1
|
Wang H, Han J, Zhang XA. Interplay of m6A RNA methylation and gut microbiota in modulating gut injury. Gut Microbes 2025; 17:2467213. [PMID: 39960310 PMCID: PMC11834532 DOI: 10.1080/19490976.2025.2467213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiota undergoes continuous variations among individuals and across their lifespan, shaped by diverse factors encompassing diet, age, lifestyle choices, medication intake, and disease states. These microbial inhabitants play a pivotal role in orchestrating physiological metabolic pathways through the production of metabolites like bile acids, choline, short-chain fatty acids, and neurotransmitters, thereby establishing a dynamic "gut-organ axis" with the host. The intricate interplay between the gut microbiota and the host is indispensable for gut health, and RNA N6-methyladenosine modification, a pivotal epigenetic mark on RNA, emerges as a key player in this process. M6A modification, the most prevalent internal modification of eukaryotic RNA, has garnered significant attention in the realm of RNA epigenetics. Recent findings underscore its potential to influence gut microbiota diversity and intestinal barrier function by modulating host gene expression patterns. Conversely, the gut microbiota, through its impact on the epigenetic landscape of host cells, may indirectly regulate the recruitment and activity of RNA m6A-modifying enzymes. This review endeavors to delve into the biological functions of m6A modification and its consequences on intestinal injury and disease pathogenesis, elucidating the partial possible mechanisms by which the gut microbiota and its metabolites maintain host intestinal health and homeostasis. Furthermore, it also explores the intricate crosstalk between them in intestinal injury, offering a novel perspective that deepens our understanding of the mechanisms underlying intestinal diseases.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
2
|
Aminian-Dehkordi J, Dickson A, Valiei A, Mofrad MRK. MetaBiome: a multiscale model integrating agent-based and metabolic networks to reveal spatial regulation in gut mucosal microbial communities. mSystems 2025:e0165224. [PMID: 40183581 DOI: 10.1128/msystems.01652-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Mucosal microbial communities (MMCs) are complex ecosystems near the mucosal layers of the gut essential for maintaining health and modulating disease states. Despite advances in high-throughput omics technologies, current methodologies struggle to capture the dynamic metabolic interactions and spatiotemporal variations within MMCs. In this work, we present MetaBiome, a multiscale model integrating agent-based modeling (ABM), finite volume methods, and constraint-based models to explore the metabolic interactions within these communities. Integrating ABM allows for the detailed representation of individual microbial agents each governed by rules that dictate cell growth, division, and interactions with their surroundings. Through a layered approach-encompassing microenvironmental conditions, agent information, and metabolic pathways-we simulated different communities to showcase the potential of the model. Using our in-silico platform, we explored the dynamics and spatiotemporal patterns of MMCs in the proximal small intestine and the cecum, simulating the physiological conditions of the two gut regions. Our findings revealed how specific microbes adapt their metabolic processes based on substrate availability and local environmental conditions, shedding light on spatial metabolite regulation and informing targeted therapies for localized gut diseases. MetaBiome provides a detailed representation of microbial agents and their interactions, surpassing the limitations of traditional grid-based systems. This work marks a significant advancement in microbial ecology, as it offers new insights into predicting and analyzing microbial communities.IMPORTANCEOur study presents a novel multiscale model that combines agent-based modeling, finite volume methods, and genome-scale metabolic models to simulate the complex dynamics of mucosal microbial communities in the gut. This integrated approach allows us to capture spatial and temporal variations in microbial interactions and metabolism that are difficult to study experimentally. Key findings from our model include the following: (i) prediction of metabolic cross-feeding and spatial organization in multi-species communities, (ii) insights into how oxygen gradients and nutrient availability shape community composition in different gut regions, and (iii) identification of spatiallyregulated metabolic pathways and enzymes in E. coli. We believe this work represents a significant advance in computational modeling of microbial communities and provides new insights into the spatial regulation of gut microbiome metabolism. The multiscale modeling approach we have developed could be broadly applicable for studying other complex microbial ecosystems.
