1
|
Lee JG. Microbiota, Gut Health, and Laryngopharyngeal Reflux Disease. Otolaryngol Clin North Am 2025; 58:433-440. [PMID: 39472179 DOI: 10.1016/j.otc.2024.09.006] [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: 01/02/2025]
Abstract
Recent advances in technology have allowed examination of microbial communities in efforts to classify microbiomes of "healthy" individuals. The gut and the oral cavity have been extensively researched but the upper airway (including the pharynx and larynx) has not received the same attention. This review details the emergence of microbiota as a field of interest and reviews existing evidence supporting a relationship between microbiome alterations and laryngopharyngeal reflux symptoms, as well as potential therapeutic interventions.
Collapse
Affiliation(s)
- Jessica G Lee
- Charleston ENT and Allergy, 2295 Henry Tecklenburg Drive, Charleston, SC 29414, USA.
| |
Collapse
|
2
|
Mukhopadhya I, Louis P. Gut microbiota-derived short-chain fatty acids and their role in human health and disease. Nat Rev Microbiol 2025:10.1038/s41579-025-01183-w. [PMID: 40360779 DOI: 10.1038/s41579-025-01183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/15/2025]
Abstract
Short-chain fatty acids (SCFAs) are a group of organic compounds produced by the fermentation of dietary fibre by the human gut microbiota. They play diverse roles in different physiological processes of the host with implications for human health and disease. This Review provides an overview of the complex microbial metabolism underlying SCFA formation, considering microbial interactions and modulating factors of the gut environment. We explore the multifaceted mechanistic interactions between SCFAs and the host, with a particular focus on the local actions of SCFAs in the gut and their complex interactions with the immune system. We also discuss how these actions influence intestinal and extraintestinal diseases and emerging therapeutic strategies using SCFAs.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Petra Louis
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
3
|
Ramar M, Wiscovitch-Russo R, Yano N, Singh H, Lamere E, Short M, Gonzalez-Juarbe N, Fedulov AV. Live bacteria in gut microbiome dictate asthma onset triggered by environmental particles via modulation of DNA methylation in dendritic cells. Cell Rep 2025; 44:115684. [PMID: 40372916 DOI: 10.1016/j.celrep.2025.115684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/24/2025] [Accepted: 04/18/2025] [Indexed: 05/17/2025] Open
Abstract
Despite broad knowledge of the pathogenesis, our understanding of the origin of allergy and asthma remains poor, preventing etiotropic treatments. The gut microbiome is seen to be altered in asthmatics; however, proof of causality of the microbiome alterations is lacking. We report on gut microbiome transplantation (GMT) from mice predisposed to asthma by maternal exposure to pro-allergy environmental particles into naive recipients. This GMT confers asthma predisposition, and the effect is abrogated by gamma sterilization of the transplant material or by co-administration of antibacterials, indicating that viable bacteria are mediating the effect. Metagenomics identifies key changes in the "pro-asthma" microbiome, and metabolomics links the identified species to altered production of butyrate known to act on immune cells and epigenetic mechanisms. We further show that transplant recipients develop DNA methylation alterations in dendritic cells. Finally, dendritic cells with an altered methylome present allergen to T cells, and this effect is abrogated by an epigenetically acting drug in vitro.
Collapse
Affiliation(s)
- Mohankumar Ramar
- Department of Surgery, Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Rosana Wiscovitch-Russo
- Department of Infectious Diseases and Genomic Medicine, J. Craig Venter Institute, Rockville, MD, USA
| | - Naohiro Yano
- Department of Surgery, Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Harinder Singh
- Department of Infectious Diseases and Genomic Medicine, J. Craig Venter Institute, Rockville, MD, USA
| | - Edward Lamere
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael Short
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Norberto Gonzalez-Juarbe
- Department of Infectious Diseases and Genomic Medicine, J. Craig Venter Institute, Rockville, MD, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA.
| | - Alexey V Fedulov
- Department of Surgery, Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
4
|
Liu L, Zhao W, Zhang H, Shang Y, Huang W, Cheng Q. Relationship between pediatric asthma and respiratory microbiota, intestinal microbiota: a narrative review. Front Microbiol 2025; 16:1550783. [PMID: 40415934 PMCID: PMC12099452 DOI: 10.3389/fmicb.2025.1550783] [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: 01/14/2025] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Pediatric asthma is a common chronic airway inflammatory disease that begins in childhood and its impact persists throughout all age stages of patients. With the continuous progress of detection technologies, numerous studies have firmly demonstrated that gut microbiota and respiratory microbiota are closely related to the occurrence and development of asthma, and related research is increasing day by day. This article elaborates in detail on the characteristics, composition of normal gut microbiota and lung microbiota at different ages and in different sites, as well as the connection of the gut-lung axis. Subsequently, it deeply analyzes various factors influencing microbiota colonization, including host factor, delivery mode, maternal dietary and infant feeding patterns, environmental microbial exposure and pollutants, and the use of antibiotics in early life. These factors are highly likely to play a crucial role in the onset process and disease progression of asthma. Research shows that obvious changes have occurred in the respiratory and gut microbiota of asthma patients, and these microbiomes exhibit different characteristics according to the phenotypes and endotypes of asthma. Finally, the article summarizes the microbiota-related treatment approaches for asthma carried out in recent years, including the application of probiotics, nutritional interventions, and fecal microbiota transplantation. These treatment modalities are expected to become new directions for future asthma treatment and bring new hope for solving the problem of childhood asthma.
Collapse
Affiliation(s)
- Lian Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wenqi Zhao
- School of Clinical Medicine, Qilu Medical University, Zibo, China
| | - Han Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanjie Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi Cheng
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Hu J, Liu J, Wu H, Zhao T, Li Z, Luo J. Differences in the gut and pharyngeal microbiomes before and after treatment of an acute exacerbation of chronic obstructive pulmonary disease. Microb Pathog 2025; 205:107635. [PMID: 40288425 DOI: 10.1016/j.micpath.2025.107635] [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: 04/11/2024] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
This study investigated the gut microbiota and pharyngeal microbiome before and after treatment of an acute exacerbation of chronic obstructive pulmonary disease (AECOPD). The abundance and diversity of microorganisms in the gut and pharynx were examined in 24 patients before and after treatment of AECOPD. Enzyme-linked immunosorbent assay was used to detect inflammatory factors in venous blood and 16S rDNA sequencing was performed. The concentration of short-chain fatty acids (SCFAs) in fecal samples was measured by chromatography-mass spectrometry. The results indicated that the diversity and richness of the gut microbiota decreased post-treatment. The linear discriminant analysis effect size (LEfSe) algorithm revealed an increase in the abundance of f_Dietziaceae, g_Dietzia, g_Megasphaera, g_Robinsoniella, s_Salivarius, and s_Peoriensis in the gut after treatment. There was also a post-treatment decrease in the richness of the pharyngeal microbiome. LEfSe revealed a high abundance of p_Actinobacteria, f_Bacteriodaceae, o_Thermales, g_Bacteroides, and g_Thermus in the pharynx before treatment, and an increased abundance of o_Enterobacterales, f_Enterobacteriaceae, f_Ruminococcaceae, and g_Faecalibacterium after treatment. There were no post-treatment changes in SCFA levels. However, the serum C-reactive protein level decreased after treatment. Levels of other inflammatory factors, including tumor necrosis factor-alpha, interleukin (IL)-1β, IL-6, IL-10, transforming growth factor-beta, IL-23, IL-17, and interferon-gamma, were consistent before and after treatment. In this study, changes in the gut microbiota and pharyngeal microbiome occurred after treatment for AECOPD, with no changes in levels of SCFAs or inflammatory factors, except for a decrease in the C-reactive protein level.
Collapse
Affiliation(s)
- Jiangxia Hu
- Department of Respiratory Medicine, Yongkang Hospital, 2 Qianhuayuan, Shengli Street, JiangJinhua, Yongkang City, Zhejiang Province, 321399, China
| | - Juan Liu
- Department of Respiratory Medicine, Zhejiang Hospital, 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310012, China
| | - Haiyan Wu
- Department of Respiratory Medicine, Zhejiang Hospital, 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310012, China
| | - Tian Zhao
- Department of Respiratory Medicine, Zhejiang Hospital, 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310012, China
| | - Zhijun Li
- Department of Respiratory Medicine, Zhejiang Hospital, 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310012, China.
| | - Jinjian Luo
- Department of Pulmonary and Critical Care Medicine of Anji County People's Hospital, 699 Tianmu Road, Anji County, HuZhou, Zhejiang Province, 313399, China.
| |
Collapse
|
6
|
Ye F, Li L, Wang J, Yang H. Advances in gut-lung axis research: clinical perspectives on pneumonia prevention and treatment. Front Immunol 2025; 16:1576141. [PMID: 40330490 PMCID: PMC12052896 DOI: 10.3389/fimmu.2025.1576141] [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: 02/13/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
In recent years, the study of the interaction between gut microbiota and distant organs such as the heart, lungs, brain, and liver has become a hot topic in the field of gut microbiology. With a deeper understanding of its immune regulation and mechanisms of action, these findings have increasingly highlighted their guiding value in clinical practice. The gut is not only the largest digestive organ in the human body but also the habitat for most microorganisms. Imbalances in gut microbial communities have been associated with various lung diseases, such as allergic asthma and cystic fibrosis. Furthermore, gut microbial communities have significant impacts on metabolic function and immune responses. Their metabolites not only regulate gastrointestinal immune systems but may also affect distant organs such as the lungs and brain. As one of the most common types of respiratory system diseases worldwide, pulmonary infections have high morbidity and mortality rates. Pulmonary infections caused by immune dysfunction can lead to gastrointestinal problems like diarrhea, further resulting in imbalances within complex interactions that are associated with abnormal manifestations under disequilibrium conditions. Meanwhile, clinical interventions can significantly modulate the composition of gut microbiota, and alteration in gut microbiota may subsequently indicate susceptibility to pulmonary infections and even contribute to the prevention or regulation of their progression. This review delves into the interaction between gut microbiota and pulmonary infections, elucidating the latest advancements in gut-lung axis research and providing a fresh perspective for the treatment and prevention of pneumonia.
Collapse
Affiliation(s)
| | | | | | - Hongfeng Yang
- Department of Critical Care Medicine, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
7
|
Zhang F, Luan J, Suo L, Wang H, Zhao Y, Sun T, Ni Y, Cao H, Zou X, Liu B. Altered gut microbiota and metabolite profiles in community-acquired pneumonia: a metagenomic and metabolomic study. Microbiol Spectr 2025; 13:e0263924. [PMID: 40062854 PMCID: PMC11960049 DOI: 10.1128/spectrum.02639-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/08/2025] [Indexed: 04/03/2025] Open
Abstract
Emerging evidence suggests that altered gut microbiota is linked to community-acquired pneumonia (CAP), but the potential mechanisms by which gut microbiota and its metabolites contribute to the development of CAP remain unclear. Fecal samples from 32 CAP patients and 36 healthy controls were analyzed through metagenomic sequencing and metabolomic profiling. The gut microbiota composition in CAP patients showed significant differences and lower diversity compared to healthy controls. Genera involved in short-chain fatty acid (SCFA) production, such as Faecalibacterium, Ruminococcus, and Eubacterium, as well as species like Faecalibacterium prausnitzii, Bifidobacterium adolescentis, Eubacterium rectale, Prevotella copri, and Ruminococcus bromii, were significantly depleted in CAP patients. Bacterial co-occurrence network analysis revealed an over-representation of pro-inflammatory bacteria, which contributed to the core gut microbiome in CAP patients. Metabolomic analysis of fecal samples identified a distinct metabolic profile, with a notable increase in arachidonic acid, but a decrease in secondary bile acids, such as deoxycholic acid, lithocholic acid, and ursodeoxycholic acid, compared to healthy controls. Spearman correlation analysis between differential microbiota and bile acids showed that Faecalibacterium prausnitzii, Bifidobacterium adolescentis, Eubacterium rectale, and Prevotella copri were positively correlated with ursocholic acid, lithocholic acid, and ursodeoxycholic acid, respectively. Our results suggest that the reduction in secondary bile acids, insufficient production of SCFAs, and an overabundance of pro-inflammatory bacteria may contribute to metabolic inflammation in the body. These factors could play a key role in the pathogenesis of CAP, driven by gut microbiota alterations. IMPORTANCE This study presents a comprehensive metagenomic and metabolomic analysis of fecal samples from community-acquired pneumonia (CAP) patients, identifying key characteristics, such as decreased secondary bile acids, imbalanced short-chain fatty acid production, and increased pro-inflammatory bacteria. These findings provide valuable insights into the mechanisms linking gut microbiota alterations to CAP pathogenesis and suggest that targeting the gut microbiota could be a promising strategy for intervening in CAP.
