evaluated whether the antiviral response induced by L. johnsonii N6.2-derived EVs, has an inhibitory effect on an RNA viral insult using murine norovirus (MNV-1) as the viral infection model. We found that RAW 264.7 Macrophages treated with EVs significantly decreased the levels of MNV-1 genome. These results were consistent with an increase in expression of Oas1b, Oas2, Oasl, Mx1, Mx2 and Ifi44l (6 hours post infection). Out of six proteins enriched in EVs, we found that SH3b2 domain of Sdp was the only protein effector molecule able to recapitulate the activation of the OAS pathway. In C57BL6 mice, the administration of live L. johnsonii N6.2, EVs, and Sdp-SH3b2/liposomes significantly decreased MNV-1 titers in the distal ileum, in contrast to the controls with PBS and liposomes alone that did not affect MNV-1. These results establish that the SH3b2 domain of Sdp, which is enriched in L. johnsonii derived EVs, is an effector molecule in EVs that can orchestrate the control of viral infections in vivo.
Collapse
Affiliation(s)
- Danilo R. da Silva
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Asra B. Sharjeel
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Reagan Beliakoff
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Leandro D. Teixeira
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Peter E. Kima
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Melissa K. Jones
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Claudio F. Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Graciela L. Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
8
|
Chen X, Mo X, Zhang Y, He D, Xiao R, Cheng Q, Wang H, Liu L, Li WW, Xie P. A comprehensive analysis of the differential expression in the hippocampus of depression induced by gut microbiota compared to traditional stress. Gene 2024; 927:148633. [PMID: 38838871 DOI: 10.1016/j.gene.2024.148633] [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: 02/17/2024] [Revised: 05/22/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Depression, which is a disease of heterogeneous etiology, is characterized by high disability and mortality rates. Gut microbiota are associated with the development of depression. To further explore any differences in the mechanisms of depression induced by gut microbiota and traditional stresses, as well as facilitate the development of microbiota-based interventions, a fecal microbiota transplantation (FMT) depression model was made. This was achieved by transplanting feces from major depressive disorder (MDD) patients into germ-free mice. Second, the mechanisms of the depression induced by gut microbiota were analyzed in comparison with those of the depression caused by different forms of stress. It turned out that mice exhibited depressive-like behavior after FMT. Then, PCR array analysis was performed on the hippocampus of the depressed mice to identify differentially expressed genes (DEGs). The KEGG analysis revealed that the pathways of depression induced by gut microbes are closely associated with immuno-inflammation. To determine the pathogenic pathways of physiological stress and psychological stress-induced depression, raw data was extracted from several databases and KEGG analysis was performed. The results from the analysis revealed that the mechanisms of depression induced by physiological and psychological stress are closely related to the regulation of neurotransmitters and energy metabolism. Interestingly, the immunoinflammatory response was distinct across different etiologies that induced depression. The findings showed that gut microbiota dysbiosis-induced depression was mainly associated with adaptive immunity, while physiological stress-induced depression was more linked to innate immunity. This study compared the pathogenesis of depression caused by gut microbiota dysbiosis, and physiological and psychological stress. We explored new intervention methods for depression and laid the foundation for precise treatment.
Collapse
Affiliation(s)
- Xueyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolong Mo
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yangdong Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dian He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Xiao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Qisheng Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haiyang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lanxiang Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Wen-Wen Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
9
|
Huang Q, Xing J, Tang F, Ren J, Wang C, Xue F. Recombinant Lactiplantibacilllus plantarum modulate gut microbial diversity and function. BMC Microbiol 2024; 24:423. [PMID: 39438791 PMCID: PMC11494753 DOI: 10.1186/s12866-024-03570-4] [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: 10/17/2023] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gut microbes are important regulators of host health and can also function as disease indicators. Lactiplantibacilllus plantarum(L. plantarum)used as express and delivery vaccines for mucosal immunity have been shown to activate specific immune responses in numerous studies. RESULTS The interaction between recombinant L. plantarum and the gut microbiota was investigated in this study. The results indicated a change in the amount of gut OTU by recombinant L. plantarum. Recombinant L. plantarum dramatically boosted the species diversity of gut bacteria based on the Shannon-Wiener index. Beta diversity analysis showed that microbial structure was changed by recombinant L. plantarum. Furthermore, recombinant NC8 L. plantarum expressing a fusion between the P14.5 protein of the African swine fever virus and IL-33 enhanced the functions of gut bacteria in metabolism and immune regulation. Increased levels of IgG and IgG1 in serum and sIgA in feces, as well as enrichment of CD4+ T cells and IgA+ B cells, indicated that the gut microbiota exerted an immunomodulatory role when mediated by recombinant L. plantarum. CONCLUSIONS These results revealed that recombinant L. plantarum exerted its potential role in the gut microbiota and gut immunity.These fndings contribute to a broader understanding and utilization of L. plantarum bacteria in various therapeutic applications.
Collapse
Affiliation(s)
- Quntao Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Junhong Xing
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jianluan Ren
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chunfeng Wang
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
10
|
Heidari M, Maleki Vareki S, Yaghobi R, Karimi MH. Microbiota activation and regulation of adaptive immunity. Front Immunol 2024; 15:1429436. [PMID: 39445008 PMCID: PMC11496076 DOI: 10.3389/fimmu.2024.1429436] [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: 05/08/2024] [Accepted: 08/30/2024] [Indexed: 10/25/2024] Open
Abstract
In the mucosa, T cells and B cells of the immune system are essential for maintaining immune homeostasis by suppressing reactions to harmless antigens and upholding the integrity of intestinal mucosal barrier functions. Host immunity and homeostasis are regulated by metabolites produced by the gut microbiota, which has developed through the long-term coevolution of the host and the gut biome. This is achieved by the immunological system's tolerance for symbiote microbiota, and its ability to generate a proinflammatory response against invasive organisms. The imbalance of the intestinal immune system with commensal organisms is causing a disturbance in the homeostasis of the gut microbiome. The lack of balance results in microbiota dysbiosis, the weakened integrity of the gut barrier, and the development of inflammatory immune reactions toward symbiotic organisms. Researchers may uncover potential therapeutic targets for preventing or regulating inflammatory diseases by understanding the interactions between adaptive immunity and the microbiota. This discussion will explore the connection between adaptive immunity and microbiota.
Collapse
Affiliation(s)
- Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Maleki Vareki
- Department of Oncology, Western University, London, ON, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Ramin Yaghobi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
11
|
Morovati S, Baghkheirati AA, Sekhavati MH, Razmyar J. A Review on cLF36, a Novel Recombinant Antimicrobial Peptide-Derived Camel Lactoferrin. Probiotics Antimicrob Proteins 2024; 16:1886-1905. [PMID: 38722550 DOI: 10.1007/s12602-024-10285-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 10/02/2024]
Abstract
Lactoferrin is an antimicrobial peptide (AMP) playing a pivotal role in numerous biological processes. The primary antimicrobial efficacy of lactoferrin is associated with its N-terminal end, which contains various peptides, such as lactoferricin and lactoferrampin. In this context, our research team has developed a refined chimeric 42-mer peptide known as cLF36 over the past few years. This peptide encompasses the complete amino acid sequence of camel lactoferrampin and partial amino acid sequence of lactoferricin. The peptide's activity against human, avian, and plant bacterial pathogens has been assessed using different biological platforms, including prokaryotic (P170 and pET) and eukaryotic (HEK293) expression systems. The peptide positively influenced the growth performance and intestinal morphology of chickens challenged with pathogen bacteria. Computational methods and in vitro studies showed the peptide's antiviral effects against hepatitis C virus, influenza virus, and rotavirus. The chimeric peptide exhibited higher activity against certain tumor cell lines compared to normal cells, which may be attributed to the peptide's interaction with negatively charged glycosaminoglycans on the surface of tumor cells. Importantly, this peptide exhibited no toxicity against host cells and demonstrated remarkable thermal and protease stability in serum. In conclusion, while our investigations suggest that the chimeric peptide, cLF36, may offer potential as a candidate or complementary option to some available antibiotics, antiviral agents, and chemical pesticides, significant uncertainties remain regarding its cost-effectiveness, as well as its pharmacodynamic and pharmacokinetic characteristics, which require further elucidation.
Collapse
Affiliation(s)
- Solmaz Morovati
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | | | - Mohammad Hadi Sekhavati
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Jamshid Razmyar
- Department of Avian Diseases, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
12
|
Fellows RC, Chun SK, Larson N, Fortin BM, Mahieu AL, Song WA, Seldin MM, Pannunzio NR, Masri S. Disruption of the intestinal clock drives dysbiosis and impaired barrier function in colorectal cancer. SCIENCE ADVANCES 2024; 10:eado1458. [PMID: 39331712 PMCID: PMC11430476 DOI: 10.1126/sciadv.ado1458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/22/2024] [Indexed: 09/29/2024]
Abstract
Diet is a robust entrainment cue that regulates diurnal rhythms of the gut microbiome. We and others have shown that disruption of the circadian clock drives the progression of colorectal cancer (CRC). While certain bacterial species have been suggested to play driver roles in CRC, it is unknown whether the intestinal clock impinges on the microbiome to accelerate CRC pathogenesis. To address this, genetic disruption of the circadian clock, in an Apc-driven mouse model of CRC, was used to define the impact on the gut microbiome. When clock disruption is combined with CRC, metagenomic sequencing identified dysregulation of many bacterial genera including Bacteroides, Helicobacter, and Megasphaera. We identify functional changes to microbial pathways including dysregulated nucleic acid, amino acid, and carbohydrate metabolism, as well as disruption of intestinal barrier function. Our findings suggest that clock disruption impinges on microbiota composition and intestinal permeability that may contribute to CRC pathogenesis.
Collapse
Affiliation(s)
- Rachel C. Fellows
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Sung Kook Chun
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Natalie Larson
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Bridget M. Fortin
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Alisa L. Mahieu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Wei A. Song
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Marcus M. Seldin
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Nicholas R. Pannunzio
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, 92697, USA
- Department of Medicine, Division of Hematology/Oncology, University of California Irvine, Irvine, CA 92697, USA
| | - Selma Masri
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Caspani G, Ruffell SGD, Tsang W, Netzband N, Rohani-Shukla C, Swann JR, Jefferies WA. Mind over matter: the microbial mindscapes of psychedelics and the gut-brain axis. Pharmacol Res 2024; 207:107338. [PMID: 39111558 DOI: 10.1016/j.phrs.2024.107338] [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: 07/16/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Psychedelics have emerged as promising therapeutics for several psychiatric disorders. Hypotheses around their mechanisms have revolved around their partial agonism at the serotonin 2 A receptor, leading to enhanced neuroplasticity and brain connectivity changes that underlie positive mindset shifts. However, these accounts fail to recognise that the gut microbiota, acting via the gut-brain axis, may also have a role in mediating the positive effects of psychedelics on behaviour. In this review, we present existing evidence that the composition of the gut microbiota may be responsive to psychedelic drugs, and in turn, that the effect of psychedelics could be modulated by microbial metabolism. We discuss various alternative mechanistic models and emphasize the importance of incorporating hypotheses that address the contributions of the microbiome in future research. Awareness of the microbial contribution to psychedelic action has the potential to significantly shape clinical practice, for example, by allowing personalised psychedelic therapies based on the heterogeneity of the gut microbiota.