Collapse
Affiliation(s)
- Javad Aminian-Dehkordi
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
| | - Andrew Dickson
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
| | - Amin Valiei
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California, USA
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, California, USA
| |
Collapse
|
3
|
Yu H, Nie Y, Zhang B, Xue J, Xue K, Huang X, Zhang X. Creatine supplementation in largemouth bass (Micropterus salmoides) diets: Improving intestinal health and alleviating enteritis. FISH & SHELLFISH IMMUNOLOGY 2025; 159:110164. [PMID: 39894092 DOI: 10.1016/j.fsi.2025.110164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Creatine plays an important role in regulating intestinal epithelial cell energy metabolism, epithelial integrity, and intestinal barrier function. In this study, three feeds with varying creatine concentrations (0 %, 0.5 %, and 4 %, labeled CR0, CR0.5, and CR4, respectively) were formulated and administered to juvenile largemouth bass (Micropterus salmoides) for 8 weeks. Creatine-containing diets significantly improved growth performance and intestinal villus height. Microbiota analysis revealed that creatine-containing diets changed the beta diversity of gut microbes and increased the relative proportion of Cetobacterium. Enteritis was induced for 7 days using the corresponding feeds containing creatine and 2 % DSS (labeled CR0, DCR0, DCR0.5, and DCR4). Enteritis resulted in an increase in hif1α expression in the DCR0.5 and DCR4 groups and a significant increase expression of creatine transporter SLC6A8. QPCR and Western blotting of intestinal barrier-related genes (e.g., Claudin1, Claudin4, and ZO1), MUC2 immunohistochemistry, and PAS mucus staining were used to show intestinal barrier status, these results suggest that dietary creatine attenuates the extent of intestinal barrier damage. After TUNEL and KI67 immunofluorescence analyses of the intestine and detection of the expression of relevant genes at the protein and transcript levels, the results showed that dietary addition of creatine significantly alleviated intestinal apoptosis and cellular inflammatory responses due to DSS-induced enteritis. These findings indicate long-term dietary supplementation with creatine modulated the microbial composition of the intestinal lumen of juvenile largemouth bass, promoted intestinal health, and improved anti-inflammatory properties following enteritis induction. This study provides a theoretical foundation for largemouth bass feed formulation optimization and fish enteritis control.
Collapse
Affiliation(s)
- Haodong Yu
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Yukang Nie
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Boran Zhang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Jiajie Xue
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Kun Xue
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Xixuan Huang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Xuezhen Zhang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; Research Institute of Huanong-Tianchen, Wuhan, 430070, China.
| |
Collapse
|
4
|
Kasahara K, Seiffarth J, Stute B, von Lieres E, Drepper T, Nöh K, Kohlheyer D. Unveiling microbial single-cell growth dynamics under rapid periodic oxygen oscillations. LAB ON A CHIP 2025. [PMID: 40159892 DOI: 10.1039/d5lc00065c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Microbial metabolism and growth are tightly linked to oxygen (O2). Microbes experience fluctuating O2 levels in natural environments; however, our understanding of how cells respond to fluctuating O2 over various time scales remains limited due to challenges in observing microbial growth at single-cell resolution under controlled O2 conditions and in linking individual cell growth with the specific O2 microenvironment. We performed time-resolved microbial growth analyses at single-cell resolution under a temporally controlled O2 supply. A multilayer microfluidic device was developed, featuring a gas supply above a cultivation layer, separated by a thin membrane enabling efficient gas transfer. This platform allows microbial cultivation under constant, dynamic, and oscillating O2 conditions. Automated time-lapse microscopy and deep-learning-based image analysis provide access to spatiotemporally resolved growth data at the single-cell level. O2 switching within tens of seconds, coupled with precise microenvironment monitoring, allows us to accurately correlate cellular growth with local O2 concentrations. Growing Escherichia coli microcolonies subjected to varying O2 oscillation periods show distinct growth dynamics characterized by response and recovery phases. The comprehensive growth data and insights gained from our unique platform are a crucial step forward to systematically study cell response and adaptation to fluctuating O2 environments at single-cell resolution.