Collapse
Affiliation(s)
- Fuxin Zhang
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
| | - Jiahui Luan
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
| | - Lijun Suo
- Department of Pulmonary and Critical Care Medicine, Zibo Municipal Hospital, Zibo, China
| | - Haiyan Wang
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
| | - Yi Zhao
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
| | - Tianyu Sun
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
| | - Yawen Ni
- National Center for Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Changping Laboratory, Beijing, China
| | - Hongyun Cao
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
| | - Xiaohui Zou
- National Center for Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Bo Liu
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
- Department of Pulmonary and Critical Care Medicine, Zibo Municipal Hospital, Zibo, China
- Department of Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
- Department of Pulmonary and Critical Care Medicine, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
8
|
Simon P, Török É, Szalontai K, Kari B, Neuperger P, Zavala N, Kanizsai I, Puskás LG, Török S, Szebeni GJ. Nutritional Support of Chronic Obstructive Pulmonary Disease. Nutrients 2025; 17:1149. [PMID: 40218907 PMCID: PMC11990120 DOI: 10.3390/nu17071149] [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: 02/05/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background: COPD is a heterogenous disease of the respiratory tract caused by diverse genetic factors along with environmental and lifestyle-related effects such as industrial dust inhalation and, most frequently, cigarette smoking. These factors lead to airflow obstruction and chronic respiratory symptoms. Additionally, the increased risk of infections exacerbates airway inflammation in COPD patients. As a consequence of the complex pathomechanisms and difficulty in treatment, COPD is among the leading causes of mortality both in the western countries and in the developing world. Results: The management of COPD is still a challenge for the clinicians; however, alternative interventions such as smoking cessation and lifestyle changes from a sedentary life to moderate physical activity with special attention to the diet may ameliorate patients' health. Here, we reviewed the effects of different dietary components and supplements on the conditions of COPD. Conclusions: COPD patients are continuously exposed to heavy metals, which are commonly present in cigarette smoke and polluted air. Meanwhile, they often experience significant nutrient deficiencies, which affect the detoxification of these toxic metals. This in turn can further disrupt nutritional balance by interfering with the absorption, metabolism, and utilization of essential micronutrients. Therefore, awareness and deliberate efforts should be made to check levels of micronutrients, with special attention to ensuring adequate levels of antioxidants, vitamin D, vitamin K2, magnesium, and iron, as these may be particularly important in reducing the risk of COPD development and limiting disease severity.
Collapse
Grants
- 2023-1.1.1-PIACI_FÓKUSZ-2024-00036 National Research, Development, and Innovation Office (NKFI), Hungary
- 2020-1.1.6-JÖVŐ-2021-00003 National Research, Development, and Innovation Office (NKFI), Hungary
- 2022-1.2.6-TÉT-IPARI-TR-2022-00023 National Research, Development, and Innovation Office (NKFI), Hungary
- 142877 FK22 National Research, Development, and Innovation Office (NKFI), Hungary.
- BO/00582/22/8 János Bolyai Research Scholarship of the Hungarian Academy of Sciences
Collapse
Affiliation(s)
- Péter Simon
- National Korányi Institute of Pulmonology, 1121 Budapest, Hungary;
| | - Éva Török
- Gastroenterology Center Buda, 1117 Budapest, Hungary;
| | - Klára Szalontai
- Department of Pulmonology, Szent-Györgyi Albert Medical Center, University of Szeged, 6772 Deszk, Hungary;
| | - Beáta Kari
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | - Patrícia Neuperger
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | - Norma Zavala
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
| | | | - László G. Puskás
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
- Anthelos Ltd., 6726 Szeged, Hungary
| | - Szilvia Török
- National Korányi Institute of Pulmonology, 1121 Budapest, Hungary;
| | - Gabor J. Szebeni
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.K.); (P.N.); (N.Z.); (L.G.P.)
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
| |
Collapse
|
9
|
Khan M, Alteneder M, Reiter W, Krausgruber T, Dobnikar L, Madern M, Waldherr M, Bock C, Hartl M, Ellmeier W, Henriksson J, Boucheron N. Single-cell and chromatin accessibility profiling reveals regulatory programs of pathogenic Th2 cells in allergic asthma. Nat Commun 2025; 16:2565. [PMID: 40089475 PMCID: PMC11910648 DOI: 10.1038/s41467-025-57590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Lung pathogenic T helper type 2 (pTh2) cells are important in mediating allergic asthma, but fundamental questions remain regarding their heterogeneity and epigenetic regulation. Here we investigate immune regulation in allergic asthma by single-cell RNA sequencing in mice challenged with house dust mite, in the presence and absence of histone deacetylase 1 (HDAC1) function. Our analyses indicate two distinct highly proinflammatory subsets of lung pTh2 cells and pinpoint thymic stromal lymphopoietin (TSLP) and Tumour Necrosis Factor Receptor Superfamily (TNFRSF) members as important drivers to generate pTh2 cells in vitro. Using our in vitro model, we uncover how signalling via TSLP and a TNFRSF member shapes chromatin accessibility at the type 2 cytokine gene loci by modulating HDAC1 repressive function. In summary, we have generated insights into pTh2 cell biology and establish an in vitro model for investigating pTh2 cells that proves useful for discovering molecular mechanisms involved in pTh2-mediated allergic asthma.
Collapse
Affiliation(s)
- Matarr Khan
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Marlis Alteneder
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Wolfgang Reiter
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Lina Dobnikar
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Moritz Madern
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Monika Waldherr
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
- FH Campus Wien, Department of Applied Life Sciences/Bioengineering/Bioinformatics, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Markus Hartl
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Johan Henriksson
- Umeå University, Umeå Centre for Microbial Research (UCMR), Integrated Science Lab (Icelab), Department of Molecular Biology, Umeå, Sweden
| | - Nicole Boucheron
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria.
| |
Collapse
|
10
|
Huang YJ. The Microbiome in Asthma Heterogeneity: The Role of Multi-Omic Investigations. Immunol Rev 2025; 330:e70015. [PMID: 40072031 PMCID: PMC11899502 DOI: 10.1111/imr.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/13/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Asthma is one of the most prevalent and extensively studied chronic respiratory conditions, yet the heterogeneity of asthma remains biologically puzzling. Established factors like exogenous exposures and treatment adherence contribute to variability in asthma risk and clinical outcomes. It is also clear that the endogenous factors of genetics and immune system response patterns play key roles in asthma. Despite significant existing knowledge in the above, divergent clinical trajectories and outcomes are still observed, even among individuals with similar risk profiles, biomarkers, and optimal medical management. This suggests uncaptured biological interactions that contribute to asthma's heterogeneity, for which the role of host microbiota has lately attracted much research attention. This review will highlight recent evidence in this area, focusing on bedside-to-bench investigations that have leveraged omic technologies to uncover microbiome links to asthma outcomes and immunobiology. Studies centered on the respiratory system and the use of multi-omics are noted in particular. These represent a new generation of reverse-translational investigations revealing potential functional crosstalk in host microbiomes that may drive phenotypic heterogeneity in chronic diseases like asthma. Multi-omic data offer a wide lens into ecosystem interactions within a host. This informs new hypotheses and experimental work to elucidate mechanistic pathways for unresolved asthma endotypes. Further incorporation of multi-omics into patient-centered investigations can yield new insights that hopefully lead to even more precise, microbiome-informed strategies to reduce asthma burden.
Collapse
Affiliation(s)
- Yvonne J. Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
11
|
Koceva H, Amiratashani M, Akbarimoghaddam P, Hoffmann B, Zhurgenbayeva G, Gresnigt MS, Marcelino VR, Eggeling C, Figge MT, Amorim MJ, Mosig AS. Deciphering respiratory viral infections by harnessing organ-on-chip technology to explore the gut-lung axis. Open Biol 2025; 15:240231. [PMID: 40037530 PMCID: PMC11879621 DOI: 10.1098/rsob.240231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
The lung microbiome has recently gained attention for potentially affecting respiratory viral infections, including influenza A virus, respiratory syncytial virus (RSV) and SARS-CoV-2. We will discuss the complexities of the lung microenvironment in the context of viral infections and the use of organ-on-chip (OoC) models in replicating the respiratory tract milieu to aid in understanding the role of temporary microbial colonization. Leveraging the innovative capabilities of OoC, particularly through integrating gut and lung models, opens new avenues to understand the mechanisms linking inter-organ crosstalk and respiratory infections. We will discuss technical aspects of OoC lung models, ranging from the selection of cell substrates for extracellular matrix mimicry, mechanical strain, breathing mechanisms and air-liquid interface to the integration of immune cells and use of microscopy tools for algorithm-based image analysis and systems biology to study viral infection in vitro. OoC offers exciting new options to study viral infections across host species and to investigate human cellular physiology at a personalized level. This review bridges the gap between complex biological phenomena and the technical prowess of OoC models, providing a comprehensive roadmap for researchers in the field.
Collapse
Affiliation(s)
- Hristina Koceva
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
| | - Mona Amiratashani
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Parastoo Akbarimoghaddam
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Bianca Hoffmann
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Gaukhar Zhurgenbayeva
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Leibniz Institute of Photonic Technologies e.V., Member of the Leibniz Centre for Photonics in Infection Research (LPI), Jena, Germany
| | - Mark S. Gresnigt
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Vanessa Rossetto Marcelino
- Melbourne Integrative Genomics, School of BioSciences, University of Melbourne, Parkville, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, Australia
| | - Christian Eggeling
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Leibniz Institute of Photonic Technologies e.V., Member of the Leibniz Centre for Photonics in Infection Research (LPI), Jena, Germany
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
- Jena Center for Soft Matter, Jena, Germany
| | - Marc Thilo Figge
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Maria-João Amorim
- Católica Biomédical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Alexander S. Mosig
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Jena Center for Soft Matter, Jena, Germany
- Center of Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
12
|
Kim YJ, Ihrie VM, Shi P, Ihrie MD, Womble JT, Meares AH, Granek JA, Gunsch CK, Ingram JL. Glucagon-Like Peptide 1 Receptor ( Glp1r) Deficiency Does Not Appreciably Alter Airway Inflammation or Gut-Lung Microbiome Axis in a Mouse Model of Obese Allergic Airways Disease and Bariatric Surgery. J Asthma Allergy 2025; 18:285-305. [PMID: 40046174 PMCID: PMC11880686 DOI: 10.2147/jaa.s478329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/22/2025] [Indexed: 03/09/2025] Open
Abstract
Purpose High body mass index (≥30 kg/m2) is associated with asthma severity, and nearly 40% of asthma patients exhibit obesity. Furthermore, over 40% of patients with obesity and asthma that receive bariatric surgery no longer require asthma medication. Increased levels of glucagon-like peptide 1 (GLP-1) occur after bariatric surgery, and recent studies suggest that GLP-1 receptor (GLP-1R) signaling may regulate the gut microbiome and have anti-inflammatory properties in the lung. Thus, we hypothesized that increased GLP-1R signaling following metabolic surgery in obese and allergen-challenged mice leads to gut/lung microbiome alterations, which together contribute to improved features of allergic airways disease. Methods Male and female Glp1r-deficient (Glp1r-/- ) and replete (Glp1r+/+) mice were administered high fat diet (HFD) to induce obesity with simultaneous intranasal challenge with house dust mite (HDM) allergen to model allergic airway disease with appropriate controls. Mice on HFD received either no surgery, sham surgery, or vertical sleeve gastrectomy (VSG) on week 10 and were sacrificed on week 13. Data were collected with regard to fecal and lung tissue microbiome, lung histology, metabolic markers, and respiratory inflammation. Results HFD led to metabolic imbalance characterized by lower GLP-1 and higher leptin levels, increased glucose intolerance, and alterations in gut microbiome composition. Prevalence of bacteria associated with short chain fatty acid (SCFA) production, namely Bifidobacterium, Lachnospiraceae UCG-001, and Parasutterella, was reduced in mice fed HFD and positively associated with serum GLP-1 levels. Intranasal HDM exposure induced airway inflammation. While Glp1r-/- genotype affected fecal microbiome beta diversity metrics, its effect was limited. Conclusion Herein, GLP-1R deficiency had surprisingly little effect on host gut and lung microbiomes and health, despite recent studies suggesting that GLP-1 receptor agonists are protective against lung inflammation.