Collapse
Affiliation(s)
- Giorgia Caspani
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, East Mall, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Department of Urologic Sciences, University of British Columbia, Gordon & Leslie Diamond Health Care Centre, Level 6, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada.
| | - Simon G D Ruffell
- Psychae Institute, Melbourne, Australia; School of Population and Global Health, University of Melbourne, 207 Bouverie St, Carlton, VIC 3053, Australia
| | - WaiFung Tsang
- Institute of Psychiatry, Psychology & Neuroscience, King'sCollege London, Department of Psychology, De Crespigny Park, London SE5 8AF, UK
| | - Nigel Netzband
- University of West of England, Frenchay Campus, Coldharbour Lane, Bristol BS16 1QY, UK
| | - Cyrus Rohani-Shukla
- Centre for Psychedelic Research, Imperial College London, Hammersmith Hospital, Du Cane Rd, London W12 0HS, UK
| | - Jonathan R Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, 12 University Rd, Southampton SO17 1BJ, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, East Mall, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Department of Urologic Sciences, University of British Columbia, Gordon & Leslie Diamond Health Care Centre, Level 6, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
14
|
Gormley A, Garavito-Duarte Y, Kim SW. The Role of Milk Oligosaccharides in Enhancing Intestinal Microbiota, Intestinal Integrity, and Immune Function in Pigs: A Comparative Review. BIOLOGY 2024; 13:663. [PMID: 39336091 PMCID: PMC11428639 DOI: 10.3390/biology13090663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
The objective of this review was to identify the characteristics and functional roles of milk coproducts from human, bovine, and porcine sources and their impacts on the intestinal microbiota and intestinal immunity of suckling and nursery pigs. Modern pig production weans piglets at 3 to 4 weeks of age, which is earlier than pigs would naturally be weaned outside of artificial rearing. As a result, the immature intestines of suckling and nursery pigs face many challenges associated with intestinal dysbiosis, which can be caused by weaning stress or the colonization of the intestines by enteric pathogens. Milk oligosaccharides are found in sow milk and function as a prebiotic in the intestines of pigs as they cannot be degraded by mammalian enzymes and are thus utilized by intestinal microbial populations. The consumption of milk oligosaccharides during suckling and through the nursery phase can provide benefits to young pigs by encouraging the proliferation of beneficial microbial populations, preventing pathogen adhesion to enterocytes, and through directly modulating immune responses. Therefore, this review aims to summarize the specific functional components of milk oligosaccharides from human, bovine, and porcine sources, and identify potential strategies to utilize milk oligosaccharides to benefit young pigs through the suckling and nursery periods.
Collapse
Affiliation(s)
| | | | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (A.G.); (Y.G.-D.)
| |
Collapse
|
15
|
Raajaraam L, Raman K. Modeling Microbial Communities: Perspective and Challenges. ACS Synth Biol 2024; 13:2260-2270. [PMID: 39148432 DOI: 10.1021/acssynbio.4c00116] [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] [Indexed: 08/17/2024]
Abstract
Microbial communities are immensely important due to their widespread presence and profound impact on various facets of life. Understanding these complex systems necessitates mathematical modeling, a powerful tool for simulating and predicting microbial community behavior. This review offers a critical analysis of metabolic modeling and highlights key areas that would greatly benefit from broader discussion and collaboration. Moreover, we explore the challenges and opportunities linked to the intricate nature of these communities, spanning data generation, modeling, and validation. We are confident that ongoing advancements in modeling techniques, such as machine learning, coupled with interdisciplinary collaborations, will unlock the full potential of microbial communities across diverse applications.
Collapse
Affiliation(s)
- Lavanya Raajaraam
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology (IIT) Madras, Chennai 600 036, India
- Centre for Integrative Biology and Systems mEdicine, IIT Madras, Chennai 600 036, India
- Robert Bosch Centre for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai 600 036, India
| | - Karthik Raman
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology (IIT) Madras, Chennai 600 036, India
- Centre for Integrative Biology and Systems mEdicine, IIT Madras, Chennai 600 036, India
- Robert Bosch Centre for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai 600 036, India
- Department of Data Science and AI, Wadhwani School of Data Science and Artificial Intelligence, IIT Madras, Chennai 600 036, India
| |
Collapse
|
16
|
Li H, Liu H, Bi L, Liu Y, Jin L, Peng R. Immunotoxicity of microplastics in fish. FISH & SHELLFISH IMMUNOLOGY 2024; 150:109619. [PMID: 38735599 DOI: 10.1016/j.fsi.2024.109619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/17/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Plastic waste degrades slowly in aquatic environments, transforming into microplastics (MPs) and nanoplastics (NPs), which are subsequently ingested by fish and other aquatic organisms, causing both physical blockages and chemical toxicity. The fish immune system serves as a crucial defense against viruses and pollutants present in water. It is imperative to comprehend the detrimental effects of MPs on the fish immune system and conduct further research on immunological assessments. In this paper, the immune response and immunotoxicity of MPs and its combination with environmental pollutants on fish were reviewed. MPs not only inflict physical harm on the natural defense barriers like fish gills and vital immune organs such as the liver and intestinal tract but also penetrate cells, disrupting intracellular signaling pathways, altering the levels of immune cytokines and gene expression, perturbing immune homeostasis, and ultimately compromising specific immunity. Initially, fish exposed to MPs recruit a significant number of macrophages and T cells while activating lysosomes. Over time, this exposure leads to apoptosis of immune cells, a decline in lysosomal degradation capacity, lysosomal activity, and complement levels. MPs possess a small specific surface area and can efficiently bind with heavy metals, organic pollutants, and viruses, enhancing immune responses. Hence, there is a need for comprehensive studies on the shape, size, additives released from MPs, along with their immunotoxic effects and mechanisms in conjunction with other pollutants and viruses. These studies aim to solidify existing knowledge and delineate future research directions concerning the immunotoxicity of MPs on fish, which has implications for human health.
Collapse
Affiliation(s)
- Huiqi Li
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Huanpeng Liu
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Liuliu Bi
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yinai Liu
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Libo Jin
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Renyi Peng
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
17
|
Gong J, Xu F, Li Y, He Y, Liang Z, Chen X, Zhang X, Liu L, Zhou L, Huang X. Metagenomic analysis of intestinal microbial function and key genes responsive to acute high-salinity stress in Nile tilapia (Oreochromis niloticus). Gene 2024; 913:148371. [PMID: 38485034 DOI: 10.1016/j.gene.2024.148371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/26/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
The intestinal microbiota is increasingly recognized as playing an important role in aquatic animals. To investigate the functional roles and mechanisms of the intestinal microbial genes/enzymes responding to salinity stress or osmotic pressure in fish, metagenomic analysis was carried out to evaluate the response of intestinal microbiota and especially their functional genes/enzymes from freshwater (the control group) to acute high salinity stress (the treatment group) in Nile tilapia. Our results showed that at the microbial community level, the intestinal microbiota in Nile tilapia generally underwent significant changes in diversity after acute high salinity stress. Among them, the shift in the bacterial community (mainly from Actinobacteria to Proteobacteria) dominated and had a large impact, the fungal community showed a very limited response, and other microbiota, such as phages, likely had a negligible response. At the functional level, the intestinal bacteriadecreased the normal physiological demand and processes, such as those of the digestive system and nervous system, but enhanced energy metabolism. Furthermore, at the gene level, some gene biomarkers, such as glutathione S-transferase, myo-inositol-1(or 4)-monophosphatase, glycine betaine/proline transport system permease protein, and some families of carbohydrate-active enzymes (GT4, GT2), were significantly enriched. However, GH15, GH23 and so on were significantly reduced. Exploring the functional details of the intestinal microbial genes/enzymes that respond to salinity stress in Nile tilapia sheds light on the mechanism of action of the intestinal microbiota with respect to the salinity adaptation of fish.
Collapse
Affiliation(s)
- Jiayi Gong
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Fengmeng Xu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China; Guangzhou Fishtech Biotechnology Co., Ltd., Guangzhou 510640, China
| | - Yao Li
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Yiyong He
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Zhizheng Liang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiao Chen
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiaoyong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Li Liu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Lei Zhou
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China.
| | - Xiande Huang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
18
|
Kim H, Jeong S, Kim SW, Kim HJ, Kim DY, Yook TH, Yang G. Indigo Naturalis in Inflammatory Bowel Disease: mechanisms of action and insights from clinical trials. J Pharmacopuncture 2024; 27:59-69. [PMID: 38948310 PMCID: PMC11194518 DOI: 10.3831/kpi.2024.27.2.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/02/2024] [Accepted: 03/20/2024] [Indexed: 07/02/2024] Open
Abstract
This study investigates the therapeutic potential of Indigo Naturalis (IN) in treating a Inflammatory Bowel Disease (IBD). The objective is to comprehensively examine the effects and pharmacological mechanisms of IN on IBD, assessing its potential as an novel treatment for IBD. Analysis of 11 selected papers is conducted to understand the effects of IN, focusing on compounds like indirubin, isatin, indigo, and tryptanthrin. This study evaluates their impact on Disease Activity Index (DAI) score, colon length, mucosal damage, and macrophage infiltration in Dextran Sulfate Sodium (DSS)-induced colitis mice. Additionally, It investigate into the anti-inflammatory mechanisms, including Aryl hydrocarbon Receptor (AhR) pathway activation, Nuclear Factor kappa B (NF-κB)/nod-like receptor family pyrin domain containing 3 (NLRP3)/Interleukin 1 beta (IL-1β) inhibition, and modulation of Toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MYD88)/NF-κB and Mitogen Activated Protein Kinase (MAPK) pathways. Immunomodulatory effects on T helper 17 (Th17)/regulatory T cell (Treg cell) balance and Glycogen synthase kinase-3 beta (GSK3-β) expression are also explored. Furthermore, the study addresses the role of IN in restoring intestinal microbiota diversity, reducing pathogenic bacteria, and increasing beneficial bacteria. The findings reveal that IN, particularly indirubin and indigo, demonstrates significant improvements in DAI score, colon length, mucosal damage, and macrophage infiltration in DSS-induced colitis mice. The anti-inflammatory effects are attributed to the activation of the AhR pathway, inhibition of inflammatory pathways, and modulation of immune responses. These results exhibit the potential of IN in IBD treatment. Notably, the restoration of intestinal microbiota diversity and balance further supports its efficacy. IN emerges as a promising and effective treatment for IBD, demonstrating anti-inflammatory effects and positive outcomes in preclinical studies. However, potential side effects necessitate further investigation for safe therapeutic development. The study underscores the need for future research to explore a broader range of active ingredients in IN to enhance therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Hyeonjin Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Soohyun Jeong
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Sung Wook Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Hyung-Jin Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Dae Yong Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Tae Han Yook
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Gabsik Yang
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| |
Collapse
|
19
|
Huang YL, Zheng JM, Shi ZY, Chen HH, Wang XT, Kong FB. Inflammatory proteins may mediate the causal relationship between gut microbiota and inflammatory bowel disease: A mediation and multivariable Mendelian randomization study. Medicine (Baltimore) 2024; 103:e38551. [PMID: 38905376 PMCID: PMC11191895 DOI: 10.1097/md.0000000000038551] [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: 04/01/2024] [Accepted: 05/21/2024] [Indexed: 06/23/2024] Open
Abstract
This research investigates the causal relationships among gut microbiota, inflammatory proteins, and inflammatory bowel disease (IBD), including crohn disease (CD) and ulcerative colitis (UC), and identifies the role of inflammatory proteins as potential mediators. Our study analyzed gut microbiome data from 13,266 samples collected by the MiBioGen alliance, along with inflammatory protein data from recent research by Zhao et al, and genetic data on CD and UC from the International Inflammatory Bowel Disease Genetics Consortium (IIBDGC). We used Mendelian randomization (MR) to explore the associations, complemented by replication, meta-analysis, and multivariable MR techniques for enhanced accuracy and robustness. Our analysis employed several statistical methods, including inverse-variance weighting, MR-Egger, and the weighted median method, ensuring comprehensive and precise evaluation. After MR analysis, replication and meta-analysis, we revealed significant associations between 11 types of gut microbiota and 17 inflammatory proteins were associated with CD and UC. Mediator MR analysis and multivariable MR analysis showed that in CD, the CD40L receptor mediated the causal effect of Defluviitaleaceae UCG-011 on CD (mediation ratio 8.3%), and the Hepatocyte growth factor mediated the causal effect of Odoribacter on CD (mediation ratio 18%). In UC, the C-C motif chemokine 4 mediated the causal effect of Ruminococcus2 on UC (mediation ratio 4%). This research demonstrates the interactions between specific gut microbiota, inflammatory proteins, and CD and UC. Furthermore, the CD40L receptor may mediate the relationship between Defluviitaleaceae UCG-011 and CD; the Hepatocyte growth factor may mediate the relationship between Odoribacter and CD; and the C-C motif chemokine 4 may mediate the relationship between Ruminococcus2 and UC. The identified associations and mediation effects offer insights into potential therapeutic approaches targeting the gut microbiome for managing CD and UC.