Collapse
Affiliation(s)
- Yeon Ji Kim
- Department of Civil and Environmental Engineering, Pratt School of Engineering, DukeUniversity, Durham, NC, USA
| | - Victoria M Ihrie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Pixu Shi
- Biostatistics and Bioinformatics, Division of Integrative Genomics, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Mark D Ihrie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jack T Womble
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Anna Hill Meares
- Department of Civil and Environmental Engineering, Pratt School of Engineering, DukeUniversity, Durham, NC, USA
| | - Joshua A Granek
- Biostatistics and Bioinformatics, Division of Integrative Genomics, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Claudia K Gunsch
- Department of Civil and Environmental Engineering, Pratt School of Engineering, DukeUniversity, Durham, NC, USA
| | - Jennifer L Ingram
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
13
|
Zheng J, Huang Y, Zhang L, Liu T, Zou Y, He L, Guo S. Role of the Gut-Lung Microbiome Axis in Airway Inflammation in OVA-Challenged Mice and the Effect of Azithromycin. J Inflamm Res 2025; 18:2661-2676. [PMID: 40008084 PMCID: PMC11853874 DOI: 10.2147/jir.s506688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Objective This study aimed to investigate the role of the gut-lung microbiome axis in airway inflammation in asthma and to evaluate the effect of azithromycin on this axis, with a focus on the potential mechanism by which azithromycin reduces allergic airway inflammation. Methods Haematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining were used to assess pathological changes in the lung tissues of asthmatic mice. Leukocyte cell types in bronchoalveolar lavage fluid (BALF) samples were quantified following Wright-Giemsa staining. Total IgE, OVA-specific IgE, IL-4, IL-6, and IL-17A levels in BALF and total IgE in serum were measured by ELISA. The respiratory and gut microbiota were analysed using 16S rRNA gene sequencing and subsequent taxonomic analysis. Results OVA-challenged asthmatic mice with gut microbiota dysbiosis exhibited alterations in the respiratory microbiota, resulting in further aggravation of airway inflammation. Following faecal microbiota transplantation (FMT) to restore gut microbiota, respiratory microbiota dysbiosis was partially improved, and airway inflammation was significantly alleviated. Furthermore, azithromycin reduced airway inflammation in asthmatic mice, particularly non-eosinophilic inflammation, for which low-dose azithromycin combined with budesonide proved more effective. Azithromycin significantly enhanced the diversity and microbial composition of the gut microbiota and also affected the respiratory microbiota. At the phylum level, azithromycin decreased the abundance of Proteobacteria in the gut microbiota. At the genus level, azithromycin reduced the abundance of Pseudomonas in the respiratory microbiota. Conclusion The gut-lung microbiome axis plays a crucial role in airway inflammation in asthma. Azithromycin may reduce airway inflammation in asthma through modulation of the gut-lung microbiome axis.
Collapse
Affiliation(s)
- Jun Zheng
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yuying Huang
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Liang Zhang
- Department of Endocrine, Genetics and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Tiantian Liu
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ya Zou
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Li He
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Sheng Guo
- Department of Endocrine, Genetics and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
14
|
Chakravarty K, Gaur S, Kumar R, Jha NK, Gupta PK. Exploring the Multifaceted Therapeutic Potential of Probiotics: A Review of Current Insights and Applications. Probiotics Antimicrob Proteins 2025; 17:341-363. [PMID: 39069588 DOI: 10.1007/s12602-024-10328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
The interplay between human health and the microbiome has gained extensive attention, with probiotics emerging as pivotal therapeutic agents due to their vast potential in treating various health issues. As significant modulators of the gut microbiota, probiotics are crucial in maintaining intestinal homeostasis and enhancing the synthesis of short-chain fatty acids. Despite extensive research over the past decades, there remains an urgent need for a comprehensive and detailed review that encapsulates probiotics' latest insights and applications. This review focusses on the multifaceted roles of probiotics in promoting health and preventing disease, highlighting the complex mechanisms through which these beneficial bacteria influence both gut flora and the human body at large. This paper also explores probiotics' neurological and gastrointestinal applications, focussing on their significant impact on the gut-brain axis and their therapeutic potential in a broad spectrum of pathological conditions. Current innovations in probiotic formulations, mainly focusing on integrating genomics and biotechnological advancements, have also been comprehensively discussed herein. This paper also critically examines the regulatory landscape that governs probiotic use, ensuring safety and efficacy in clinical and dietary settings. By presenting a comprehensive overview of recent studies and emerging trends, this review aims to illuminate probiotics' extensive therapeutic capabilities, leading to future research and clinical applications. However, besides extensive research, further advanced explorations into probiotic interactions and mechanisms will be essential for developing more targeted and effective therapeutic strategies, potentially revolutionizing health care practices for consumers.
Collapse
Affiliation(s)
- Kashyapi Chakravarty
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India.
| | - Rohit Kumar
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Niraj Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 602105, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, 140401, India
| | - Piyush Kumar Gupta
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, 248002, India.
| |
Collapse
|
15
|
Crabtree D, Seidler K, Barrow M. Pathophysiological mechanisms of gut dysbiosis and food allergy and an investigation of probiotics as an intervention for atopic disease. Clin Nutr ESPEN 2025; 65:189-204. [PMID: 39571752 DOI: 10.1016/j.clnesp.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND AND AIMS Epidemiological studies have associated reduced bacterial diversity and abundance and food allergy. This mechanistic review investigated the link between gut dysbiosis and food allergy with a focus on the role of short-chain fatty acids (SCFAs) in modulating T-cells. T-cell differentiation poses an opportunity to direct the immune cells towards an anergic regulatory T cell (Treg) or allergic T helper 2 (Th2) response. Probiotic intervention to prevent and/or treat atopic disease symptoms through this mechanistic pathway was explored. METHODOLOGY A narrative review was conducted following a three-stage systematic literature search of EMBASE and Medline databases. Ninety-six of 571 papers were accepted and critically appraised using ARRIVE and SIGN50 forms. Thematic analysis identified key pathophysiological mechanisms within the narrative of included papers. RESULTS Preclinical studies provided compelling evidence for SCFAs' modulation of T-cell differentiation, which may act through G-protein coupled receptors 41, 43 and 109a and histone deacetylase inhibition. Foxp3 transcription factor was implicated in the upregulation of Tregs. Human probiotic intervention studies aimed at increasing SCFAs and Tregs and preventing atopic disease showed inconclusive results. However, evidence for probiotic intervention in children with cow's milk protein allergy (CMPA) was more promising and warrants further investigation. CONCLUSION Preclinical evidence suggests that the mechanism of gut dysbiosis and reduced SCFAs may skew T-cell differentiation towards a Th2 response, thus inducing allergy symptoms. Probiotic trials were inconclusive: probiotics were predominantly unsuccessful in the prevention of allergic disease, however, may be able to modulate food allergy symptoms in infants with CMPA.
Collapse
Affiliation(s)
- Danielle Crabtree
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| | - Karin Seidler
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| | - Michelle Barrow
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| |
Collapse
|
16
|
Hoskinson C, Petersen C, Turvey SE. How the early life microbiome shapes immune programming in childhood asthma and allergies. Mucosal Immunol 2025; 18:26-35. [PMID: 39675725 DOI: 10.1016/j.mucimm.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
Despite advances in our understanding of their diagnosis and treatment, pediatric allergies impose substantial burdens on affected children, families, and healthcare systems. Further, the prevalence of allergic diseases has dramatically increased over the past half-century, leading to additional concerns and concerted efforts to identify the origins, potential predictors and preventions, and therapies of allergic diseases. Together with the increase in allergic diseases, changes in lifestyle and early-life environmental influences have corresponded with changes in colonization patterns of the infant gut microbiome. The gut microbiome plays a key role in developing the immune system, thus greatly influencing the development of allergic disease. In this review, we specifically highlight the importance of the proper maturation and composition of the gut microbiome as an essential step in healthy child development or disease progression. By exploring the intertwined development of the immune system and microbiome across pediatric allergic diseases, we provide insights into potential novel strategies for their prevention and management.
Collapse
Affiliation(s)
- Courtney Hoskinson
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Charisse Petersen
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
17
|
Zhu H, Wu C, Wu H, Liu J, Ye W, Zhao T, Li Z. The gut microbiota-SCFA-inflammation axis in patients with AECOPD. PLoS One 2025; 20:e0312606. [PMID: 39787116 PMCID: PMC11717293 DOI: 10.1371/journal.pone.0312606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/09/2024] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVES The aim of the study was to explore the alteration of microbiota and SCFA in gut and inflammation in acute exacerbation chronic obstructive pulmonary disease (AECOPD) patients, and to test the hypothesis that a disorder of gut microbiota will lead to the alteration of SCFA, which will aggravate inflammation in AECOPD patients. METHODS AND RESULTS 24 patients with AECOPD and 18 healthy volunteers were included in the study. Gut microbiota were analyzed by 16S rDNA and serum was used to detect levels of inflammatory factors by ELISA. Fatty acid concentrations were determined in lumen via gas chromatography-mass spectrometry. The richness and diversity of gut microbiota were decreased in AECOPD patients. β-diversity analysis revealed differences between AECOPD patients and healthy controls. p_Bacteroidetes, g_Paraprevotella, g_Ruminococcus2, g_Parasutterella, o_Rhodospirillales, and g_Romboutsia in the healthy controls and p_Firmicutes, o_Actinomycetales, f_Actinomycetadeae, g_Actinomyces, g_Mogibacterium, f_Veillonellaceae, f_Enterococcaceae, and g_Enterococcus in AECOPD patients were the most abundant microbiota. SCFA levels were decreased in patients with AECOPD. In addition, the results demonstrated that except for a reduction in IL-6, there was no change in inflammatory markers in AECOPD patients. CONCLUSION In AECOPD patients, the gut microbiota-SCFA-inflammation axis is augmented, with decreased diversity and abundance of gut microbiota, leading to a reduction in SCFA and an imbalance of inflammation.
Collapse
Affiliation(s)
- Hengjing Zhu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Chen Wu
- Department of Respiratory Medicine in Jiashan County Second People’s Hospital, Jiaxing, Zhejiang Province, China
| | - Haiyan Wu
- Department of Respiratory Medicine in Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Juan Liu
- Department of Respiratory Medicine in Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Wu Ye
- Department of Respiratory Medicine in Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Tian Zhao
- Department of Respiratory Medicine in Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Zhijun Li
- Department of Respiratory Medicine in Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
18
|
Lai Y, Qiu R, Zhou J, Ren L, Qu Y, Zhang G. Fecal Microbiota Transplantation Alleviates Airway Inflammation in Asthmatic Rats by Increasing the Level of Short-Chain Fatty Acids in the Intestine. Inflammation 2025:10.1007/s10753-024-02233-w. [PMID: 39775370 DOI: 10.1007/s10753-024-02233-w] [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: 10/12/2024] [Revised: 11/22/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025]
Abstract
Asthma is a prevalent chronic inflammatory disorder of the respiratory tract that not only manifests with respiratory symptoms but also often involves intestinal flora disorders and gastrointestinal dysfunction. Recent studies have confirmed the close relationship between the gut and lungs, known as the "gut-lung axis" theory. Fecal microbiota transplantation (FMT), a method for restoring normal intestinal flora, has shown promise in treating common gastrointestinal diseases. The "gut-lung axis" theory suggests that FMT may have significant therapeutic potential for asthma. In this study, we established an Ovalbumin (OVA)-induced rat model of asthma to investigate the protective effect of FMT on airway inflammation and the restoration of intestinal short-chain fatty acids (SCFAs), aiming to explore its underlying mechanism. Rats in the Control group underwent fecal treatment via gavage (Control-FMT, C-FMT group), while rats in the Asthma group underwent fecal treatment via gavage after asthma induction (Asthma-FMT, A-FMT group). Following a two-week period of continuous intragastric administration, various measurements were conducted to assess pulmonary function, peripheral blood neutrophil, lymphocyte, and eosinophil content, lung tissue pathology, and collagen fiber deposition in the lungs. Additionally, neutrophil and eosinophil content in bronchoalveolar lavage fluid (BALF), expression levels of Interleukin-4 (IL-4), IL-5, IL-13, IL-17, IL-33, leukotrienes (LT), thymic stromal lymphopoietin (TSLP), prostaglandin D2 (PGD2) protein and mRNA in lung tissue, and SCFAs content in stool were evaluated. In the C-FMT group, lung function significantly improved, inflammatory cell content in peripheral blood and BALF decreased, lung tissue pathology and collagen fiber deposition significantly improved, the protein and mRNA levels of lung inflammatory factors IL-4, IL-5, IL-13, IL-17, IL-33, LT, TSLP, PGD2 were significantly decreased, and SCFAs such as acetate (C2), propionate (C3), butyrate (C4), isobutyric acid (I-C4), valeric acid (C5), and isovaleric acid (I-C5) content in stool significantly increased. However, the indexes in the A-FMT group did not show significant recovery, and the treatment effect on asthma symptoms in rats was inferior to that in the C-FMT group. Asthma induced intestinal flora disorders in rats, and FMT treatment improved the inflammatory response in asthmatic rat models and corrected their intestinal SCFAs disorders. Encouraging the recovery of intestinal SCFAs may play a significant role, and beneficial bacteria present in feces may improve asthma symptoms by promoting the remodeling of intestinal flora. This experiment provides further scientific evidence supporting the "gut-lung axis" theory.
Collapse
Affiliation(s)
- Yitian Lai
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ranran Qiu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Jingying Zhou
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ling Ren
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yizhuo Qu
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Guoshan Zhang
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
19
|
Rowland SN, Green CG, Halliwill JR, Singanayagam A, Heaney LM. Gut feelings on short-chain fatty acids to regulate respiratory health. Trends Endocrinol Metab 2025:S1043-2760(24)00329-1. [PMID: 39757060 DOI: 10.1016/j.tem.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025]
Abstract
Respiratory infections and diseases pose significant challenges to society and healthcare systems, underscoring the need for preventative and therapeutic strategies. Recent research in rodent models indicates that short-chain fatty acids (SCFAs), metabolites produced by gut bacteria, may offer medicinal benefits for respiratory conditions. In this opinion, we summarize the current literature that highlights the potential of SCFAs to enhance immune balance in humans. SCFAs have demonstrated the potential to decrease the risk of primary and secondary respiratory infections, modulate allergic airway exacerbations, and improve overall epithelial pathogen defenses. Therefore, we suggest that systemic SCFA levels could be targeted to support gut and respiratory health in specific groups, such as patients in hospital, women and their offspring, children, older adults, and athletes/military personnel.
Collapse
Affiliation(s)
- Samantha N Rowland
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Christopher G Green
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - John R Halliwill
- Department of Human Physiology, University of Oregon, Eugene, OR, USA
| | - Aran Singanayagam
- Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| |
Collapse
|
20
|
Pan J, Zhang X, Shi D, Tian X, Xu L, Lu X, Dong M, Yao P, Pan Z, Ling Z, Wu N, Yao H. Short-chain fatty acids play a key role in antibody response to SARS-CoV-2 infection in people living with HIV. Sci Rep 2024; 14:31211. [PMID: 39732792 DOI: 10.1038/s41598-024-82596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
High SARS-CoV-2-specific antibody levels can protect against SARS-CoV-2 reinfection. The gut microbiome can affect a host's immune response. However, its role in the antibody response to SARS-CoV-2 in people living with HIV (PLWH) remains poorly understood. Here, we categorised PLWH and healthy individuals into high- and low-antibody-response groups. Shotgun metagenomic sequencing and targeted metabolomic assays were used to investigate the differences in the gut microbiome and metabolic functions between the high- and low-antibody-response groups. PLWH demonstrated a higher abundance of short-chain fatty acid (SCFA)-producing species, accompanied by high serum levels of several SCFAs, in the high-antibody-response group than in the low-antibody-response group. In contrast, healthy individuals demonstrated higher enrichment of pilus-bearing bacterial species, with flagella-expressing genes, in the high-antibody-response group than in the low-antibody-response group. Therefore, gut-microbiota-derived SCFAs play a key role in antibody responses in PLWH but not in healthy individuals. Our results afford a novel understanding of how the gut microbiome and its metabolites are associated with host immunity. Moreover, they may facilitate the exploration of modalities to prevent SARS-CoV-2 reinfection through various gut-microbiota-targeted interventions tailored to different populations.
Collapse
Affiliation(s)
- Jingying Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Xiaodi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Danrong Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Xuebin Tian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Lijun Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Xiangyun Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Mingqing Dong
- Department of Infectious Disease, Zhejiang Qingchun Hospital, Zhejiang University, Hangzhou, 310000, China
| | - Peng Yao
- Department of Infectious Disease, Zhejiang Qingchun Hospital, Zhejiang University, Hangzhou, 310000, China
| | - Zhaoyi Pan
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China.
| |
Collapse
|
21
|
Wang J, Hou Y, Mu L, Yang M, Ai X. Gut microbiota contributes to the intestinal and extraintestinal immune homeostasis by balancing Th17/Treg cells. Int Immunopharmacol 2024; 143:113570. [PMID: 39547012 DOI: 10.1016/j.intimp.2024.113570] [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: 08/09/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Gut microbiota is generally considered to play an important role in host health due to its extensive immunomodulatory activities. Th17 and Treg cells are two important CD4+ T cell subsets involved in immune regulation, and their imbalance is closely tied to many immune diseases. Recently, abundant researches have highlighted the importance of gut microbiota in supporting intestinal and extraintestinal immunity through the balance of Th17 and Treg cells. Here, we presented a comprehensive review of these findings. This review first provided an overview of gut microbiota, along with Th17/Treg cell differentiation and cytokine production. Subsequently, the review summarized the regulatory effects of gut microbiota (in terms of species, components, and metabolites) on the Th17/Treg cell balance in the local intestines and extraintestinal organs, such as lung, liver, brain, kidney, and bone. Specifically, the Th17 and Treg cells that can be modulated by gut microbiota originate not only from the gut and extraintestinal organs, but also from peripheral blood and spleen. Then, the microbial therapeutics, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT), were also reviewed because of their therapeutic potentials in addressing intestinal and extraintestinal diseases via the Th17/Treg axis. Finally, the review discussed the clinical applications and future study prospects of microbial therapeutics by targeting the Th17/Treg cell balance. In conclusion, this review focused on elucidating the regulatory effects of gut microbiota in balancing Th17/Treg cells to maintain intestinal and extraintestinal immune homeostasis, contributing to the further development and promotion of microbial therapeutics.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yaqin Hou
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Lifeng Mu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
22
|
Bhutta NK, Xu X, Jian C, Wang Y, Liu Y, Sun J, Han B, Wu S, Javeed A. Gut microbiota mediated T cells regulation and autoimmune diseases. Front Microbiol 2024; 15:1477187. [PMID: 39749132 PMCID: PMC11694513 DOI: 10.3389/fmicb.2024.1477187] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Gut microbiota regulates the immune system, the development and progression of autoimmune diseases (AIDs) and overall health. Recent studies have played a crucial part in understanding the specific role of different gut bacterial strains and their metabolites in different AIDs. Microbial signatures in AIDs are revealed by advanced sequencing and metabolomics studies. Microbes such as Faecalibacterium prausnitzii, Akkermansia muciniphila, Anaerostipes caccae, Bacteroides sp., Roseburia sp., Blautia sp., Blautia faecis, Clostridium lavalense, Christensenellaceae sp., Coprococcus sp., Firmicutes sp., Ruminococcaceae sp., Lachnospiraceae sp., Megamonas sp., Monoglobus sp., Streptococcus pneumoniae and Bifidobacterium sp. help maintain immune homeostasis; whereas, Prevotella copri, Ruminococcus gnavus, Lactobacillus salivarius, Enterococcus gallinarum, Elizabeth menigoseptica, Collinsella sp., Escherichia sp., Fusobacterium sp., Enterobacter ludwigii, Enterobacteriaceae sp., Proteobacteria, Porphyromonas gingivalis, Porphyromonas nigrescens, Dorea sp., and Clostridium sp. cause immuno-pathogenesis. A complex web of interactions is revealed by understanding the influence of gut microbiota on immune cells and various T cell subsets such as CD4+ T cells, CD8+ T cells, natural killer T cells, γδ T cells, etc. Certain AIDs, including rheumatoid arthritis, diabetes mellitus, atopic asthma, inflammatory bowel disease and non-alcoholic fatty liver disease exhibit a state of dysbiosis, characterized by alterations in microbial diversity and relative abundance of specific taxa. This review summarizes recent developments in understanding the role of certain microbiota composition in specific AIDs, and the factors affecting specific regulatory T cells through certain microbial metabolites and also focuses the potential application and therapeutic significance of gut microbiota-based interventions as novel adjunctive therapies for AIDs. Further research to determine the precise association of each gut bacterial strain in specific diseases is required.
Collapse
Affiliation(s)
- Nabeel Khalid Bhutta
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiujin Xu
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Cuiqin Jian
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yifan Wang
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yi Liu
- Hangzhou Zheda Dixun Biological Gene Engineering Co., Ltd., Hangzhou, China
| | - Jinlyu Sun
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, Department of Allergy, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bingnan Han
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Shandong Wu
- Hangzhou Zheda Dixun Biological Gene Engineering Co., Ltd., Hangzhou, China
| | - Ansar Javeed
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
23
|
Sun Y, Wang Y, Yang Z, Han X, Zhang Y, Chen L, Huo J, Wu R, Wang W, Wang N. Neutral Polysaccharide from Platycodonis Radix-Ameliorated PM 2.5-Induced Lung Injury by Inhibiting the TLR4/NF-κB p65 Pathway and Regulating the Lung and Gut Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27923-27938. [PMID: 39626068 DOI: 10.1021/acs.jafc.4c07319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Platycodonis radix (PR) has been reported to play a protective role in lung injury. However, much less is known about the protective effect and mechanism of its main component PR polysaccharides (PRPs) in particulate matter (PM2.5)-induced lung injury. Here, a neutral polysaccharide (MW: 244.56 kDa) was isolated from PR, mainly composed of Rha, Ara, Gal, Glc, Xyl, and Man. PRPs significantly improved PM2.5-induced pulmonary edema, oxidative damage, and cell apoptosis and downregulated inflammatory factor levels in bronchoalveolar lavage fluid. Mechanistically, PRPs reduced intestinal mucosal barrier damage, thereby lowering serum lipopolysaccharide levels and inhibiting the overactivation of the TLR4/NF-κB signaling pathway in the lung tissue. Notably, PRPs could optimize the composition of pulmonary and intestinal microbiota. Oral administration of PRPs resulted in enrichment of short-chain fatty acid (SCFA)-producing bacteria, thereby upregulating the levels of acetate, butyrate, and isovalerate. Taken together, PRPs have great potential in preventing and repairing the lung injury caused by PM2.5.
Collapse
Affiliation(s)
- Yang Sun
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yanchun Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zaiming Yang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xianlei Han
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yue Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Liyan Chen
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, China
| | - Jinhai Huo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Weiming Wang
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
24
|
Lin H, Perkins NJ, Nkoy F, Stanford JB, Schliep KC, Peddada SD. A Study of Short-Chain Fatty Acids During the Canalicular and Early Saccular Phases of Fetal Lung Development and Childhood Asthma. Genes (Basel) 2024; 15:1595. [PMID: 39766862 PMCID: PMC11675564 DOI: 10.3390/genes15121595] [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: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Emerging literature indicates that the microbiome and its byproducts, such as short-chain fatty acids (SCFAs), play an important role in childhood diseases such as allergies and asthma. Specifically, there is evidence suggesting that SCFAs play a critical role in fetal immunoprogramming during the late saccular phase of fetal lung development. An increase in acetate during the late saccular phase is known to play a critical role in inhibiting histone deacetylases (HDACs), resulting in a cascade of events, including Treg immune regulation, involved in fetal immunoprogramming, and reduction in the asthma phenotype. However, it is not known whether changes in SCFA levels, especially acetate, occurred during the canalicular or early saccular phase among pregnant women whose children did not develop asthma. METHODS In this research, we investigated this question using plasma samples obtained from mothers during the 20th and 28th weeks of pregnancy. Mothers whose children developed asthma were categorized as cases, while those whose children did not were categorized as controls. The specimens were assayed for a panel of SCFAs consisting of acetate, propionate, butyrate, valerate, isobutyrate, and isovalerate. RESULTS The resulting data indicated no significant differences between the cases and controls, either at week 20 or week 28, in any of the SCFAs measured, despite the vascularization during these phases. CONCLUSIONS We did not find differences in measured SCFAs at week 20 or at week 28. A larger prospective study covering multiple time points is necessary to confirm the findings of this preliminary study. Such a study, together with the published literature regarding later time points, may help discover critical windows during pregnancy when simple manipulation of diet will result in healthier outcomes for infants.
Collapse
Affiliation(s)
- Huang Lin
- Department of Epidemiology and Biostatistics, University of Maryland, College Park, MD 20742, USA;
| | - Neil J. Perkins
- Biostatistics and Bioinformatics Branch (BBB), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD 20817, USA;
| | - Flory Nkoy
- Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA;
| | - Joseph B. Stanford
- Division of Public Health, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA; (J.B.S.); (K.C.S.)
| | - Karen C. Schliep
- Division of Public Health, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA; (J.B.S.); (K.C.S.)
| | - Shyamal D. Peddada
- Biostatistics and Computational Biology Branch (BCBB), National Institute of Environmental Health Sciences (NIEHS), NIH, Durham, NC 27709, USA
| |
Collapse
|
25
|
Daglia M, Drago L, Ullah H, Di Minno A, Brindisi G, Brunese FP, Dinardo G, Gori A, Indolfi C, Naso M, Tondina E, Trincianti C, Varricchio A, Zicari AM, Ciprandi G. Effects of the supplementation of single and multi-strain probiotics, alone or in combination with other treatments, on asthma in children: A systematic review of the randomized, placebo-controlled clinical studies. J Funct Foods 2024; 123:106599. [DOI: 10.1016/j.jff.2024.106599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
26
|
Kim J, Lee Y, Chung Y. Control of T-cell immunity by fatty acid metabolism. Ann Pediatr Endocrinol Metab 2024; 29:356-364. [PMID: 39778404 PMCID: PMC11725633 DOI: 10.6065/apem.2448160.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Fatty acids play critical roles in maintaining the cellular functions of T cells and regulating T-cell immunity. This review synthesizes current research on the influence of fatty acids on T-cell subsets, including CD8+ T cells, TH1, TH17, Treg (regulatory T cells), and TFH (T follicular helper) cells. Fatty acids impact T cells by modulating signaling pathways, inducing metabolic changes, altering cellular structures, and regulating gene expression epigenetically. These processes affect T-cell activation, differentiation, and function, with implications for diseases such as autoimmune disease and cancer. Based on these insights, fatty acid pathways can potentially be modulated by novel therapeutics, paving the way for novel treatment approaches for immune-mediated disorders and cancer immunotherapy.