Collapse
Affiliation(s)
- Yu-Liang Huang
- Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Jin-Min Zheng
- Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Zheng-Yi Shi
- Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Huan-Huan Chen
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xiao-Tong Wang
- Departments of Gastrointestinal, Hernia and Enterofistula Surgery, People’s Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Fan-Biao Kong
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
20
|
Lin K, Zheng W, Guo M, Zhou R, Zhang M, Liu T. The intestinal microbial metabolite acetyl l-carnitine improves gut inflammation and immune homeostasis via CADM2. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167089. [PMID: 38369215 DOI: 10.1016/j.bbadis.2024.167089] [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: 08/12/2023] [Revised: 01/27/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Intestinal symbiotic bacteria play a key role in the regulation of immune tolerance in inflammatory bowel disease (IBD) hosts. However, the bacterial strains directly involved in this regulation and their related metabolites are largely unknown. We sought to investigate the effects of intestinal microbial metabolites on intestinal epithelium and to elucidate their therapeutic potential in regulating intestinal mucosal inflammation and immune homeostasis. Here, we used metagenomic data from Crohn's disease (CD) patients to analyze the composition of intestinal flora and identify metabolite profiles associated with disease behavior, and used the mouse model of dextran sodium sulfate (DSS)-induced colitis to characterize the therapeutic effects of the flora metabolite acetyl l-carnitine (ALC) on DSS-induced colitis. We found that intraperitoneal injection of ALC treatment could significantly alleviate the symptoms of DSS-induced colitis in mice, including prevention of weight loss, reduction in disease activity index (DAI) scores, increasing of colonic length, reduction in histological scores, and improvement in intestinal barrier function. Further, transcriptome sequencing analysis and gene silencing experiments revealed that the absence of CADM2 abolished the inhibitory effect of ALC on the TLR-MyD88 pathway in colonic epithelial cells, thereby reducing the release of inflammatory factors in colon epithelial cells. And we confirmed a significant downregulation of CADM2 expression in intestinal tissues of CD patients compared to healthy people in a population cohort. In addition, we also found that ALC increased the ratio of Treg cells in colon, and decreased the ratio of Th17 cells and macrophages, thereby improving the immune tolerance of the organism. The proposed study could be a potential approach for the treatment of CD.
Collapse
Affiliation(s)
- Kai Lin
- Medical Research Center, Peking Union Medical College Hospital, Beijing, China.
| | - Weiyang Zheng
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Mingyue Guo
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Runing Zhou
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Mengmeng Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Tingting Liu
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
21
|
Zhao BC, Wang TH, Chen J, Qiu BH, Xu YR, Li JL. Essential oils improve nursery pigs' performance and appetite via modulation of intestinal health and microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:174-188. [PMID: 38357573 PMCID: PMC10864218 DOI: 10.1016/j.aninu.2023.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/22/2023] [Accepted: 10/12/2023] [Indexed: 02/16/2024]
Abstract
Optimal intestinal health and functionality are essential for animal health and performance, and simultaneously intestinal nutrient transporters and intestinal peptides are also involved in appetite and feed intake control mechanisms. Given the potential of essential oil (EO) in improving animal performance and improving feed palatability, we hypothesized that dietary supplementation of cinnamaldehyde and carvacrol could improve performance and appetite of nursery pigs by modulating intestinal health and microbiota. Cinnamaldehyde (100 mg/kg), carvacrol (100 mg/kg), and their mixtures (including 50 mg/kg cinnamaldehyde and 50 mg/kg carvacrol) were supplemented into the diets of 240 nursery pigs for 42 d, and data related to performance were measured. Thereafter, the influence of EO on intestinal health, appetite and gut microbiota and their correlations were explored. EO supplementation increased (P < 0.05) the body weight, average daily gain (ADG) and average daily feed intake (ADFI) of piglets, and reduced (P < 0.05) diarrhea rates in nursery pigs. Furthermore, EO increased (P < 0.05) the intestinal absorption area and the abundance of tight junction proteins, and decreased (P < 0.05) intestinal permeability and local inflammation. In terms of intestinal development and the mucus barrier, EO promoted intestinal development and increased (P < 0.05) the number of goblet cells. Additionally, we found that piglets in the EO-supplemented group had upregulated (P < 0.05) levels of transporters and digestive enzymes in the intestine, which were significantly associated with daily gain and feed utilization. In addition, EO supplementation somewhat improved appetite in nursery pigs, increased the diversity of the gut microbiome and the abundance of beneficial bacteria, and there was a correlation between altered bacterial structure and appetite-related hormones. These findings indicate that EO is effective in promoting growth performance and nutrient absorption as well as in regulating appetite by improving intestinal health and bacterial structure.
Collapse
Affiliation(s)
- Bi-Chen Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tian-Hao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jian Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bai-Hao Qiu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ya-Ru Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| |
Collapse
|
22
|
Lu W, Cen J, Dai Q, Tao H, Peng L. Gut microbiota does not play a mediating role in the causal association between inflammatory bowel disease and several its associated extraintestinal manifestations: a Mendelian randomization study. Front Immunol 2024; 14:1296889. [PMID: 38288127 PMCID: PMC10822939 DOI: 10.3389/fimmu.2023.1296889] [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: 09/19/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Background and objectives Observational study has found inflammatory bowel disease to be associated with multiple extraintestinal manifestations. To this end, we characterized the causal association between inflammatory bowel disease and extraintestinal manifestations through a Mendelian randomization study and further explored the role of intestinal flora in inflammatory bowel disease and the extraintestinal manifestations associated with it. Materials and methods We genetically predicted the causal relationship between inflammatory bowel disease and twenty IBD-related extraintestinal manifestations (including sarcoidosis, iridocyclitis, interstitial lung disease, atopic dermatitis, ankylosing spondylitis, psoriatic arthropathies, primary sclerosing cholangitis, primary biliary cholangitis). We used the full genome-wide association study (GWAS) summary statistics on gut microbiota in 18,340 participants from 24 cohorts to explore its role in the casual relationships between IBD and IBD-related extraintestinal manifestations. Inverse variance weighting (IVW) was used as the main analytical method to assess the causal associations. We performed Cochran's Q test to examine the heterogeneity. To assess the robustness of the IVW results, we further performed sensitivity analyses including the weighted median method, MR-Egger regression, and Mendelian Randomization Pleiotropy RESidual Sum and Outlier (MR-PRESSO) test. The leave-one-out sensitivity analysis was further performed to monitor if significant associations were dominated by a single nucleotide polymorphism (SNP). Result A total of eight extraintestinal manifestations were found to be at elevated risk of development due to inflammatory bowel diseases. A total of 11 causal relationships were found between IBD and gut microbiota, four of which were stable. Between gut microbiota and these eight extraintestinal manifestations, a total of 67 nominal causal associations were identified, of which 13 associations were stable, and notably 4 associations were strongly correlated. Conclusion Through the two-sample MR analysis, we identified extraintestinal manifestations that were causally associated with inflammatory bowel disease and obtained multiple associations from inflammatory bowel disease and gut microbiota, and gut microbiota and extraintestinal manifestations in further analyses. These associations may provide useful biomarkers and potential targets for pathogenesis and treatment.
Collapse
Affiliation(s)
| | | | | | - Heqing Tao
- *Correspondence: Liang Peng, ; Heqing Tao,
| | - Liang Peng
- *Correspondence: Liang Peng, ; Heqing Tao,
| |
Collapse
|
23
|
Peña-Juárez MC, Guadarrama-Escobar OR, Serrano-Castañeda P, Méndez-Albores A, Vázquez-Durán A, Vera-Graziano R, Rodríguez-Pérez B, Salgado-Machuca M, Anguiano-Almazán E, Morales-Florido MI, Rodríguez-Cruz IM, Escobar-Chávez JJ. Synergistic Effect of Retinoic Acid and Lactoferrin in the Maintenance of Gut Homeostasis. Biomolecules 2024; 14:78. [PMID: 38254678 PMCID: PMC10813542 DOI: 10.3390/biom14010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Lactoferrin (LF) is a glycoprotein that binds to iron ions (Fe2+) and other metallic ions, such as Mg2+, Zn2+, and Cu2+, and has antibacterial and immunomodulatory properties. The antibacterial properties of LF are due to its ability to sequester iron. The immunomodulatory capability of LF promotes homeostasis in the enteric environment, acting directly on the beneficial microbiota. LF can modulate antigen-presenting cell (APC) biology, including migration and cell activation. Nonetheless, some gut microbiota strains produce toxic metabolites, and APCs are responsible for initiating the process that inhibits the inflammatory response against them. Thus, eliminating harmful strains lowers the risk of inducing chronic inflammation, and consequently, metabolic disease, which can progress to type 2 diabetes mellitus (T2DM). LF and retinoic acid (RA) exhibit immunomodulatory properties such as decreasing cytokine production, thus modifying the inflammatory response. Their activities have been observed both in vitro and in vivo. The combined, simultaneous effect of these molecules has not been studied; however, the synergistic effect of LF and RA may be employed for enhancing the secretion of humoral factors, such as IgA. We speculate that the combination of LF and RA could be a potential prophylactic alternative for the treatment of metabolic dysregulations such as T2DM. The present review focuses on the importance of a healthy diet for a balanced gut and describes how probiotics and prebiotics with immunomodulatory activity as well as inductors of differentiation and cell proliferation could be acquired directly from the diet or indirectly through the oral administration of formulations aimed to maintain gut health or restore a eubiotic state in an intestinal environment that has been dysregulated by external factors such as stress and a high-fat diet.
Collapse
Affiliation(s)
- Ma. Concepción Peña-Juárez
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Omar Rodrigo Guadarrama-Escobar
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Pablo Serrano-Castañeda
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Abraham Méndez-Albores
- Unidad de Investigación Multidisciplinaria Lab-14 (Ciencia y Tecnología de los Materiales), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (A.M.-A.); (A.V.-D.)
| | - Alma Vázquez-Durán
- Unidad de Investigación Multidisciplinaria Lab-14 (Ciencia y Tecnología de los Materiales), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (A.M.-A.); (A.V.-D.)
| | - Ricardo Vera-Graziano
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México, Apartado Postal 70-360, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, Mexico;
| | - Betsabé Rodríguez-Pérez
- Laboratorio de Servicio de Análisis de Propóleos (LASAP), Unidad de Investigación Multidisciplinaria (UIM), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54714, Mexico;
| | - Mariana Salgado-Machuca
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Ericka Anguiano-Almazán
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Miriam Isabel Morales-Florido
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
- Laboratorio de Farmacia Molecular y Liberación Controlada, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico
| | - Isabel Marlene Rodríguez-Cruz
- Unidad de Enseñanza e Investigación, Hospital Regional e Alta Especialidad de Sumpango, Carretera Zumpango-Jilotzingo #400, Barrio de Santiago, 2ª Sección, Zumpango 55600, Mexico;
| | - José Juan Escobar-Chávez
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| |
Collapse
|
24
|
Bao H, Wang Y, Xiong H, Xia Y, Cui Z, Liu L. Mechanism of Iron Ion Homeostasis in Intestinal Immunity and Gut Microbiota Remodeling. Int J Mol Sci 2024; 25:727. [PMID: 38255801 PMCID: PMC10815743 DOI: 10.3390/ijms25020727] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Iron is a vital trace element that plays an important role in humans and other organisms. It plays an active role in the growth, development, and reproduction of bacteria, such as Bifidobacteria. Iron deficiency or excess can negatively affect bacterial hosts. Studies have reported a major role of iron in the human intestine, which is necessary for maintaining body homeostasis and intestinal barrier function. Organisms can maintain their normal activities and regulate some cancer cells in the body by regulating iron excretion and iron-dependent ferroptosis. In addition, iron can modify the interaction between hosts and microorganisms by altering their growth and virulence or by affecting the immune system of the host. Lactic acid bacteria such as Lactobacillus acidophilus (L. acidophilus), Lactobacillus rhamnosus (L. rhamnosus), and Lactobacillus casei (L. casei) were reported to increase trace elements, protect the host intestinal barrier, mitigate intestinal inflammation, and regulate immune function. This review article focuses on the two aspects of the iron and gut and generally summarizes the mechanistic role of iron ions in intestinal immunity and the remodeling of gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | - Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (H.B.); (Y.W.); (H.X.); (Y.X.)
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (H.B.); (Y.W.); (H.X.); (Y.X.)
| |
Collapse
|
25
|
Abu YF, Singh S, Tao J, Chupikova I, Singh P, Meng J, Roy S. Opioid-induced dysbiosis of maternal gut microbiota during gestation alters offspring gut microbiota and pain sensitivity. Gut Microbes 2024; 16:2292224. [PMID: 38108125 PMCID: PMC10730209 DOI: 10.1080/19490976.2023.2292224] [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: 07/27/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
There has been a rapid increase in neonates born with a history of prenatal opioid exposure. How prenatal opioid exposure affects pain sensitivity in offspring is of interest, as this may perpetuate the opioid epidemic. While few studies have reported hypersensitivity to thermal pain, potential mechanisms have not been described. This study posits that alterations in the gut microbiome may underly hypersensitivity to pain in prenatally methadone-exposed 3-week-old male offspring, which were generated using a mouse model of prenatal methadone exposure. Fecal samples collected from dams and their offspring were subjected to 16s rRNA sequencing. Thermal and mechanical pain were assessed using the tail flick and Von Frey assays. Transcriptomic changes in whole brain samples of opioid or saline-exposed offspring were investigated using RNA-sequencing, and midbrain sections from these animals were subjected to qPCR profiling of genes related to neuropathic and inflammatory pain pathways. Prenatal methadone exposure increased sensitivity to thermal and mechanical pain and elevated serum levels of IL-17a. Taxonomical analysis revealed that prenatal methadone exposure resulted in significant alterations in fecal gut microbiota composition, including depletion of Lactobacillus, Bifidobacterium, and Lachnospiracea sp and increased relative abundance of Akkermansia, Clostridium sensu stricto 1, and Lachnoclostridium. Supplementation of the probiotic VSL#3 in dams rescued hypersensitivity to thermal and mechanical pain in prenatally methadone-exposed offspring. Similarly, cross-fostering prenatally methadone-exposed offspring to control dams also attenuated hypersensitivity to thermal pain in opioid-exposed offspring. Modulation of the maternal and neonatal gut microbiome with probiotics resulted in transcriptional changes in genes related to neuropathic and immune-related signaling in whole brain and midbrain samples of prenatally methadone-exposed offspring. Together, our work provides compelling evidence of the gut-brain-axis in mediating pain sensitivity in prenatally opioid-exposed offspring.
Collapse
Affiliation(s)
- Yaa F. Abu
- Department of Microbiology and Immunology, University of Miami, Miami, FL, USA
| | - Salma Singh
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Junyi Tao
- Department of Surgery, University of Miami, Miami, FL, USA
| | | | - Praveen Singh
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Jingjing Meng
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Sabita Roy
- Department of Surgery, University of Miami, Miami, FL, USA
| |
Collapse
|
26
|
Waldbaum JD, Xhumari J, Akinsuyi OS, Arjmandi B, Anton S, Roesch LFW. Association between Dysbiosis in the Gut Microbiota of Primary Osteoporosis Patients and Bone Loss. Aging Dis 2023; 14:2081-2095. [PMID: 37199579 PMCID: PMC10676803 DOI: 10.14336/ad.2023.0425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/25/2023] [Indexed: 05/19/2023] Open
Abstract
In recent decades, gut microbiome research has experienced significant growth, driven by technological advances that enable quantifying bacterial taxa with greater precision. Age, diet, and living environment have emerged as three key factors influencing gut microbes. Dysbiosis, resulting from alterations in these factors, may lead to changes in bacterial metabolites that regulate pro- and anti-inflammatory processes and consequently impact bone health. Restoration of a healthy microbiome signature could mitigate inflammation and potentially reduce bone loss associated with osteoporosis or experienced by astronauts during spaceflight. However, current research is hindered by contradictory findings, insufficient sample sizes, and inconsistency in experimental conditions and controls. Despite progress in sequencing technology, defining a healthy gut microbiome across global populations remains elusive. Challenges persist in identifying accurate gut bacterial metabolics, specific taxa, and their effects on host physiology. We suggest greater attention be directed towards this issue in Western countries as the cost of treating osteoporosis in the United States reaches billions of dollars annually, with expenses projected to continue rising.
Collapse
Affiliation(s)
- Julien D.H. Waldbaum
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| | - Jessica Xhumari
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| | - Oluwamayowa S. Akinsuyi
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| | - Bahram Arjmandi
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Florida, USA.
| | - Stephen Anton
- Department of Physiology and Aging, College of Public Health and Health Professions, College of Medicine, University of Florida, Florida, USA.
| | - Luiz Fernando Wurdig Roesch
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| |
Collapse
|
27
|
Xie Q, Cui D, Zhu Q, Qin X, Ren D, Xu X. Supplementing maternal diet with milk oligosaccharides and probiotics helps develop the immune system and intestinal flora of offsprings. Food Sci Nutr 2023; 11:6868-6877. [PMID: 37970377 PMCID: PMC10630837 DOI: 10.1002/fsn3.3579] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 11/17/2023] Open
Abstract
Intestinal flora is very important for improving the development of the immune system in newborns. Maternal diet during pregnancy and lactation is one of the key factors affecting the growth and development of offspring. The objective of the present study was to examine whether supplementation of maternal diet with milk oligosaccharides and Bifidobacterium could influence the development of the intestinal flora and immune system of neonatal mice. In total, 30 pregnant Institute of Cancer Research (ICR) mice were randomly divided into six groups: a control group (basal diet) and five intervention groups (basal diet supplemented with different doses of 2'-fucosyllactose [2'-FL] and Bifidobacterium Bb12) during the pregnancy period. All female mice were monitored for physical health during gavage. After delivery, the number of mice in each litter, any deformity, and the development of the offspring were recorded. The spleen, blood, and fecal samples of six groups of 10-12 day-old offspring were collected. The results demonstrated that maternal milk oligosaccharides and probiotics conferred protective effects against lipopolysaccharide (LPS)-induced immunosuppression in mice offspring by significantly enhancing the immune organ indexes, splenocyte proliferation, immunoglobulin (immunoglobulin G, A, M) production as well as improving the macrophage phagocytosis (p < .05). The abundance of Lactobacilli and Bifidobacteria in the feces of offspring mice in the intervention groups was significantly higher than that of the offspring mice in the control group (p < .05). These findings suggest that the combination of 2'-FL and Bifidobacterium Bb12 displayed synergistic interactions between the two components that could promote the development of the immune system of the offsprings and improve their microbiota through maternal ingestion.
Collapse
Affiliation(s)
- Qinggang Xie
- College of Food ScienceNortheast Agricultural UniversityHarbinChina
| | | | - Qinchao Zhu
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Xuewen Qin
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Daxi Ren
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Xiaoxi Xu
- College of Food ScienceNortheast Agricultural UniversityHarbinChina
| |
Collapse
|
28
|
Li H, Ma X, Li Y, Liu Q, Tian Q, Yang X, Zhou Z, Ren J, Sun B, Feng X, Zhang H, Yin X, Li H, Ding X. The metagenomic and metabolomic profile of the gut microbes in Chinese full-term and late preterm infants treated with Clostridium butyricum. Sci Rep 2023; 13:18775. [PMID: 37907561 PMCID: PMC10618524 DOI: 10.1038/s41598-023-45586-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023] Open
Abstract
The present study investigated the composition, abundance, and diversity of gut microbes in full-term and late-preterm infants from a medical center in eastern China. A total of 144 genomes of stool samples were captured for 16S rRNA metagenomic analyses. A high abundance of commensal intestinal bacteria was detected in these samples such as Phocaeicola vulgatus, Escherichia coli, and Faecalibacterium prausnitzii, indicating a relatively consistent diversity of gut microbes in the present full-term infants aged 38-40 weeks. However, late preterm infants (n = 50) with mandatory antimicrobials feeding exhibited lower diversity but a higher composition of opportunistic pathogens such as Enterococcus species. Centralized on the situation, we explored the regulatory effect of Clostridium butyricum as probiotics on these late preterm infants. The consumption of C. butyricum did not restore the composition of gut microbes altered by antimicrobials to normal levels, although several opportunistic pathogens decreased significantly after probiotic therapy including Staphylococcus aureus, Sphingomonas echinoides, and Pseudomonas putida. We also compared the effects of day-fed versus night-fed probiotics. Intriguingly, the nighttime feeding showed a higher proportion of C. butyricum compared with probiotic day-feeding. Finally, fecal metabolome and metabolites were analyzed in late preterm infants with (n = 20) or without probiotic therapy (n = 20). The KEGG enrichment analysis demonstrated that vitamin digestion and absorption, synaptic vesicle cycle, and biotin metabolism were significantly increased in the probiotic-treated group, while MSEA indicated that a series of metabolism were significantly enriched in probiotic-treated infants including glycerolipid, biotin, and lysine, indicating the complex effects of probiotic therapy on glutathione metabolism and nutrients digestion and absorption in late preterm infants. Overall, this study provided metagenomic and metabolomic profile of the gut microbes in full-term newborns and late preterm infants in eastern China. Further studies are needed to support and elucidate the role of probiotic feeding in late preterm infants with mandatory antimicrobial treatment.