Collapse
Affiliation(s)
- Jaemin Kim
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yoosun Lee
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
27
|
Shi M, Xue Q, Xie J, Yang Q, Tong J, Zhu J, Gao Y, Ma X, Wu D, Li Z. Protective effect of Shenqi Wenfei Formula against lipopolysaccharide/cigarette smoke-induced COPD in Rat based on gut microbiota and network pharmacology analysis. Front Microbiol 2024; 15:1441015. [PMID: 39629210 PMCID: PMC11611827 DOI: 10.3389/fmicb.2024.1441015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction The incidence of chronic obstructive pulmonary disease (COPD) appears to be increasing and evidence suggests that the intestinal flora may play a causative role in its development. Previous studies found that the Shenqi Wenfei Formula (SQWF) can regulate pyroptosis via the NLRP3/GSDMD pathway, thereby reducing the inflammatory response in the lungs of COPD model rats. However, there is no information on whether the drug's effects are associated with intestinal flora. Therefore, this study investigates whether the effects of SQWF are mediated through the regulation of intestinal flora, aiming to elucidate the underlying mechanisms of its therapeutic impact on COPD. Methods COPD was induced in rats using lipopolysaccharide and cigarette smoke, followed by intragastric administration of SQWF or physiological saline The targets of SQWF, associated signaling pathways, and key bacterial groups were investigated using 16S rRNA sequencing, network pharmacology, and bioinformatics techniques. The prediction results were validated using quantitative reverse transcription PCR, western blotting, and immunofluorescence, among other methods. Results SQWF treatment was found to alleviate COPD in model rats. Treatment was also observed to restore the balance of the intestinal flora in the rats, especially by reducing the abundance of g_Parabacteroides. Bioinformatics predictions identified g_Parabacteroides metabolites, RelA, HDAC1, and enriched neutrophil extracellular trap formation pathways as core targets of SQWF in COPD. qRT-PCR and Western blotting results showed that SQWF treatment reduced ReLA and HDAC1 mRNA and protein expression, along with decreased myeloperoxidase and neutrophil elastase levels in the nucleus. Conclusion Treatment with SQWF was found to restore the imbalance of intestinal g_Parabacteroides in COPD and also regulate the expression of the ReLA and HDAC1 genes, thereby reducing pulmonary neutrophil extracellular traps and alleviating lung inflammation.
Collapse
Affiliation(s)
- Mengyao Shi
- Anhui University of Chinese Medicine, Hefei, China
| | - Qian Xue
- Anhui University of Chinese Medicine, Hefei, China
| | - Jinghui Xie
- Anhui University of Chinese Medicine, Hefei, China
| | - Qinjun Yang
- Anhui University of Chinese Medicine, Hefei, China
- Chinese Medicine Respiratory Disease Prevention Institute, Hefei, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, China
| | - Jiabing Tong
- Anhui University of Chinese Medicine, Hefei, China
- Chinese Medicine Respiratory Disease Prevention Institute, Hefei, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, China
| | - Jie Zhu
- Anhui University of Chinese Medicine, Hefei, China
- Chinese Medicine Respiratory Disease Prevention Institute, Hefei, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, China
| | - Yating Gao
- Chinese Medicine Respiratory Disease Prevention Institute, Hefei, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, China
- First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Xiao Ma
- Anhui University of Chinese Medicine, Hefei, China
| | - Di Wu
- Anhui University of Chinese Medicine, Hefei, China
- First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zegeng Li
- Anhui University of Chinese Medicine, Hefei, China
- Chinese Medicine Respiratory Disease Prevention Institute, Hefei, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, China
- First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
28
|
Druszczynska M, Sadowska B, Kulesza J, Gąsienica-Gliwa N, Kulesza E, Fol M. The Intriguing Connection Between the Gut and Lung Microbiomes. Pathogens 2024; 13:1005. [PMID: 39599558 PMCID: PMC11597816 DOI: 10.3390/pathogens13111005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Recent advances in microbiome research have uncovered a dynamic and complex connection between the gut and lungs, known as the gut-lung axis. This bidirectional communication network plays a critical role in modulating immune responses and maintaining respiratory health. Mediated by immune interactions, metabolic byproducts, and microbial communities in both organs, this axis demonstrates how gut-derived signals, such as metabolites and immune modulators, can reach the lung tissue via systemic circulation, influencing respiratory function and disease susceptibility. To explore the implications of this connection, we conducted a systematic review of studies published between 2001 and 2024 (with as much as nearly 60% covering the period 2020-2024), using keywords such as "gut-lung axis", "microbiome", "respiratory disease", and "immune signaling". Studies were selected based on their relevance to gut-lung communication mechanisms, the impact of dysbiosis, and the role of the gut microbiota in respiratory diseases. This review provides a comprehensive overview of the gut-lung microbiome axis, emphasizing its importance in regulating inflammatory and immune responses linked to respiratory health. Understanding this intricate pathway opens new avenues for microbiota-targeted therapeutic strategies, which could offer promising interventions for respiratory diseases like asthma, chronic obstructive pulmonary disease, and even infections. The insights gained through this research underscore the potential of the gut-lung axis as a novel target for preventative and therapeutic approaches in respiratory medicine, with implications for enhancing both gut and lung health.
Collapse
Affiliation(s)
- Magdalena Druszczynska
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, Institute of Microbiology, Biotechnology and Immunology, University of Lodz, 90-237 Lodz, Poland; (B.S.); (N.G.-G.); (M.F.)
| | - Beata Sadowska
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, Institute of Microbiology, Biotechnology and Immunology, University of Lodz, 90-237 Lodz, Poland; (B.S.); (N.G.-G.); (M.F.)
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, 91-347 Lodz, Poland;
| | - Nikodem Gąsienica-Gliwa
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, Institute of Microbiology, Biotechnology and Immunology, University of Lodz, 90-237 Lodz, Poland; (B.S.); (N.G.-G.); (M.F.)
| | - Ewelina Kulesza
- Department of Rheumatology and Internal Diseases, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Marek Fol
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, Institute of Microbiology, Biotechnology and Immunology, University of Lodz, 90-237 Lodz, Poland; (B.S.); (N.G.-G.); (M.F.)
| |
Collapse
|
29
|
Kabil A, Nayyar N, Brassard J, Li Y, Chopra S, Hughes MR, McNagny KM. Microbial intestinal dysbiosis drives long-term allergic susceptibility by sculpting an ILC2-B1 cell-innate IgE axis. J Allergy Clin Immunol 2024; 154:1260-1276.e9. [PMID: 39134158 DOI: 10.1016/j.jaci.2024.07.023] [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: 02/27/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND The abundance and diversity of intestinal commensal bacteria influence systemic immunity with impact on disease susceptibility and severity. For example, loss of short chain fatty acid (SCFA)-fermenting bacteria in early life (humans and mice) is associated with enhanced type 2 immune responses in peripheral tissues including the lung. OBJECTIVE Our goal was to reveal the microbiome-dependent cellular and molecular mechanisms driving enhanced susceptibility to type 2 allergic lung disease. METHODS We used low-dose vancomycin to selectively deplete SCFA-fermenting bacteria in wild-type mice. We then examined the frequency and activation status of innate and adaptive immune cell lineages with and without SCFA supplementation. Finally, we used ILC2-deficient and signal transducer and activator of transcription 6 (STAT6)-deficient transgenic mouse strains to delineate the cellular and cytokine pathways leading to enhanced allergic disease susceptibility. RESULTS Mice with vancomycin-induced dysbiosis exhibited a 2-fold increase in lung ILC2 primed to produce elevated levels of IL-2, -5, and -13. In addition, upon IL-33 inhalation, mouse lung ILC2 displayed a novel ability to produce high levels of IL-4. These expanded and primed ILC2s drove B1 cell expansion and IL-4-dependent production of IgE that in turn led to exacerbated allergic inflammation. Importantly, these enhanced lung inflammatory phenotypes in mice with vancomycin-induced dysbiosis were reversed by administration of dietary SCFA (specifically butyrate). CONCLUSION SCFAs regulate an ILC2-B1 cell-IgE axis. Early-life administration of vancomycin, an antibiotic known to deplete SCFA-fermenting gut bacteria, primes and amplifies this axis and leads to lifelong enhanced susceptibility to type 2 allergic lung disease.
Collapse
Affiliation(s)
- Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Natalia Nayyar
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yicong Li
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sameeksha Chopra
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael R Hughes
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Kelly M McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada; Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
30
|
Hou Y, Wang D, Zhou S, Huo C, Chen H, Li F, Ding M, Li H, Zhao H, He J, Da H, Ma Y, Qiang Z, Chen X, Bai C, Cui J, Gao N, Liu Y. Probiotics combined with prebiotics alleviated seasonal allergic rhinitis by altering the composition and metabolic function of intestinal microbiota: a prospective, randomized, double-blind, placebo-controlled clinical trial. Front Immunol 2024; 15:1439830. [PMID: 39555052 PMCID: PMC11563974 DOI: 10.3389/fimmu.2024.1439830] [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: 05/28/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Background Numerous studies have established that probiotics or prebiotics can relieve the symptoms of allergic rhinitis (AR), but their mechanism of action remain underexplored. This study aimed to observe the clinical efficacy of probiotics combined with prebiotics in seasonal AR patients and explore their underlying mechanisms. Methods We conducted a prospective, randomized, double-blind, placebo-controlled clinical trial. The test group was given probiotics combined with prebiotics, whereas the placebo group was administered simulated preparation for 90 days. Outcome measures included total nasal symptom score (TNSS), visual analog scale, rhinitis quality of life questionnaire, fractional exhaled nitric oxide, and the rate and intensity of Loratadine use. Serum TNF-α, INF-γ, IL-4, IL-17, and IgE levels were measured by enzyme-linked immunosorbent assay. Intestinal microbiota was detected by 16S rRNA gene sequencing and quantitative PCR. Short-chain fatty acids were analyzed by gas chromatography-mass spectrometry. Results 106 participants (N = 53 for both test group and placebo group) completed the study. From baseline to day 91, mean difference between groups (MDBG) in the reduction of TNSS was -1.1 (-2.2, -0.1) (P = 0.04); MDBG in the increment of TNF-α was 7.1 pg/ml (95% CI: 0.8, 13.4, P = 0.03); the INF-γ level was significantly increased (P = 0.01), whereas that of IL-17 (P = 0.005) was significantly decreased in the test group, whilst mean difference within groups was not statistically significant in the placebo group; MDBG in the increment of acetate was 12.4% (95% CI: 7.1%, 17.6%, P <0.001). After the administration of probiotics and prebiotics, the composition and metabolic function of the intestinal microbiota were significantly altered and positively related to the beneficial effect on seasonal AR patients. Conclusion Probiotics combined with prebiotics administered for 90 days significantly attenuated the symptoms of seasonal AR patients, which may related to fluctuations in the composition and metabolic function of the intestinal microbiota and further ameliorating host immunity.
Collapse
Affiliation(s)
- Yangfan Hou
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Shuru Zhou
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Caifang Huo
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Haijuan Chen
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Fangxia Li
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Minjuan Ding
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Hongxin Li
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Hongyan Zhao
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Jin He
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Hongju Da
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Yu Ma
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Zhihui Qiang
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Xiushan Chen
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
| | - Cairong Bai
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
- Department of Allergy, Yulin No.2 Hospital, Yulin, China
| | - Jing Cui
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
| | - Na Gao
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
- Department of Allergy, Yulin No.2 Hospital, Yulin, China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| |
Collapse
|
31
|
Donald K, Finlay BB. Mechanisms of microbe-mediated immune development in the context of antibiotics and asthma. FRONTIERS IN ALLERGY 2024; 5:1469426. [PMID: 39469482 PMCID: PMC11513386 DOI: 10.3389/falgy.2024.1469426] [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: 07/23/2024] [Accepted: 08/28/2024] [Indexed: 10/30/2024] Open
Abstract
The gut houses 70%-80% of the body's immune cells and represents the main point of contact between the immune system and the outside world. Immune maturation occurs largely after birth and is guided by the gut microbiota. In addition to the many human clinical studies that have identified relationships between gut microbiota composition and disease outcomes, experimental research has demonstrated a plethora of mechanisms by which specific microbes and microbial metabolites train the developing immune system. The healthy maturation of the gut microbiota has been well-characterized and discreet stages marked by changes in abundance of specific microbes have been identified. Building on Chapter 8, which discusses experimental models used to study the relationship between the gut microbiota and asthma, the present review aims to dive deeper into the specific microbes and metabolites that drive key processes in immune development. The implications of microbiota maturation patterns in the context of asthma and allergies, as well as the effects of antibiotics on microbe-immune crosstalk, will also be discussed.
Collapse
Affiliation(s)
- Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Departmentof Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Departmentof Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Chen Y, Wen Y, Zhao R, Zhu Y, Chen Z, Zhao C, Mu W. Human milk oligosaccharides in preventing food allergy: A review through gut microbiota and immune regulation. Int J Biol Macromol 2024; 278:134868. [PMID: 39163965 DOI: 10.1016/j.ijbiomac.2024.134868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
Food allergy (FA) has increasingly attracted global attention in past decades. However, the mechanism and effect of FA are complex and varied, rendering it hard to prevention and management. Most of the allergens identified so far are macromolecular proteins in food and may have potential cross-reactions. Human milk oligosaccharides (HMOs) have been regarded as an ideal nutrient component for infants, as they can enhance the immunomodulatory capacity to inhibit the progress of FA. HMOs may intervene in the development of allergies by modifying gut microbiota and increasing specific short-chain fatty acids levels. Additionally, HMOs could improve the intestinal permeability and directly or indirectly regulate the balance of T helper cells and regulatory T cells by enhancing the inflammatory signaling pathways to combat FA. This review will discuss the influence factors of FA, key species of gut microbiota involved in FA, types of FA, and profiles of HMOs and provide evidence for future research trends to advance HMOs as potential therapeutic aids in preventing the progress of FA.