Collapse
Affiliation(s)
- Hong Li
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Xingling Ma
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Yongfu Li
- Neonatology Department, Suzhou Science and Technology Town Hospital, Suzhou, Jiangsu, China
| | - Qin Liu
- Neonatology Department, Suzhou Science and Technology Town Hospital, Suzhou, Jiangsu, China
- Pediatric Department, Suzhou New District Yangshan Community Health Service Center, Suzhou, China
| | - Qiuyan Tian
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Xiaofeng Yang
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Zhemin Zhou
- Pasteurien College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Jing Ren
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Bin Sun
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Xing Feng
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China
| | - Hong Zhang
- Taixing People's Hospital, Taizhou, Jiangsu, China
| | - Xiaoping Yin
- Taixing People's Hospital, Taizhou, Jiangsu, China
| | - Heng Li
- Pasteurien College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| | - Xin Ding
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, #303 Jingde Road, Gusu District, Suzhou, 215003, Jiangsu, China.
| |
Collapse
|
29
|
Mahapatro A, Bawna F, Kumar V, Daryagasht AA, Gupta S, Raghuma N, Moghdam SS, Kolla A, Mahapatra SS, Sattari N, Amini-Salehi E, Nayak SS. Anti-inflammatory effects of probiotics and synbiotics on patients with non-alcoholic fatty liver disease: An umbrella study on meta-analyses. Clin Nutr ESPEN 2023; 57:475-486. [PMID: 37739694 DOI: 10.1016/j.clnesp.2023.07.087] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/28/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND AIM The impact of chronic low-grade inflammation in the development of non-alcoholic fatty liver disease (NAFLD) has been studied widely. Previous studies showed gut pathogens' effects on inflammation development in NAFLD patients; hence, hypothetically, gut microbial therapy by administration of probiotics, synbiotics, and prebiotics may alleviate inflammation in these individuals. Several studies were performed in this regard; however, conflicting results were obtained. In this study, we aimed to comprehensively evaluate the effects of gut microbial therapy on inflammatory markers in NAFLD patients in a meta-umbrella design. METHODS Two independent researchers investigated international databases, including PubMed, Web of Science, Scopus, and Cochrane Library, from inception until March 2023. Meta-analyses evaluating the impact of probiotics, synbiotics, or prebiotics on inflammatory markers of patients with NAFLD were eligible for our study. AMASTAR 2 checklist was used to evaluate the quality of included studies. Random effect model was performed for the analysis, and Egger's regression test was conducted to determine publication bias. RESULTS A total number of 12 studies were entered into our analysis. Our findings revealed that gut microbial therapy could significantly reduce serum C-reactive protein (CRP) levels among NAFLD patients (ES: -0.58; 95% CI: -0.73, -0.44, P < 0.001). In subgroup analysis, this reduction was observed with both probiotics (ES: -0.63; 95% CI: -0.81, -0.45, P < 0.001) and synbiotics (ES: -0.49; 95% CI: -0.74, -0.24, P < 0.001). In addition, gut microbial therapy could significantly decrease tumor necrosis factor-a (TNF-a) levels in NAFLD patients (ES: -0.48; 95% CI: -0.67 to -0.30, P < 0.001). In subgroup analysis, this decrease was observed with probiotics (ES: -0.32; 95% CI: -0.53, -0.11, P = 0.002) and synbiotics (ES: -0.96; 95% CI: -1.32, -0.60, P < 0.001). Not enough information was available for assessing prebiotics' impacts. CONCLUSION The results of this umbrella review suggest that probiotics and synbiotics have promising effects on inflammatory markers, including TNF-a and CRP; however, more research is needed regarding the effects of prebiotics. PROSPERO REGISTRATION CODE CRD42022346998.
Collapse
Affiliation(s)
| | - Fnu Bawna
- Dow University of Health Sciences, Karachi, Pakistan
| | | | | | - Siddharth Gupta
- Baptist Memorial Hospital, North Mississippi, Mississippi, USA
| | - Nakka Raghuma
- GSL Medical College and General Hospital, Rajamahendravaram, Andhra Pradesh, India
| | | | - Akshita Kolla
- SRM Medical College Hospital and Research Center, Chennai, India
| | | | - Nazila Sattari
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Sandeep S Nayak
- Department of Internal Medicine, Bridgeport Hospital, Bridgeport, USA
| |
Collapse
|
30
|
Kohda C, Ino S, Ishikawa H, Kuno Y, Nagashima R, Iyoda M. The essential role of intestinal microbiota in cytomegalovirus reactivation. Microbiol Spectr 2023; 11:e0234123. [PMID: 37754566 PMCID: PMC10581228 DOI: 10.1128/spectrum.02341-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/10/2023] [Indexed: 09/28/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a member of Herpesviridae. It has been reported that HCMV is reactivated in the breast milk of HCMV-seropositive lactating women. As we have reported various aspects of the roles of indigenous microbiota, its role in the murine CMV (MCMV) reactivation was examined in this study. MCMV was latently infected in the salivary gland, mammary tissues, and colon in the pregnant mice. When the salivary gland, mammary tissues, and colon were removed 5 days after delivery, MCMV reactivation of latent infection in each organ was confirmed by the detection of MCMV IE1 mRNA using reverse transcription-quantitative PCR. MCMV reactivation was observed in 100% of the mice during pregnancy. Next, for the elimination of intestinal microbiota, the pregnant mice were treated with low-dose or high-dose non-absorbable antibiotics. Although the numbers of aerobe/anaerobe in cecal content in low-dose antibiotic-treated mice were comparable to those in untreated controls, high-dose antibiotic treatment decreased the number of aerobe/anaerobe microbes from ca.9.0 Log10 to ca.3.0 Log10 (cfu/g). However, it could not be confirmed in 16S rRNA analysis that specific bacterial phylum or genus was eliminated by this high-dose treatment. Interestingly, MCMV reactivation was also observed in 100% of low-dose antibiotic-treated mice, whereas, in high-dose antibiotic-treated mice, MCMV reactivation was not observed in the salivary gland or colon. MCMV IE1 mRNA was detected only in 33% of the mammary tissues of those high-dose-treated mice. These results suggest that the indigenous microbiota played a crucial role in the reactivation of latent infection. IMPORTANCE Human cytomegalovirus (HCMV) infection via breast milk is a serious problem for very preterm infants such as developing a sepsis-like syndrome, cholestasis, or bronchopulmonary dysplasia, among others. It has been reported that HCMV is reactivated in the breast milk of HCMV-seropositive lactating women. In this study, the roles of indigenous microbiota in the murine CMV (MCMV) reactivation were examined using a mouse model. In MCMV latently infected mice, MCMV reactivation was observed in 100% of the mice during pregnancy. For the elimination of intestinal microbiota, MCMV-latent mice were treated with non-absorbable antibiotics. After delivery, MCMV reactivation was not observed in antibiotic-treated mice. This result suggested that the indigenous microbiota played a crucial role in the reactivation of latent infection.
Collapse
Affiliation(s)
- Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Satoshi Ino
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
- Department of Medicine, Division of Nephrology, Showa University School of Medicine, Tokyo, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
- Department of Medicine, Division of Nephrology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Abreu Nascimento MD, Matta Alvarez Pimenta ND, Aiceles de Medeiros Pinto Polastri V, Cardoso Chamon R, Sarto Figueiredo M. Immunonutrients and intestinal microbiota: a gap in the literature. Crit Rev Food Sci Nutr 2023; 64:13058-13071. [PMID: 37751225 DOI: 10.1080/10408398.2023.2260468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The human intestinal microbiota is composed of a wide variety of microorganisms that play an important role in intestinal permeability, digestion, and especially, in the maturation of host's immune system. At the same time, effectiveness of immunomodulatory nutrients is known, especially in situations of stress and in strengthening body's defenses. However, the influence of the use of immunonutrients on microbiota's composition and variability is still poorly investigated. Studies indicate that the use of immunomodulators such as omega 3, glutamine, and arginine, can play a role in its modulation, through the immunological enhancement of the hosts. Therefore, this article sought to concentrate the latest evidence on the influence of the use of the main immunonutrients used in clinical practice on human gut microbiota, and their potential benefits.
Collapse
Affiliation(s)
| | - Nina da Matta Alvarez Pimenta
- Graduate Program in Nutrition Science, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil, Niterói, Brazil
| | | | - Raiane Cardoso Chamon
- Graduate Program in Pathology, Department of Pathology, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | | |
Collapse
|
32
|
Liu S, Luo L, Zuo F, Huang X, Zhong L, Liu S, Geng Y, Ou Y, Chen D, Cai W, Deng Y. Ammonia nitrogen stress damages the intestinal mucosal barrier of yellow catfish ( Pelteobagrus fulvidraco) and induces intestinal inflammation. Front Physiol 2023; 14:1279051. [PMID: 37791345 PMCID: PMC10542119 DOI: 10.3389/fphys.2023.1279051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Nitrogen from ammonia is one of the most common pollutants toxics to aquatic species in aquatic environment. The intestinal mucosa is one of the key mucosal defenses of aquatic species, and the accumulation of ammonia nitrogen in water environment will cause irreversible damage to intestinal function. In this study, histology, immunohistochemistry, ultrastructural pathology, enzyme activity analysis and qRT-PCR were performed to reveal the toxic effect of ammonia nitrogen stress on the intestine of Pelteobagrus fulvidraco. According to histological findings, ammonia nitrogen stress caused structural damage to the intestine and reduced the number of mucous cells. Enzyme activity analysis revealed that the activity of bactericidal substances (Lysozyme, alkaline phosphatase, and ACP) had decreased. The ultrastructure revealed sparse and shortened microvilli as well as badly degraded tight junctions. Immunohistochemistry for ZO-1 demonstrated an impaired intestinal mucosal barrier. Furthermore, qRT-PCR revealed that tight junction related genes (ZO-1, Occludin, Claudin-1) were downregulated, while the pore-forming protein Claudin-2 was upregulated. Furthermore, as ammonia nitrogen concentration grew, so did the positive signal of Zap-70 (T/NK cell) and the expression of inflammation-related genes (TNF, IL-1β, IL-8, IL-10). In light of the above findings, we conclude that ammonia nitrogen stress damages intestinal mucosal barrier of Pelteobagrus fulvidraco and induces intestinal inflammation.
Collapse
Affiliation(s)
- Senyue Liu
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, China
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lin Luo
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Fengyuan Zuo
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoli Huang
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liang Zhong
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- State Key Lab of Marine Pollution, Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sha Liu
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yi Geng
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yangping Ou
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Defang Chen
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Wenlong Cai
- State Key Lab of Marine Pollution, Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yongqiang Deng
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
34
|
Smith KB, Murack M, Ismail N. The sex-dependent and enduring impact of pubertal stress on health and disease. Brain Res Bull 2023; 200:110701. [PMID: 37422090 DOI: 10.1016/j.brainresbull.2023.110701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/02/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Illness is often predicated long before the manifestation of its symptoms. Exposure to stressful experiences particularly during critical periods of development, such as puberty and adolescence, can induce various physical and mental illnesses. Puberty is a critical period of maturation for neuroendocrine systems, such as the hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-adrenal (HPA) axes. Exposure to adverse experiences during puberty can impede normal brain reorganizing and remodelling and result in enduring consequences on brain functioning and behaviour. Stress responsivity differs between the sexes during the pubertal period. This sex difference is partly due to differences in circulating sex hormones between males and females, impacting stress and immune responses differently. The effects of stress during puberty on physical and mental health remains under-examined. The purpose of this review is to summarize the most recent findings pertaining to age and sex differences in HPA axis, HPG axis, and immune system development, and describe how disruption in the functioning of these systems can propagate disease. Lastly, we delve into the notable neuroimmune contributions, sex differences, and the mediating role of the gut microbiome on stress and health outcomes. Understanding the enduring consequences of adverse experiences during puberty on physical and mental health will allow a greater proficiency in treating and preventing stress-related diseases early in development.
Collapse
Affiliation(s)
- Kevin B Smith
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada
| | - Michael Murack
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada
| | - Nafissa Ismail
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada; LIFE Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
35
|
Hickey JW, Becker WR, Nevins SA, Horning A, Perez AE, Zhu C, Zhu B, Wei B, Chiu R, Chen DC, Cotter DL, Esplin ED, Weimer AK, Caraccio C, Venkataraaman V, Schürch CM, Black S, Brbić M, Cao K, Chen S, Zhang W, Monte E, Zhang NR, Ma Z, Leskovec J, Zhang Z, Lin S, Longacre T, Plevritis SK, Lin Y, Nolan GP, Greenleaf WJ, Snyder M. Organization of the human intestine at single-cell resolution. Nature 2023; 619:572-584. [PMID: 37468586 PMCID: PMC10356619 DOI: 10.1038/s41586-023-05915-x] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/02/2023] [Indexed: 07/21/2023]
Abstract
The intestine is a complex organ that promotes digestion, extracts nutrients, participates in immune surveillance, maintains critical symbiotic relationships with microbiota and affects overall health1. The intesting has a length of over nine metres, along which there are differences in structure and function2. The localization of individual cell types, cell type development trajectories and detailed cell transcriptional programs probably drive these differences in function. Here, to better understand these differences, we evaluated the organization of single cells using multiplexed imaging and single-nucleus RNA and open chromatin assays across eight different intestinal sites from nine donors. Through systematic analyses, we find cell compositions that differ substantially across regions of the intestine and demonstrate the complexity of epithelial subtypes, and find that the same cell types are organized into distinct neighbourhoods and communities, highlighting distinct immunological niches that are present in the intestine. We also map gene regulatory differences in these cells that are suggestive of a regulatory differentiation cascade, and associate intestinal disease heritability with specific cell types. These results describe the complexity of the cell composition, regulation and organization for this organ, and serve as an important reference map for understanding human biology and disease.
Collapse
Affiliation(s)
- John W Hickey
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Winston R Becker
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Aaron Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Almudena Espin Perez
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Bokai Zhu
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Bei Wei
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Roxanne Chiu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Derek C Chen
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Daniel L Cotter
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Edward D Esplin
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Annika K Weimer
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Chiara Caraccio
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | | | - Christian M Schürch
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Sarah Black
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Maria Brbić
- Department of Computer Science, Stanford University, Stanford, CA, USA
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kaidi Cao
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Shuxiao Chen
- Department of Statistics and Data Science, University of Pennsylvania, Pennsylvania, PA, USA
| | - Weiruo Zhang
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Nancy R Zhang
- Department of Statistics and Data Science, University of Pennsylvania, Pennsylvania, PA, USA
| | - Zongming Ma
- Department of Statistics and Data Science, University of Pennsylvania, Pennsylvania, PA, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Zhengyan Zhang
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Shin Lin
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Teri Longacre
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, USA
| | - Yiing Lin
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Garry P Nolan
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA.
| | | | - Michael Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
36
|
Jeong H, Kim S, Hwang US, Choi H, Park YS. Immunostimulatory Activity of Lactococcus lactis subsp. lactis CAB701 Isolated from Jeju Cabbage. Microorganisms 2023; 11:1718. [PMID: 37512890 PMCID: PMC10385365 DOI: 10.3390/microorganisms11071718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/19/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
This study explored the potential of Lactococcus lactis subsp. lactis CAB701 as a probiotic strain, focusing on its immunostimulatory properties. Despite adverse conditions in the gastrointestinal environment, this strain exhibited remarkable survivability, as evidenced by its tolerance to acid, bile, and pancreatin, coupled with its impressive ability to adhere to Caco-2 cells. It also exhibited significant antioxidant activity, similar to the established probiotic Lacticaseibacillus rhamnosus GG (LGG). Our research elucidates the potent immunostimulatory effects of L. lactis subsp. lactis CAB701. This strain significantly enhanced nitric oxide production in RAW 264.7, far exceeding that obtained with LGG. An in-depth examination revealed elevated expression of key inflammatory mediators, including inducible nitric oxide synthase, tumor necrosis factor-alpha, cyclooxygenase-2, interleukin (IL)-1 beta, and IL-6. L. lactis subsp. lactis CAB701 increases the expression of critical signaling proteins in the mitogen-activated protein kinase pathway. This prompted a substantial increase in the expression of phosphorylated c-Jun N-terminal kinases and extracellular signal-regulated kinases, suggesting their role in modulating these immune-related pathways. Overall, these findings demonstrate the significant immunostimulatory capacity of L. lactis subsp. lactis CAB701, positioning it as a potential candidate for probiotic use, especially in applications that enhance immune responses.
Collapse
Affiliation(s)
- Huijin Jeong
- Department of Food Science and Biotechnology, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Suin Kim
- Department of Food Science and Biotechnology, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Un-Sik Hwang
- BKbio Co., Ltd., Jeju-si 63359, Republic of Korea
| | | | - Young-Seo Park
- Department of Food Science and Biotechnology, Gachon University, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
37
|
Li J, Feng S, Wang Z, He J, Zhang Z, Zou H, Wu Z, Liu X, Wei H, Tao S. Limosilactobacillus mucosae-derived extracellular vesicles modulates macrophage phenotype and orchestrates gut homeostasis in a diarrheal piglet model. NPJ Biofilms Microbiomes 2023; 9:33. [PMID: 37280255 DOI: 10.1038/s41522-023-00403-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/22/2023] [Indexed: 06/08/2023] Open
Abstract
The diarrheal disease causes high mortality, especially in children and young animals. The gut microbiome is strongly associated with diarrheal disease, and some specific strains of bacteria have demonstrated antidiarrheal effects. However, the antidiarrheal mechanisms of probiotic strains have not been elucidated. Here, we used neonatal piglets as a translational model and found that gut microbiota dysbiosis observed in diarrheal piglets was mainly characterized by a deficiency of Lactobacillus, an abundance of Escherichia coli, and enriched lipopolysaccharide biosynthesis. Limosilactobacillus mucosae and Limosilactobacillus reuteri were a signature bacterium that differentiated healthy and diarrheal piglets. Germ-free (GF) mice transplanted with fecal microbiota from diarrheal piglets reproduced diarrheal disease symptoms. Administration of Limosilactobacillus mucosae but not Limosilactobacillus reuteri alleviated diarrheal disease symptoms induced by fecal microbiota of diarrheal piglets and by ETEC K88 challenge. Notably, Limosilactobacillus mucosae-derived extracellular vesicles alleviated diarrheal disease symptoms caused by ETEC K88 by regulating macrophage phenotypes. Macrophage elimination experiments demonstrated that the extracellular vesicles alleviated diarrheal disease symptoms in a macrophage-dependent manner. Our findings provide insights into the pathogenesis of diarrheal disease from the perspective of intestinal microbiota and the development of probiotic-based antidiarrheal therapeutic strategies.
Collapse
Affiliation(s)
- Jingjing Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuaifei Feng
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jinhui He
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zeyue Zhang
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huicong Zou
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhifeng Wu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiangdong Liu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Hong Wei
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
38
|
Liu L, Yin M, Gao J, Yu C, Lin J, Wu A, Zhu J, Xu C, Liu X. Intestinal Barrier Function in the Pathogenesis of Nonalcoholic Fatty Liver Disease. J Clin Transl Hepatol 2023; 11:452-458. [PMID: 36643028 PMCID: PMC9817057 DOI: 10.14218/jcth.2022.00089] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/09/2022] [Accepted: 07/29/2022] [Indexed: 01/18/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide. The mechanisms involved in NAFLD onset are complicated and multifactorial. Recent literature has indicated that altered intestinal barrier function is related to the occurrence and progression of liver disease. The intestinal barrier is important for absorbing nutrients and electrolytes and for defending against toxins and antigens in the enteric environment. Major mechanisms by which the intestinal barrier influences the development of NAFLD involve the altered epithelial layer, decreased intracellular junction integrity, and increased intestinal barrier permeability. Increased intestinal permeability leads to luminal dysbiosis and allows the translocation of pathogenic bacteria and metabolites into the liver, inducing inflammation, immune response, and hepatocyte injury in NAFLD. Although research has been directed to NAFLD in recent decades, the pathophysiological changes in NAFLD initiation and progression are still not completely understood, and the therapeutic targets remain limited. A deeper understanding on the correlation between NAFLD pathogenesis and intestinal barrier regulation must be attained. Therefore, in this review, the components of the intestinal barrier and their respective functions and disruptions during the progression of NAFLD are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chunfang Xu
- Correspondence to: Xiaolin Liu and Chunfang Xu, Department of Gastroenterology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215006, China. ORCID: https://orcid.org/0000-0003-4560-7589 (XL) and https://orcid.org/0000-0001-5648-3003 (CX). Tel/Fax: +86-512-65223637, E-mail: (XL) and (CX)
| | - Xiaolin Liu
- Correspondence to: Xiaolin Liu and Chunfang Xu, Department of Gastroenterology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215006, China. ORCID: https://orcid.org/0000-0003-4560-7589 (XL) and https://orcid.org/0000-0001-5648-3003 (CX). Tel/Fax: +86-512-65223637, E-mail: (XL) and (CX)
| |
Collapse
|
39
|
Kouidhi S, Zidi O, Belkhiria Z, Rais H, Ayadi A, Ben Ayed F, Mosbah A, Cherif A, El Gaaied ABA. Gut microbiota, an emergent target to shape the efficiency of cancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:240-265. [PMID: 37205307 PMCID: PMC10185446 DOI: 10.37349/etat.2023.00132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/04/2023] [Indexed: 05/21/2023] Open
Abstract
It is now well-acknowledged that microbiota has a profound influence on both human health and illness. The gut microbiota has recently come to light as a crucial element that influences cancer through a variety of mechanisms. The connections between the microbiome and cancer therapy are further highlighted by a number of preclinical and clinical evidence, suggesting that these complicated interactions may vary by cancer type, treatment, or even by tumor stage. The paradoxical relationship between gut microbiota and cancer therapies is that in some cancers, the gut microbiota may be necessary to maintain therapeutic efficacy, whereas, in other cancers, gut microbiota depletion significantly increases efficacy. Actually, mounting research has shown that the gut microbiota plays a crucial role in regulating the host immune response and boosting the efficacy of anticancer medications like chemotherapy and immunotherapy. Therefore, gut microbiota modulation, which aims to restore gut microbial balance, is a viable technique for cancer prevention and therapy given the expanding understanding of how the gut microbiome regulates treatment response and contributes to carcinogenesis. This review will provide an outline of the gut microbiota's role in health and disease, along with a summary of the most recent research on how it may influence the effectiveness of various anticancer medicines and affect the growth of cancer. This study will next cover the newly developed microbiota-targeting strategies including prebiotics, probiotics, and fecal microbiota transplantation (FMT) to enhance anticancer therapy effectiveness, given its significance.