Collapse
Affiliation(s)
- Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Yuxi Wen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Department of Analytical and Food Chemistry, Faculty of Sciences, Universidade de Vigo, 32004 Ourense, Spain
| | - Runfan Zhao
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Zhengxin Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
33
|
Lu K, Li C, Men J, Xu B, Chen Y, Yan P, Gai Z, Zhang Q, Zhang L. Traditional Chinese medicine to improve immune imbalance of asthma: focus on the adjustment of gut microbiota. Front Microbiol 2024; 15:1409128. [PMID: 39411430 PMCID: PMC11473343 DOI: 10.3389/fmicb.2024.1409128] [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: 03/29/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Asthma, being the prevailing respiratory ailment globally, remains enigmatic in terms of its pathogenesis. In recent times, the advancement of traditional Chinese medicine pertaining to the intestinal microbiota has yielded a plethora of investigations, which have substantiated the potential of traditional Chinese medicine in disease prevention and treatment through modulation of the intestinal microbiota. Both animal models and clinical trials have unequivocally demonstrated the indispensable role of the intestinal microbiota in the pathogenesis of asthma. This article presents a summary of the therapeutic effects of traditional Chinese medicine in the context of regulating gut microbiota and its metabolites, thereby achieving immune regulation and inhibiting airway inflammation associated with asthma. It elucidates the mechanism by which traditional Chinese medicine modulates the gut microbiota to enhance asthma management, offering a scientific foundation for the utilization of traditional Chinese medicine in the treatment of asthma.
Collapse
Affiliation(s)
- Ke Lu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingwen Men
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bin Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizheng Yan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhibo Gai
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingxiang Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lu Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
34
|
Gao Y, Yao Q, Meng L, Wang J, Zheng N. Double-side role of short chain fatty acids on host health via the gut-organ axes. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:322-339. [PMID: 39290857 PMCID: PMC11406094 DOI: 10.1016/j.aninu.2024.05.001] [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: 05/23/2023] [Revised: 01/29/2024] [Accepted: 05/14/2024] [Indexed: 09/19/2024]
Abstract
Short chain fatty acids (SCFA) exist in dietary foods and are produced by the fermentation of gut microbiota, and are considered an important element for regulating host health. Through blood circulation, SCFA produced in the gut and obtained from foods have an impact on the intestinal health as well as vital organs of the host. It has been recognized that the gut is the "vital organ" in the host. As the gut microbial metabolites, SCFA could create an "axis" connecting the gut and to other organs. Therefore, the "gut-organ axes" have become a focus of research in recent years to analyze organism health. In this review, we summarized the sources, absorption properties, and the function of SCFA in both gut and other peripheral tissues (brain, kidney, liver, lung, bone and cardiovascular) in the way of "gut-organ axes". Short chain fatty acids exert both beneficial and pathological role in gut and other organs in various ways, in which the beneficial effects are more pronounced. In addition, the beneficial effects are reflected in both preventive and therapeutic effects. More importantly, the mechanisms behinds the gut and other tissues provided insight into the function of SCFA, assisting in the development of novel preventive and therapeutic strategies for maintaining the host health.
Collapse
Affiliation(s)
- Yanan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qianqian Yao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège 4000, Belgium
| | - Lu Meng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
35
|
Kim YC, Sohn KH, Kang HR. Gut microbiota dysbiosis and its impact on asthma and other lung diseases: potential therapeutic approaches. Korean J Intern Med 2024; 39:746-758. [PMID: 39252487 PMCID: PMC11384250 DOI: 10.3904/kjim.2023.451] [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: 10/21/2023] [Revised: 03/08/2024] [Accepted: 04/07/2024] [Indexed: 09/11/2024] Open
Abstract
The emerging field of gut-lung axis research has revealed a complex interplay between the gut microbiota and respiratory health, particularly in asthma. This review comprehensively explored the intricate relationship between these two systems, focusing on their influence on immune responses, inflammation, and the pathogenesis of respiratory diseases. Recent studies have demonstrated that gut microbiota dysbiosis can contribute to asthma onset and exacerbation, prompting investigations into therapeutic strategies to correct this imbalance. Probiotics and prebiotics, known for their ability to modulate gut microbial compositions, were discussed as potential interventions to restore immune homeostasis. The impact of antibiotics and metabolites, including short-chain fatty acids produced by the gut microbiota, on immune regulation was examined. Fecal microbiota transplantation has shown promise in various diseases, but its role in respiratory disorders is not established. Innovative approaches, including mucus transplants, inhaled probiotics, and microencapsulation strategies, have been proposed as novel therapeutic avenues. Despite challenges, including the sophisticated adaptability of microbial communities and the need for mechanistic clarity, the potential for microbiota-based interventions is considerable. Collaboration between researchers, clinicians, and other experts is essential to unravel the complexities of the gut-lung axis, paving a way for innovative strategies that could transform the management of respiratory diseases.
Collapse
Affiliation(s)
- Young-Chan Kim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyoung-Hee Sohn
- Division of Respiratory, Allergy and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Hospital, Seoul, Korea
| | - Hye-Ryun Kang
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Zhao M, Tian J, Hou W, Yin L, Li W. Global research trends on the associations between the microbiota and lung cancer: a visualization bibliometric analysis (2008-2023). Front Microbiol 2024; 15:1416385. [PMID: 39282557 PMCID: PMC11392740 DOI: 10.3389/fmicb.2024.1416385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Numerous papers have been published on the microbiota in lung cancer in recent years. However, there is still a lack of bibliometric analysis of the microbiota in lung cancer in this field. Our paper did bibliometric analyses and elucidated the knowledge structure and study hotspots related to the microbiota in lung cancer patients. We screened publications reporting on the microbiota in lung cancer from 2008 to 2023 from the Web of Science Core Collection (WoSCC) database, and carried out bibliometric analyses by the application of the VOSviewers, CiteSpace and R package "bibliometrix." The 684 documents enrolled in the analysis were obtained from 331 institutions in 67 regions by 4,661 authors and were recorded in 340 journals. Annual papers are growing rapidly, and the countries of China, the United States and Italy are contributing the most to this area of research. Zhejiang University is the main research organization. Science and Cancer had significant impacts on this area. Zhang Yan had the most articles, and the Bertrand Routy had the most co-cited times. Exploring the mechanism of action of the lung and/or gut microbiota in lung cancer and therapeutic strategies involving immune checkpoint inhibitors in lung cancer are the main topics. Moreover, "gut microbiota," "immunotherapy," and "short-chain fatty acids" are important keywords for upcoming study hotspots. In conclusion, microbiota research offers promising opportunities in lung cancer, with pivotal studies exploring the mechanisms that link lung and gut microbiota to therapeutic strategies, particularly through immune checkpoint inhibitors. Moreover, the gut-lung axis emerges as a novel target for innovative treatments. Further research is essential to unravel the detailed mechanisms of this connection.
Collapse
Affiliation(s)
- Maoyuan Zhao
- Lung Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wang Hou
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liyuan Yin
- Lung Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
37
|
Li X, Shang S, Wu M, Song Q, Chen D. Gut microbial metabolites in lung cancer development and immunotherapy: Novel insights into gut-lung axis. Cancer Lett 2024; 598:217096. [PMID: 38969161 DOI: 10.1016/j.canlet.2024.217096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Metabolic derivatives of numerous microorganisms inhabiting the human gut can participate in regulating physiological activities and immune status of the lungs through the gut-lung axis. The current well-established microbial metabolites include short-chain fatty acids (SCFAs), tryptophan and its derivatives, polyamines (PAs), secondary bile acids (SBAs), etc. As the study continues to deepen, the critical function of microbial metabolites in the occurrence and treatment of lung cancer has gradually been revealed. Microbial derivates can enter the circulation system to modulate the immune microenvironment of lung cancer. Mechanistically, oncometabolites damage host DNA and promote the occurrence of lung cancer, while tumor-suppresive metabolites directly affect the immune system to combat the malignant properties of cancer cells and even show considerable application potential in improving the efficacy of lung cancer immunotherapy. Considering the crosstalk along the gut-lung axis, in-depth exploration of microbial metabolites in patients' feces or serum will provide novel guidance for lung cancer diagnosis and treatment selection strategies. In addition, targeted therapeutics on microbial metabolites are expected to overcome the bottleneck of lung cancer immunotherapy and alleviate adverse reactions, including fecal microbiota transplantation, microecological preparations, metabolite synthesis and drugs targeting metabolic pathways. In summary, this review provides novel insights and explanations on the intricate interplay between gut microbial metabolites and lung cancer development, and immunotherapy through the lens of the gut-lung axis, which further confirms the possible translational potential of the microbiome metabolome in lung cancer treatment.
Collapse
Affiliation(s)
- Xinpei Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shijie Shang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qian Song
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
38
|
Węgrzyn K, Jasińska A, Janeczek K, Feleszko W. The Role of Postbiotics in Asthma Treatment. Microorganisms 2024; 12:1642. [PMID: 39203484 PMCID: PMC11356534 DOI: 10.3390/microorganisms12081642] [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: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
In recent years, there has been abundant research concerning human microbiome and its impact on the host's health. Studies have shown that not only the commensal bacteria itself, but also postbiotics, understood as inanimate microorganisms, possibly with the presence of their components, may themselves have an effect on various elements of human physiology. In this review, we take a closer look at the specific ways in which postbiotics can alter immune response in allergic asthma, which is one of the most prevalent allergic diseases in today's world and a serious subject of concern. Through altering patients' immune response, not only to allergens but also to pathogens, postbiotics could have a significant role in lowering the number of asthma exacerbations. We suggest that more profound research should be undertaken in order to launch postbiotics into clinical standards of asthma treatment, given the greatly promising findings in terms of their immunomodulating potential.
Collapse
Affiliation(s)
- Konstancja Węgrzyn
- Central Clinical Hospital, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Agnieszka Jasińska
- Department of Pediatric Pneumonology and Allergy, University Clinical Centre, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Kamil Janeczek
- Department of Paediatric Pulmonology and Rheumatology, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Wojciech Feleszko
- Department of Pediatric Pneumonology and Allergy, University Clinical Centre, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
39
|
Magalhães J, Cappellozza BI, Dos Santos TC, Inoe F, Pessoa Araújo Júnior J, Kurissio JK, Queiroz O, Joergensen JN, Cooke RF, Vasconcelos CGC, Vasconcelos JLM. Effects of supplementing direct-fed microbials on health and growth of preweaning Gyr × Holstein dairy calves. J Dairy Sci 2024; 107:6117-6130. [PMID: 38608942 DOI: 10.3168/jds.2023-24434] [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: 11/15/2023] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
This study aimed to evaluate the effects of direct-fed microbials (DFM) on health and growth responses of preweaning Bos indicus × Bos taurus (Gyr × Holstein) crossbred calves. Ninety newborn heifer calves (initial BW of 35 ± 4.0 kg) were used. At birth, calves were ranked by initial BW and parity of the dam and assigned to: (1) whole milk without DFM supplementation (CON; n = 30), (2) whole milk with the addition of 1.0 g/calf per day of a Bacillus-based DFM (BAC; n = 30), or (3) whole milk with the addition of 1.0 g/calf per day of BAC and 1.2 g/calf per day of Enterococcus faecium 669 (MIX; n = 30). Milk was fed individually during the study (77 d), and the BAC and MIX treatments were offered daily throughout the 77-d preweaning period. All calves were offered a starter supplement and corn silage starting on d 1 and 60 of age, respectively. Milk and starter supplement intake were evaluated daily, and BW was recorded on d 0 and at weaning (d 77). Diarrhea and pneumonia were assessed daily, and fecal samples were collected on d 0, 7, 14, 21, and at weaning (d 77) for assessment of the presence of bacterial and protozoal pathogens via qPCR. All data were analyzed using SAS (v. 9.4) with calf as the experimental unit and using single-df orthogonal contrasts (BAC + MIX vs. CON; BAC vs. MIX). Daily feeding of DFM, regardless of type, improved weaning BW. Odds ratio for occurrence of pneumonia was lower for DFM-supplemented calves, but its occurrence did not differ between BAC and MIX calves. No Salmonella spp. or Escherichia coli F41 were detected in any of the calves. The proportion of calves positive for E. coli F17 was greater for DFM calves on d 7 (92% and 96% vs. 81% for BAC, MIX, and CON, respectively), on d 21 (13% and 26% vs. 7% for BAC, MIX, and CON, respectively), and at weaning (48% and 35% vs. 22% for BAC, MIX, and CON, respectively). For Clostridium difficile, more DFM calves were positive on d 7 (65% and 30% vs. 35% for BAC, MIX, and CON, respectively) and 14 (20% and 28% vs. 7% for BAC, MIX, and CON, respectively), but proportion of positive calves was also greater for BAC versus MIX on d 7. More CON calves were positive for Clostridium perfringens on d 14 (14% vs. 3% and 8% for CON, BAC, and MIX, respectively) compared with DFM-fed calves. Incidence of calves positive for C. perfringens was greater in BAC than MIX on d 7 (50% vs. 18%), and greater for MIX than BAC at weaning (9% vs. 0%). For protozoa occurrence, a lower proportion of DFM calves were positive for Cryptosporidium spp. on d 7 (58% and 48% vs. 76% for BAC, MIX, and CON, respectively), but opposite results were observed on d 21 for Cryptosporidium spp. (3% and 11% vs. 0% for BAC, MIX, and CON, respectively) and Eimeria spp. on d 14 (7% and 8% vs. 0% for BAC, MIX, and CON, respectively) and 21 (50% and 59% vs. 38% for BAC, MIX, and CON, respectively). In summary, DFM feeding alleviated the occurrence of pneumonia and improved growth rates, while also modulating the prevalence of bacteria and protozoa in preweaning Gyr × Holstein calves.