Collapse
Affiliation(s)
- Soumaya Kouidhi
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
| | - Oumaima Zidi
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
- Department of Biologu, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
| | | | - Henda Rais
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
- Service d’Oncologie Médicale, Hôpital Salah-Azaïz, Tunis 1006, Tunisia
| | - Aida Ayadi
- Department of Pathology, Abderrahman Mami Hospital, University of Tunis El Manar, Ariana 2080, Tunisia
| | - Farhat Ben Ayed
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
| | - Amor Mosbah
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
| | - Ameur Cherif
- Laboratory BVBGR-LR11ES31, Biotechnopole Sidi Thabet, University Manouba, ISBST, Ariana 2020, Tunisia
| | - Amel Ben Ammar El Gaaied
- Laboratory of Genetics, Immunology and Human Pathology, Department of Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
| |
Collapse
|
40
|
Immunity orchestrates a bridge in gut-brain axis of neurodegenerative diseases. Ageing Res Rev 2023; 85:101857. [PMID: 36669690 DOI: 10.1016/j.arr.2023.101857] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Neurodegenerative diseases, in particular for Alzheimer's disease (AD), Parkinson's disease (PD) and Multiple sclerosis (MS), are a category of diseases with progressive loss of neuronal structure or function (encompassing neuronal death) leading to neuronal dysfunction, whereas the underlying pathogenesis remains to be clarified. As the microbiological ecosystem of the intestinal microbiome serves as the second genome of the human body, it is strongly implicated as an essential element in the initiation and/or progression of neurodegenerative diseases. Nevertheless, the precise underlying principles of how the intestinal microflora impact on neurodegenerative diseases via gut-brain axis by modulating the immune function are still poorly characterized. Consequently, an overview of initiating the development of neurodegenerative diseases and the contribution of intestinal microflora on immune function is discussed in this review.
Collapse
|
41
|
Shekhar S, Brar NK, Petersen FC. Suppressive effect of therapeutic antibiotic regimen on antipneumococcal Th1/Th17 responses in neonatal mice. Pediatr Res 2023; 93:818-826. [PMID: 35778498 DOI: 10.1038/s41390-022-02115-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Antibiotics are commonly used in human neonates, but their impact on neonatal T cell immunity remains poorly understood. The aim of this study was to investigate the impact of the antibiotic piperacillin with the beta-lactamase inhibitor tazobactam on neonatal CD4+ and CD8+ T cell responses to Streptococcus pneumoniae. METHODS Splenic and lung cells were isolated from the neonatal mice receiving piperacillin and tazobactam or saline (sham) and cultured with S. pneumoniae to analyze T cell cytokine production by ELISA and flow cytometry. RESULTS Antibiotic exposure to neonatal mice resulted in reduced numbers of CD4+/CD8+ T cells in the spleen and lungs compared to control mice. Upon in vitro stimulation with S. pneumoniae, splenocytes and lung cells from antibiotic-exposed mice produced lower levels of IFN-γ (Th1)/IL-17A (Th17) and IL-17A cytokines, respectively. Flow cytometric analysis revealed that S. pneumoniae-stimulated splenic CD4+ T cells from antibiotic-exposed mice expressed decreased levels of IFN-γ and IL-17A compared to control mice, whereas lung CD4+ T cells produced lower levels of IL-17A. However, no significant difference was observed for IL-4 (Th2) production. CONCLUSIONS Neonatal mice exposure to piperacillin and tazobactam reduces the number of CD4+ and CD8+ T cells, and suppresses Th1 and Th17, but not Th2, responses to S. pneumoniae. IMPACT Exposure of neonatal mice with a combination of piperacillin and tazobactam reduces CD4+/CD8+ T cells in the spleen and lungs. Antibiotic exposure suppresses neonatal Th1 and Th17, but not Th2, responses to Streptococcus pneumoniae. Our findings may have important implications for developing better therapeutic strategies in the neonatal intensive care unit.
Collapse
Affiliation(s)
| | - Navdeep K Brar
- Institute of Oral Biology, University of Oslo, Oslo, Norway
| | | |
Collapse
|
42
|
Combination Therapy with Indigo and Indirubin for Ulcerative Colitis via Reinforcing Intestinal Barrier Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2894695. [PMID: 36825081 PMCID: PMC9943625 DOI: 10.1155/2023/2894695] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 02/16/2023]
Abstract
Indigo and indirubin, the active molecules of traditional Chinese medicine indigo naturalis, exert therapeutic activity for ulcerative colitis (UC). Indigo and indirubin are isomers and have distinctive profiles in anti-inflammation, immune regulation, intestinal microbiota regulation, oxidative stress regulation, and intestinal mucosal repair for UC treatment. Thus, exploring its combined administration's integrated advantages for UC is critical. This study is aimed at clarifying the effect and mechanisms of the combined administration of indigo and indirubin on colitis mouse models. The results showed that all the treatment groups could improve the disease symptoms, and the combined administration showed the best effect. Additionally, compared with indigo and indirubin alone, the combination group could significantly reinforce intestinal barrier function by increasing the expression of E-cadherin, occludin, ZO-1, and MUC2 and improving intestinal permeability. The treatment groups significantly improved the expression of cytokines, including TNF-α, IFN-γ, IL-12, IL-23, and IL-17A, and indirubin presented the most potent anti-inflammatory effect. Furthermore, all the treatment groups reduced the infiltration of the immune cells in intestinal lamina propria and the production of ROS/RNS. Notably, indigo exhibited a more substantial capacity to regulate natural killer (NK) cells, ILC3, neutrophils, and dendritic cells, followed by the combination group and indirubin alone. Finally, all the treatment groups modulated intestinal microbiota composition, increased the proportion of beneficial microbiota, and decreased the proportion of microbiota. Our results indicated that indigo and indirubin synergistically reinforced the intestinal barrier function, which may be associated with integrating the indirubin anti-inflammatory and intestinal microbiota regulating strength and indigo immune and ROS/RNS regulation advantage.
Collapse
|
43
|
Ding Y, Yao X, Zhang H, He X, Song Z. Maternal antibiotic treatment during pregnancy attenuates the transport and absorption of maternal antibody IgG through TLR4 and TLR2 receptor. Front Microbiol 2023; 14:1109273. [PMID: 36891396 PMCID: PMC9986424 DOI: 10.3389/fmicb.2023.1109273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Maternal antibody IgG, the main antibody in colostrum, plays an important role in neonates protection. Commensal microbiota is closely related to host antibody repertoire. However, there are few reports on how maternal gut microbiota affects maternal antibody IgG transfer. In the present study, we investigated the effects of altering the gut microbiota (treated with antibiotics during pregnancy) on maternal IgG transportation and offspring absorption and explored its underlying mechanisms. Results showed that antibiotic treatment during pregnancy significantly decreased maternal cecal microbial richness (Chao1 and Obesrved species) and diversity (Shannon and Simpson). Plasma metabolome enriched significant changes in the process of bile acid secretion pathway, and the concentration of deoxycholic acid, a secondary metabolite of microorganisms was lowered. Flow cytometry analysis indicated that antibiotic treatment promoted the number of B cells and abated the number of T, DC and M1 cells in intestinal lamina propria of dams. Surprisingly, the serum IgG level in antibiotic treated dams was significantly increased, while IgG contents in colostrum was decreased. Moreover, pregnancy antibiotic treatment in dams was reduced the expression of FcRn, TLR4 and TLR2 in breast of dams and in duodenum and jejunum of neonates. Furthermore, TLR4-/- and TLR2-/- knock-out mice showed a lower FcRn expression in breast of dams and in duodenum and jejunum of neonates. These findings suggest that maternal intestine bacteria may affect the maternal IgG transfer through regulating the breast TLR4 and TLR2 of dams.
Collapse
Affiliation(s)
- Yanan Ding
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaofeng Yao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
44
|
Immunomodulatory action of Lactococcuslactis. J Biosci Bioeng 2023; 135:1-9. [PMID: 36428209 DOI: 10.1016/j.jbiosc.2022.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/07/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022]
Abstract
Fermented foods are gaining popularity due to health-promoting properties with high levels of nutrients, phytochemicals, bioactive compounds, and probiotic microorganisms. Due to its unique fermentation process, Lactococcus lactis plays a key role in the food business, notably in the manufacturing of dairy products. The superior biological activities of L. lactis in these functional foods include anti-inflammatory and immunomodulatory capabilities. L. lactis boosted growth performance, controlled amino acid profiles, intestinal immunology, and microbiota. Besides that, the administration of L. lactis increased the rate of infection clearance. Innate and acquired immune responses would be upregulated in both local and systemic compartments, resulting in these consequences. L. lactis is often employed in the food sector and is currently being exploited as a delivery vehicle for biological research. These bacteria are being eyed as potential candidates for biotechnological applications. With this in mind, we reviewed the immunomodulatory effects of different L. lactis strains.
Collapse
|
45
|
Kang R, Li S, Perveen A, Shen J, Li C. Effects of maternal T-2 toxin exposure on microorganisms and intestinal barrier function in young mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114252. [PMID: 36332402 DOI: 10.1016/j.ecoenv.2022.114252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 06/16/2023]
Abstract
T-2 toxin belongs to the trichothecenes group A compound, mainly produced by Fusarium fungi. It has been shown that T-2 toxin could cross the placental barrier and breast milk, thus endangering the health of offspring. The present study aimed to explore the effects of maternal T-2 toxin exposure on the integrity of the intestinal barrier and the intestinal microflora of young mice. From late pregnancy (GD 14) to lactation (LD 21), pregnant mice were given T-2 toxin daily at 0, 0.005, or 0.05 mg T-2 toxin/kg BW. Postnatal day 21 (PND21), PND28, and PND56 young mice were chosen as objects to detect the influences of maternal T-2 toxin exposure to mice on the offspring. The results showed that maternal exposure to T-2 toxin disturbed the balance of the intestinal microbial flora of the young mice. Villous adhesions and fusion of ileum were observed in T-2-treated groups. In addition, supplementation of T-2 toxin significantly decreased the gene expressions of Claudin 1, Occludin, Tjp1, Il10, Il6, and Tnf in PND 21. However, in PND 28, the expressions of Tnf were significantly increased. The expressions of Claudin 1, Occludin, Tjp1, Il10, Il6 and Tnf were significantly increased after T-2 toxin treatment in PND 56. These results suggested that maternal exposure to T-2 toxin has negative influences on the intestine of young mice, which may be due to the alterations of microbial composition.
Collapse
Affiliation(s)
- Ruifen Kang
- Research Center for Livestock Environmental Control and Smart Production, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Sheng Li
- Research Center for Livestock Environmental Control and Smart Production, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Aneela Perveen
- Research Center for Livestock Environmental Control and Smart Production, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jiakun Shen
- Research Center for Livestock Environmental Control and Smart Production, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Chunmei Li
- Research Center for Livestock Environmental Control and Smart Production, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
46
|
Leung JSM. Interaction between gut microbiota and COVID-19 and its vaccines. World J Gastroenterol 2022; 28:5801-5806. [PMID: 36353201 PMCID: PMC9639653 DOI: 10.3748/wjg.v28.i40.5801] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/07/2022] [Accepted: 10/14/2022] [Indexed: 02/06/2023] Open
Abstract
The whole world has been continuously afflicted by the coronavirus disease 2019 (COVID-19) pandemic for the past 3 years. Many countries have tried many methods to control this virus infection with varying successes and failures. The gut microbiota is a biosystem spanning the entire length of the digestive tract and playing important roles in health and disease. It is much affected by COVID-19. In return it also substantially impacts infection. In particular, the gut microbiota has established a bidirectional interaction with the COVID-19 vaccines, enhancing or reducing vaccine efficacy by virtue of its varying components. Conversely, COVID-19 vaccines also make a substantial impact on the gut microbiota, re-ducing its overall population and biodiversity. It is hoped that by exploring and harnessing this bidirectional interaction we may break new ground and develop new methods to prevent and treat this formidable virus infection.
Collapse
Affiliation(s)
- John S M Leung
- Cardiothoracic Unit, St. Paul’s Hospital, Hong Kong, China
| |
Collapse
|
47
|
Qin H, Wu H, Shen K, Liu Y, Li M, Wang H, Qiao Z, Mu Z. Fermented Minor Grain Foods: Classification, Functional Components, and Probiotic Potential. Foods 2022; 11:3155. [PMID: 37430904 PMCID: PMC9601907 DOI: 10.3390/foods11203155] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 08/05/2023] Open
Abstract
Fermented minor grain (MG) foods often have unique nutritional value and functional characteristics, which are important for developing dietary culture worldwide. As a kind of special raw material in fermented food, minor grains have special functional components, such as trace elements, dietary fiber, and polyphenols. Fermented MG foods have excellent nutrients, phytochemicals, and bioactive compounds and are consumed as a rich source of probiotic microbes. Thus, the purpose of this review is to introduce the latest progress in research related to the fermentation products of MGs. Specific discussion is focused on the classification of fermented MG foods and their nutritional and health implications, including studies of microbial diversity, functional components, and probiotic potential. Furthermore, this review discusses how mixed fermentation of grain mixtures is a better method for developing new functional foods to increase the nutritional value of meals based on cereals and legumes in terms of dietary protein and micronutrients.
Collapse
Affiliation(s)
- Huibin Qin
- Center for Agricultural Genetic Resources Research, Shanxi Agricultural University, Key Laboratory of Crop Gene Resources and Germplasm Enhancement on Loess Plateau, Ministry of Agriculture, Shanxi Key Laboratory of Genetic Resources and Genetic Improvement of Minor Crops, Taiyuan 030031, China
| | - Houbin Wu
- Shennong Technology Group Co., Ltd., Jinzhong 030801, China
| | - Ke Shen
- Shennong Technology Group Co., Ltd., Jinzhong 030801, China
| | - Yilin Liu
- Shennong Technology Group Co., Ltd., Jinzhong 030801, China
| | - Meng Li
- Center for Agricultural Genetic Resources Research, Shanxi Agricultural University, Key Laboratory of Crop Gene Resources and Germplasm Enhancement on Loess Plateau, Ministry of Agriculture, Shanxi Key Laboratory of Genetic Resources and Genetic Improvement of Minor Crops, Taiyuan 030031, China
| | - Haigang Wang
- Center for Agricultural Genetic Resources Research, Shanxi Agricultural University, Key Laboratory of Crop Gene Resources and Germplasm Enhancement on Loess Plateau, Ministry of Agriculture, Shanxi Key Laboratory of Genetic Resources and Genetic Improvement of Minor Crops, Taiyuan 030031, China
| | - Zhijun Qiao
- Center for Agricultural Genetic Resources Research, Shanxi Agricultural University, Key Laboratory of Crop Gene Resources and Germplasm Enhancement on Loess Plateau, Ministry of Agriculture, Shanxi Key Laboratory of Genetic Resources and Genetic Improvement of Minor Crops, Taiyuan 030031, China
| | - Zhixin Mu
- Center for Agricultural Genetic Resources Research, Shanxi Agricultural University, Key Laboratory of Crop Gene Resources and Germplasm Enhancement on Loess Plateau, Ministry of Agriculture, Shanxi Key Laboratory of Genetic Resources and Genetic Improvement of Minor Crops, Taiyuan 030031, China
| |
Collapse
|
48
|
Effects of Two Kinds of Extracts of Cistanche deserticola on Intestinal Microbiota and Its Metabolism. Foods 2022; 11:foods11182897. [PMID: 36141024 PMCID: PMC9498788 DOI: 10.3390/foods11182897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cistanche deserticola belongs to the Liedang family. Known as "desert ginseng", it has high medicinal value and plays important roles in endocrine regulation, neuroprotection, immune regulation, and other processes. Some studies have shown that single substances such as polysaccharides and phenylethanolside can affect intestinal microbiota, but few studies have studied the synergistic effect of various components in Cistanche deserticola extracts on intestinal microbiota. Therefore, in this study, through an in vitro digestion model (Changdao Moni, CDMN) combined with 16S rRNA gene amplification sequencing technology and untargeted metabolomics technology, it was found that the two extracts all had significant effects on the enteric cavity and mucosal flora. Both extracts inhibited Bacteroides in the intestinal cavity and Parabacteroides and Ruminococcus 2 in the intestinal mucosa and promoted Bifidobacterium and Prevotella in the intestinal cavity and Megasphaera in the intestinal mucosa. The aqueous extract also inhibited Phascolarctobacterium. Both extracts also significantly increased the production of short-chain fatty acids, especially butyrate. The intake of extract had significant effects on the metabolic pathways related to amino acids and lipids. Indoles were upregulated by the aqueous extract but downregulated by the alcohol extract. In addition, the extract also had a significant effect on the hemolytic phosphorus esters. In conclusion, the two kinds of extracts have different effects on intestinal microbiota and its metabolism. This study provides guiding significance for the edibility and food development of Cistanche deserticola.
Collapse
|
49
|
Sardinha-Silva A, Alves-Ferreira EVC, Grigg ME. Intestinal immune responses to commensal and pathogenic protozoa. Front Immunol 2022; 13:963723. [PMID: 36211380 PMCID: PMC9533738 DOI: 10.3389/fimmu.2022.963723] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
The physical barrier of the intestine and associated mucosal immunity maintains a delicate homeostatic balance between the host and the external environment by regulating immune responses to commensals, as well as functioning as the first line of defense against pathogenic microorganisms. Understanding the orchestration and characteristics of the intestinal mucosal immune response during commensal or pathological conditions may provide novel insights into the mechanisms underlying microbe-induced immunological tolerance, protection, and/or pathogenesis. Over the last decade, our knowledge about the interface between the host intestinal mucosa and the gut microbiome has been dominated by studies focused on bacterial communities, helminth parasites, and intestinal viruses. In contrast, specifically how commensal and pathogenic protozoa regulate intestinal immunity is less well studied. In this review, we provide an overview of mucosal immune responses induced by intestinal protozoa, with a major focus on the role of different cell types and immune mediators triggered by commensal (Blastocystis spp. and Tritrichomonas spp.) and pathogenic (Toxoplasma gondii, Giardia intestinalis, Cryptosporidium parvum) protozoa. We will discuss how these various protozoa modulate innate and adaptive immune responses induced in experimental models of infection that benefit or harm the host.
Collapse
|
50
|
Maternal stevioside supplementation improves intestinal immune function of chicken offspring potentially via modulating gut microbiota and down-regulating the promoter methylation level of suppressor of cytokine signaling 1 (SOCS1). ANIMAL NUTRITION 2022; 10:329-346. [PMID: 35919247 PMCID: PMC9307571 DOI: 10.1016/j.aninu.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/18/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022]
Abstract
The intestinal immune function of chickens is limited during the early growing stage. Maternal nutritional intervention has been suggested to affect the innate immunity of offspring. The present study aimed to investigate the effects of maternal stevioside supplementation on the intestinal immune function of chicken offspring. A total of 120 Jinmao yellow-feathered breeder hens were fed a basal diet or a diet supplemented with 250 mg/kg stevioside for 5 weeks. During the last week, 200 breeding eggs from each group were collected for incubation. After hatching, 80 male offspring (40 chickens from each group) were randomly selected and fed the same basal diet for 28 d. In addition, 90 well-shaped fertile eggs of non-treated breeder hens were incubated for the in ovo injection experiment. Steviol dissolved in 20% glycerol was injected at 7 d of incubation. The results showed that maternal stevioside supplementation could improve embryonic development, jejunal integrity and proliferation in the jejunal crypt (P < 0.05). Maternal stevioside supplementation could also increase the innate transcription levels of cytokines and endotoxin tolerance-related factors in the jejunum of chicken offspring (P < 0.05). At 28 d of age, the offspring following maternal stevioside supplementation exhibited higher jejunal secretory immunoglobulin A and serum interferons levels (P < 0.05). A higher abundance of Lactobacillales induced by maternal stevioside supplementation was positively correlated with intestinal immune-related factors (P < 0.05). The in ovo injection with steviol did not alter either embryonic development or intestinal immune function of hatching chickens (P > 0.05). Furthermore, maternal stevioside supplementation could induce hypo-methylation on the promoter region of suppressor of cytokine signaling 1 (SOCS1). In conclusion, maternal stevioside supplementation could improve the intestinal immune function of chicken offspring potentially via modulating the gut microbiota and down-regulating the promoter methylation level of SOCS1.
Collapse
|