Collapse
Affiliation(s)
- Julia Magalhães
- School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP 18618-000, Brazil
| | - Bruno I Cappellozza
- Commercial Development, Animal & Plant Health and Nutrition, Chr. Hansen A/S, Hørsholm 2970, Denmark
| | - Taynara C Dos Santos
- School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP 18618-000, Brazil
| | - Fernanda Inoe
- School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP 18618-000, Brazil
| | - João Pessoa Araújo Júnior
- Institute of Biotechnology (IBTEC), School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP 18607-440, Brazil
| | - Jacqueline K Kurissio
- Institute of Biotechnology (IBTEC), School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP 18607-440, Brazil
| | - Oscar Queiroz
- Commercial Development, Animal & Plant Health and Nutrition, Chr. Hansen A/S, Hørsholm 2970, Denmark
| | - Jens N Joergensen
- Commercial Development, Animal & Plant Health and Nutrition, Chr. Hansen A/S, Hørsholm 2970, Denmark
| | - Reinaldo F Cooke
- Department of Animal Science, Texas A&M University, College Station, TX 77843
| | | | - José Luiz M Vasconcelos
- School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP 18618-000, Brazil.
| |
Collapse
|
40
|
Mann ER, Lam YK, Uhlig HH. Short-chain fatty acids: linking diet, the microbiome and immunity. Nat Rev Immunol 2024; 24:577-595. [PMID: 38565643 DOI: 10.1038/s41577-024-01014-8] [Citation(s) in RCA: 231] [Impact Index Per Article: 231.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
The short-chain fatty acids (SCFAs) butyrate, propionate and acetate are microbial metabolites and their availability in the gut and other organs is determined by environmental factors, such as diet and use of antibiotics, that shape the diversity and metabolism of the microbiota. SCFAs regulate epithelial barrier function as well as mucosal and systemic immunity via evolutionary conserved processes that involve G protein-coupled receptor signalling or histone deacetylase activity. Indicatively, the anti-inflammatory role of butyrate is mediated through direct effects on the differentiation of intestinal epithelial cells, phagocytes, B cells and plasma cells, and regulatory and effector T cells. Intestinally derived SCFAs also directly and indirectly affect immunity at extra-intestinal sites, such as the liver, the lungs, the reproductive tract and the brain, and have been implicated in a range of disorders, including infections, intestinal inflammation, autoimmunity, food allergies, asthma and responses to cancer therapies. An ecological understanding of microbial communities and their interrelated metabolic states, as well as the engineering of butyrogenic bacteria may support SCFA-focused interventions for the prevention and treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Elizabeth R Mann
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ying Ka Lam
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
41
|
Ezenabor EH, Adeyemi AA, Adeyemi OS. Gut Microbiota and Metabolic Syndrome: Relationships and Opportunities for New Therapeutic Strategies. SCIENTIFICA 2024; 2024:4222083. [PMID: 39041052 PMCID: PMC11262881 DOI: 10.1155/2024/4222083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/10/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024]
Abstract
Since its discovery, numerous studies have shown the role of the microbiota in well-being and disease. The gut microbiota represents an essential factor that plays a multidirectional role that affects not just the gut but also other parts of the body, including the brain, endocrine system, humoral system, immune system, and metabolic pathways, as well as host-microbiome interactions. Through a comprehensive analysis of existing literature using the desktop research methodology, this review elucidates the mechanisms by which gut microbiota dysbiosis contributes to metabolic dysfunction, including obesity, dyslipidaemia, hypertension, atherosclerosis, hyperuricemia, and hyperglycaemia. Furthermore, it examines the bidirectional communication pathways between gut microbiota and host metabolism, highlighting the role of microbial-derived metabolites, immune modulation, and gut barrier integrity in shaping metabolic homeostasis. Importantly, the review identifies promising therapeutic strategies targeting the gut microbiota as potential interventions for metabolic syndrome, including probiotics, prebiotics, symbiotics, dietary modifications, and faecal microbiota transplantation. By delineating the bidirectional interactions between gut microbiota and metabolic syndrome, the review not only advances our understanding of disease pathophysiology but also underscores the potential for innovative microbiota-based interventions to mitigate the global burden of metabolic syndrome and its associated complications.
Collapse
Affiliation(s)
- Emmanuel Henry Ezenabor
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Aishat Abimbola Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Oluyomi Stephen Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| |
Collapse
|
42
|
Korkmaz FT, Quinton LJ. Extra-pulmonary control of respiratory defense. Cell Immunol 2024; 401-402:104841. [PMID: 38878619 PMCID: PMC12002097 DOI: 10.1016/j.cellimm.2024.104841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024]
Abstract
Pneumonia persists as a public health crisis, representing the leading cause of death due to infection. Whether respiratory tract infections progress to pneumonia and its sequelae such as acute respiratory distress syndrome and sepsis depends on numerous underlying conditions related to both the causative agent and host. Regarding the former, pneumonia burden remains staggeringly high, despite the effectiveness of pathogen-targeting strategies such as vaccines and antibiotics. This demands a greater understanding of host features that collaborate to promote immune resistance and tissue resilience in the infected lung. Such features inside the pulmonary compartment have drawn much attention, where major advances have been made related to resident and recruited immune activity. By comparison, extra-pulmonary processes guiding pneumonia susceptibility are relatively elusive, constituting the focus of this review. Here we will highlight examples of when, how, and why tissues outside of the lungs dispatch signals that modulate local immunity in the airspaces. Topics include the liver, gut, bone marrow, brain and more, all of which contribute in direct and indirect ways to pneumonia outcome. When tuned appropriately, it has become clear that these responses can serve protective roles, and this will be considered distinctly from what would otherwise be aberrant responses characteristic of pneumonia-induced organ injury and sepsis. Further advances in this area may reveal novel targetable areas for clinical intervention that are not confined to the intra-pulmonary space.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology and Infectious Disease, UMass Chan Medical School, Worcester, MA 01602, United States.
| | - Lee J Quinton
- Department of Medicine, Division of Immunology and Infectious Disease, UMass Chan Medical School, Worcester, MA 01602, United States
| |
Collapse
|
43
|
Musiol S, Harris CP, Gschwendtner S, Burrell A, Amar Y, Schnautz B, Renisch D, Braun SC, Haak S, Schloter M, Schmidt-Weber CB, Zielinski CE, Alessandrini F. The impact of high-salt diet on asthma in humans and mice: Effect on specific T-cell signatures and microbiome. Allergy 2024; 79:1844-1857. [PMID: 38798015 DOI: 10.1111/all.16148] [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: 12/06/2023] [Revised: 03/25/2024] [Accepted: 04/14/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND The rise in asthma has been linked to different environmental and lifestyle factors including dietary habits. Whether dietary salt contributes to asthma incidence, remains controversial. We aimed to investigate the impact of higher salt intake on asthma incidence in humans and to evaluate underlying mechanisms using mouse models. METHODS Epidemiological research was conducted using the UK Biobank Resource. Data were obtained from 42,976 participants with a history of allergies. 24-h sodium excretion was estimated from spot urine, and its association with asthma incidence was assessed by Cox regression, adjusting for relevant covariates. For mechanistic studies, a mouse model of mite-induced allergic airway inflammation (AAI) fed with high-salt diet (HSD) or normal-salt chow was used to characterize disease development. The microbiome of lung and feces (as proxy for gut) was analyzed via 16S rRNA gene based metabarcoding approach. RESULTS In humans, urinary sodium excretion was directly associated with asthma incidence among females but not among males. HSD-fed female mice displayed an aggravated AAI characterized by increased levels of total IgE, a TH2-TH17-biased inflammatory cell infiltration accompanied by upregulation of osmosensitive stress genes. HSD induced distinct changes in serum short chain fatty acids and in both gut and lung microbiome, with a lower Bacteroidetes to Firmicutes ratio and decreased Lactobacillus relative abundance in the gut, and enriched members of Gammaproteobacteria in the lung. CONCLUSIONS High dietary salt consumption correlates with asthma incidence in female adults with a history of allergies. Female mice revealed HSD-induced T-cell lung profiles accompanied by alterations of gut and lung microbiome.
Collapse
Affiliation(s)
- Stephanie Musiol
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Carla P Harris
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Dr. von Hauner Children's Hospital, University Hospital, LMU of Munich, Munich, Germany
| | - Silvia Gschwendtner
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Amy Burrell
- Department of Infection Immunology, Leibniz Institute for Natural Product Research & Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
| | - Yacine Amar
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Benjamin Schnautz
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Dennis Renisch
- Department of Chemistry - TRIGA site, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sonja C Braun
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Faculty of Medicine, LMU of Munich, Munich, Germany
| | - Stefan Haak
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Schloter
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Christina E Zielinski
- Department of Infection Immunology, Leibniz Institute for Natural Product Research & Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Center for Translational Cancer Research & Institute of Virology, Technical University of Munich, Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
44
|
Zhang XM, Huang YC, Chen BZ, Li Q, Wu PP, Chen WH, Wu RH, Li C. Water decoction of Pericarpium citri reticulatae and Amomi fructus ameliorates alcohol-induced liver disease involved in the modulation of gut microbiota and TLR4/NF-κB pathway. Front Pharmacol 2024; 15:1392338. [PMID: 38966547 PMCID: PMC11222602 DOI: 10.3389/fphar.2024.1392338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/15/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction Alcohol consumption alters the diversity and metabolic activities of gut microbiota, leading to intestinal barrier dysfunction and contributing to the development of alcoholic liver disease (ALD), which is the most prevalent cause of advanced liver diseases. In this study, we investigated the protective effects and action mechanism of an aqueous extraction of Pericarpium citri reticulatae and Amomi fructus (PFE) on alcoholic liver injury. Methods C57BL/6 mice were used to establish the mouse model of alcoholic liver injury and orally administered 500 and 1,000 mg/kg/d of PFE for 2 weeks. Histopathology, immunohistochemistry, immunofluorescence, Western blotting, qRT-PCR, and 16S rDNA amplicon sequencing were used to analyze the mechanism of action of PFE in the treatment of alcohol-induced liver injury. Results Treatment with PFE significantly improved alcohol-induced liver injury, as illustrated by the normalization of serum alanine aminotransferase, aspartate aminotransferase, total triglyceride, and cholesterol levels in ALD mice in a dose-dependent manner. Administration of PFE not only maintained the intestinal barrier integrity prominently by upregulating mucous production and tight junction protein expressions but also sensibly reversed the dysregulation of intestinal microecology in alcohol-treated mice. Furthermore, PFE treatment significantly reduced hepatic lipopolysaccharide (LPS) and attenuated oxidative stress as well as inflammation related to the TLR4/NF-κB signaling pathway. The PFE supplementation also significantly promoted the production of short-chain fatty acids (SCFAs) in the ALD mice. Conclusion Administration of PFE effectively prevents alcohol-induced liver injury and may also regulate the LPS-involved gut-liver axis; this could provide valuable insights for the development of drugs to prevent and treat ALD.
Collapse
Affiliation(s)
- Xing-Min Zhang
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Yue-Chang Huang
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Bai-Zhong Chen
- Guangdong Xinbaotang Biotechnology Co., Ltd., Jiangmen, China
| | - Qian Li
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Pan-Pan Wu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Wen-Hua Chen
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Ri-Hui Wu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Chen Li
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| |
Collapse
|
45
|
Zhang P, Zheng Z, Sun H, Gao T, Xiao X. A review of common influencing factors and possible mechanisms associated with allergic diseases complicating tic disorders in children. Front Pediatr 2024; 12:1360420. [PMID: 38957776 PMCID: PMC11218626 DOI: 10.3389/fped.2024.1360420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024] Open
Abstract
Over the past few decades, the incidence of childhood allergic diseases has increased globally, and their impact on the affected child extends beyond the allergy itself. There is evidence of an association between childhood allergic diseases and the development of neurological disorders. Several studies have shown a correlation between allergic diseases and tic disorders (TD), and allergic diseases may be an important risk factor for TD. Possible factors influencing the development of these disorders include neurotransmitter imbalance, maternal anxiety or depression, gut microbial disorders, sleep disturbances, maternal allergic status, exposure to tobacco, and environmental factors. Moreover, gut microbial disturbances, altered immunological profiles, and DNA methylation in patients with allergic diseases may be potential mechanisms contributing to the development of TD. An in-depth investigation of the relationship between allergic diseases and TD in children will be important for preventing and treating TD.
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Child Health, Dalian Municipal Women and Children’s Medical Center (Group), Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Zhimin Zheng
- Department of Child Health, Dalian Municipal Women and Children’s Medical Center (Group), Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Hao Sun
- Department of Child Health, Dalian Municipal Women and Children’s Medical Center (Group), Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Tieying Gao
- Department of Child Health, Dalian Municipal Women and Children’s Medical Center (Group), Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Xuwu Xiao
- Department of Child Health, Dalian Municipal Women and Children’s Medical Center (Group), Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
46
|
Petakh P, Duve K, Oksenych V, Behzadi P, Kamyshnyi O. Molecular mechanisms and therapeutic possibilities of short-chain fatty acids in posttraumatic stress disorder patients: a mini-review. Front Neurosci 2024; 18:1394953. [PMID: 38887367 PMCID: PMC11182003 DOI: 10.3389/fnins.2024.1394953] [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: 03/02/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
This mini-review explores the role of short-chain fatty acids (SCFAs) in posttraumatic stress disorder (PTSD). Highlighting the microbiota-gut-brain axis, this study investigated the bidirectional communication between the gut microbiome and mental health. SCFAs, byproducts of gut microbial fermentation, have been examined for their potential impact on PTSD, with a focus on molecular mechanisms and therapeutic interventions. This review discusses changes in SCFA levels and bacterial profiles in individuals with PTSD, emphasizing the need for further research. Promising outcomes from clinical trials using probiotics and fermented formulations suggest potential avenues for PTSD management. Future directions involve establishing comprehensive human cohorts, integrating multiomics data, and employing advanced computational methods, with the goal of deepening our understanding of the role of SCFAs in PTSD and exploring microbiota-targeted interventions.
Collapse
Affiliation(s)
- Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Khrystyna Duve
- Department of Neurology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
47
|
Liu J, Wen B, Huang Y, Deng G, Yan Q, Jia L. Exploring the effect of Clostridium butyricum on lung injury associated with acute pancreatitis in mice by combined 16S rRNA and metabolomics analysis. Anaerobe 2024; 87:102854. [PMID: 38614288 DOI: 10.1016/j.anaerobe.2024.102854] [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: 11/06/2023] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024]
Abstract
OBJECTIVES Acute lung injury is a critical complication of severe acute pancreatitis (SAP). The gut microbiota and its metabolites play an important role in SAP development and may provide new targets for AP-associated lung injury. Based on the ability to reverse AP injury, we proposed that Clostridium butyricum may reduce the potential for AP-associated lung injury by modulating with intestinal microbiota and related metabolic pathways. METHODS An AP disease model was established in mice and treated with C. butyricum. The structure and composition of the intestinal microbiota in mouse feces were analyzed by 16 S rRNA gene sequencing. Non-targeted metabolite analysis was used to quantify the microbiota derivatives. The histopathology of mouse pancreas and lung tissues was examined using hematoxylin-eosin staining. Pancreatic and lung tissues from mice were stained with immunohistochemistry and protein immunoblotting to detect inflammatory factors IL-6, IL-1β, and MCP-1. RESULTS C. butyricum ameliorated the dysregulation of microbiota diversity in a model of AP combined with lung injury and affected fatty acid metabolism by lowering triglyceride levels, which were closely related to the alteration in the relative abundance of Erysipelatoclostridium and Akkermansia. In addition, C. butyricum treatment attenuated pathological damage in the pancreatic and lung tissues and significantly suppressed the expression of inflammatory factors in mice. CONCLUSIONS C. butyricum may alleviate lung injury associated with AP by interfering with the relevant intestinal microbiota and modulating relevant metabolic pathways.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China; Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Biyan Wen
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China; Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Yaoxing Huang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China; Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Guiqing Deng
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China; Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Qingqing Yan
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China; Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Lin Jia
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China; Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China.
| |
Collapse
|
48
|
Liu J, Li Y, Shen D, Li X, Wang K, Nagaoka K, Li C. Gut microbiota intervention alleviates pulmonary inflammation in broilers exposed to fine particulate matter from broiler house. Appl Environ Microbiol 2024; 90:e0217423. [PMID: 38656183 PMCID: PMC11107152 DOI: 10.1128/aem.02174-23] [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: 12/05/2023] [Accepted: 03/31/2024] [Indexed: 04/26/2024] Open
Abstract
The gut microbiota of poultry is influenced by a variety of factors, including feed, drinking water, airborne dust, and footpads, among others. Gut microbiota can affect the immune reaction and inflammation in the lungs. To investigate the effect of gut microbiota variation on lung inflammation induced by PM2.5 (fine particulate matter) in broilers, 36 Arbor Acres (AA) broilers were randomly assigned to three groups: control group (CON), PM2.5 exposure group (PM), and PM2.5 exposure plus oral antibiotics group (PMA). We used non-absorbable antibiotics (ABX: neomycin and amikacin) to modify the microbiota composition in the PMA group. The intervention was conducted from the 18th to the 28th day of age. Broilers in the PM and PMA groups were exposed to PM by a systemic exposure method from 21 to 28 days old, and the concentration of PM2.5 was controlled at 2 mg/m3. At 28 days old, the lung injury score, relative mRNA expression of inflammatory factors, T-cell differentiation, and dendritic cell function were significantly increased in the PM group compared to the CON group, and those of the PMA group were significantly decreased compared to the PM group. There were significant differences in both α and β diversity of cecal microbiota among these three groups. Numerous bacterial genera showed significant differences in relative abundance among the three groups. In conclusion, gut microbiota could affect PM2.5-induced lung inflammation in broilers by adjusting the capacity of antigen-presenting cells to activate T-cell differentiation. IMPORTANCE Gut microbes can influence the development of lung inflammation, and fine particulate matter collected from broiler houses can lead to lung inflammation in broilers. In this study, we explored the effect of gut microbes modified by intestinal non-absorbable antibiotics on particulate matter-induced lung inflammation. The results showed that modification in the composition of gut microbiota could alleviate lung inflammation by attenuating the ability of dendritic cells to stimulate T-cell differentiation, which provides a new way to protect lung health in poultry farms.
Collapse
Affiliation(s)
- Junze Liu
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuan Li
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Dan Shen
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiaoqing Li
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kai Wang
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Chunmei Li
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
49
|
Schenzel A, Geiger A, Nendel E, Yang Z, Krammer S, Leberle A, Brunst AK, Trump S, Mittler S, Rauh M, Geppert CI, Tausche P, Hohenberger K, Rieker RJ, Schieweck O, Zundler S, Finotto S. Fiber rich food suppressed airway inflammation, GATA3 + Th2 cells, and FcεRIα+ eosinophils in asthma. Front Nutr 2024; 11:1367864. [PMID: 38757128 PMCID: PMC11097976 DOI: 10.3389/fnut.2024.1367864] [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: 01/09/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Background Allergic Asthma is a disease presenting various endotypes and no current therapies act curative but alleviate disease symptoms. Dietary interventions are gaining increasing importance in regulating immune responses. Furthermore, short chain fatty acids (SFCA), as the main products of dietary fiber's fermentation by the gut bacteria, ameliorate the pathogenesis and disease burden of different illnesses including asthma. Nevertheless, the connection and crosstalk between the gut and lung is poorly understood. Objective In this work, the role of high fiber diet on the development of allergic asthma at baseline and after exacerbation of disease induced by respiratory viruses was investigated. Methods Hereby, SCFA in serum of asthmatic and non-asthmatic pre-school children before and after airway disease symptoms were analyzed. Moreover, the effect of high fiber diet in vivo in a murine model of house dust mite extract (HDM) induced allergic asthma and in the end in isolated lung and spleen cells infected ex vivo with Rhinovirus was analyzed. Results In this study, a decrease of the SCFA 3-Hydroxybutyric acid in serum of asthmatic children after symptomatic episodes at convalescent visit as compared to asthmatic and control children at baseline visit was observed. In experimental asthma, in mice fed with high fiber diet, a reduced lung GATA3 + Th2 type mediated inflammation, mucus production and collagen deposition and expression of Fc epsilon receptor Ia (FcεRIa) in eosinophils was observed. By contrast, the CD8+ memory effector T cells were induced in the lungs of asthmatic mice fed with high fiber diet. Then, total lung cells from these asthmatic mice fed with either standard food or with fiber rich food were infected with RV ex vivo. Here, RV1b mRNA was found significantly reduced in the lung cells derived from fiber rich food fed mice as compared to those derived from standard food fed asthmatic mice. Looking for the mechanism, an increase in CD8+ T cells in RV infected spleen cells derived from fiber rich fed asthmatic mice, was observed. Conclusion Convalescent preschool asthmatic children after a symptomatic episode have less serum ß-Hydroxybutyric acid as compared to control and asthmatic children at baseline visit. Fiber rich diet associated with anti-inflammatory effects as well as anti-allergic effects by decreasing Type 2 and IgE mediated immune responses and inducing CD8+ memory effector T cells in a murine model of allergic asthma. Finally, ex vivo infection with Rhinovirus (RV) of total lung cells from asthmatic mice fed with fiber rich food led to a decreased RV load as compared to mice fed with standard food. Moreover, spleen cells derived from asthmatic mice fed with fiber rich food induced CD8+ T cells after ex vivo infection with RV. Clinical implications Dietary interventions with increased content in natural fibers like pectins would ameliorate asthma exacerbations. Moreover, respiratory infection in asthma downregulated SCFA in the gut contributing to asthma exacerbations.
Collapse
Affiliation(s)
- Alicia Schenzel
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Adriana Geiger
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Elvedina Nendel
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Zuqin Yang
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susanne Krammer
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anna Leberle
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ann-Kathrin Brunst
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sonja Trump
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susanne Mittler
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Manfred Rauh
- Children’s Hospital, Department of Allergy and Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Carol I. Geppert
- Institute of Pathology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Patrick Tausche
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katja Hohenberger
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ralf J. Rieker
- Institute of Pathology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Oliver Schieweck
- Laboratory of Clinic Medicine, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
50
|
Chen HZ, Zeng YY, Cai GX, Gu WD, Yang Y. Differential analysis of serum immunology and gut microbiota in patients with gastrointestinal diseases. Front Microbiol 2024; 15:1323842. [PMID: 38751718 PMCID: PMC11094713 DOI: 10.3389/fmicb.2024.1323842] [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: 10/18/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
Objective Gastric and intestinal diseases possess distinct characteristics although they are interconnected. The primary objective of this study was to investigate the pathogenesis of gastrointestinal diseases through different analyses of clinical characteristics, serum immunology, and gut microbiota in patients with gastrointestinal diseases. Methods We collected serum samples from 89 patients with gastrointestinal diseases and 9 healthy controls for immunological assessment, stool samples for DNA extraction, library construction, sequencing, as well as clinical data for subsequent analysis. Results Regarding clinical characteristics, there were significant differences between the disease group and the healthy control (HC) group, particularly in terms of age, cancer antigen 125 (CA125), cancer antigen 199 (CA199), alpha-fetoprotein (AFP), total bilirubin (TBIL) and indirect bilirubin (IBIL). The intestinal disease (ID) group exhibited the highest IL-6 level, which significantly differed from the stomach disease (SD) group (p < 0.05). In comparing the HC with the ID groups, significant differences in abundance were detected across 46 species. The HC group displayed a greater abundance of Clostridiales, Clostridia, Firmicutes, Bifidobacterium, Bifidobacteriaceae, Bifidobacteriales, Actinobacteria, Veillonellaceae, Longum, Copri, Megamonas and Callidus than other species. Similarly, when comparing the HC with the SD groups, significant differences in abundance were identified among 49 species, with only one species that the Lachnospiraceae in the HC group exhibited a higher abundance than others. Furthermore, certain clinical characteristics, such as CA125, CA199, glucose (Glu), creatine kinase-MB (CKMB) and interleukin-22 (IL-22), displayed positive correlations with enriched gut species in the ID and SD groups, while exhibiting a negative correlation with the HC group. Conclusion The disturbance in human gut microbiota is intimately associated with the development and progression of gastrointestinal diseases. Moreover, the gut microbiota in the HC group was found more diverse than that in the ID and SD groups, and there were significant differences in microbial species among the three groups at different classification levels. Notably, a correlation was identified between specific clinical characteristics (e.g., CA125, CA199, Glu, CKMB and IL-22) and gut microbiota among patients with gastrointestinal diseases.
Collapse
Affiliation(s)
- Huan Zhu Chen
- Biochemistry Teaching and Research Office of the Basic Department of the Medical College of Jiaying University, Meizhou, China
| | - Yu Yang Zeng
- Biochemistry Teaching and Research Office of the Basic Department of the Medical College of Jiaying University, Meizhou, China
| | - Guo Xiong Cai
- Laboratory Department of the Affiliated Hospital of the Medical College of Jiaying University, Meizhou, China
| | - Wei Dan Gu
- Laboratory Department of the Affiliated Hospital of the Medical College of Jiaying University, Meizhou, China
| | - YaLi Yang
- Biochemistry Teaching and Research Office of the Basic Department of the Medical College of Jiaying University, Meizhou, China
- Guangdong Provincial Key Laboratory of Conservation and Precision Utilization of Characteristic Agricultural Resources in Mountainous Areas, Meizhou, China
| |
Collapse
|