Collapse
Affiliation(s)
- Keitaro Kasahara
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
- Computational Systems Biotechnology (AVT.CSB), RWTH Aachen University, Aachen, Germany
| | - Johannes Seiffarth
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
- Computational Systems Biotechnology (AVT.CSB), RWTH Aachen University, Aachen, Germany
| | - Birgit Stute
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Eric von Lieres
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
- Computational Systems Biotechnology (AVT.CSB), RWTH Aachen University, Aachen, Germany
| | - Thomas Drepper
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Katharina Nöh
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Dietrich Kohlheyer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| |
Collapse
|
5
|
Fanizza F, Perottoni S, Boeri L, Donnaloja F, Negro F, Pugli F, Forloni G, Giordano C, Albani D. A gut-brain axis on-a-chip platform for drug testing challenged with donepezil. LAB ON A CHIP 2025; 25:1854-1874. [PMID: 40052475 DOI: 10.1039/d4lc00273c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Current drug development pipelines are time-consuming and prone to a significant percentage of failure, partially due to the limited availability of advanced human preclinical models able to better replicate the in vivo complexity of our body. To contribute to an advancement in this field, we developed an in vitro multi-organ-on-a-chip system, that we named PEGASO platform, which enables the dynamic culturing of human cell-based models relevant for drug testing. The PEGASO platform is composed of five independent connected units, which are based on a previously developed millifluidic organ-on-a-chip device (MINERVA 2.0), hosting human primary cells and iPSC-derived cells recapitulating key biological features of the gut, immune system, liver, blood-brain-barrier and brain that were fluidically connected and challenged to model the physiological passage of donepezil, a drug prescribed for Alzheimer's disease. The nutrient medium flow rate of the connected units was tuned to obtain suitable oxygenation and shear stress values for the cells cultured in dynamic condition. A computational model was at first developed to simulate donepezil transport within the platform and to assess the drug amount reaching the last organ-on-a-chip. Then, we demonstrated that after 24 hours of donepezil administration, the drug was actually transported though the cell-based models of the platform which in turn were found viable and functional. Donepezil efficacy was confirmed by the decreased acetylcholinesterase activity at the brain model and by the increased expression of a donepezil-relevant multi-drug transporter (P-gp). Overall, the PEGASO platform is an innovative in vitro tool for drug screening and personalized medicine applications which holds the potential to be translated to preclinical research and improve new drug development pipelines.
Collapse
Affiliation(s)
- Francesca Fanizza
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Francesca Donnaloja
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Francesca Negro
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Francesca Pugli
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, Milan, Italy.
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
6
|
Kaufman D, Winkler S, Heuer C, Shibli A, Snezhko A, Livshits GI, Bahnemann J, Ben-Yoav H. Automated electrochemical oxygen sensing using a 3D-printed microfluidic lab-on-a-chip system. LAB ON A CHIP 2025; 25:1404-1415. [PMID: 39763425 DOI: 10.1039/d4lc00962b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Dissolved oxygen is crucial for metabolism, growth, and other complex physiological and pathological processes; however, standard physiological models (such as organ-on-chip systems) often use ambient oxygen levels, which do not reflect the lower levels that are typically found in vivo. Additionally, the local generation of reactive oxygen species (ROS; a key factor in physiological systems) is often overlooked in biology-mimicking models. Here, we present a microfluidic system that integrates electrochemical dissolved oxygen sensors with lab-on-a-chip technology to monitor the physiological oxygen concentrations and generate hydrogen peroxide (H2O2; a specific ROS). This microfluidic lab-on-a-chip system was fabricated using high-resolution 3D printing technology in a one-step process. It incorporates a micromixer, an on-chip bubble-trap, an electrochemical cell with fabricated gold or platinum black-coated working electrodes as well as an Ag/AgCl reference electrode, and a commercial optical oxygen sensor for validation. This device enables an automated variation of the oxygen levels as well as sensitive electrochemical oxygen monitoring (limit of detection = 11.9 ± 0.3 μM), with a statistically significant correlation with the optical sensor. The proposed system can serve as a tool to characterize and evaluate custom-made electrodes. Indeed, we envision that in the future it will be used to regulate dissolved oxygen levels and oxygen species in real time in organ-on-chip systems.
Collapse
Affiliation(s)
- Daniel Kaufman
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Steffen Winkler
- Institute of Physics, University of Augsburg, Universitätsstraße 1, 86159 Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 8615, Augsburg, Germany
| | - Christopher Heuer
- Institute of Physics, University of Augsburg, Universitätsstraße 1, 86159 Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 8615, Augsburg, Germany
| | - Ahed Shibli
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Alexander Snezhko
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Gideon I Livshits
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Janina Bahnemann
- Institute of Physics, University of Augsburg, Universitätsstraße 1, 86159 Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 8615, Augsburg, Germany
| | - Hadar Ben-Yoav
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| |
Collapse
|
7
|
Yang H, Xia R, Teame T, Meng D, Li S, Wang T, Ding Q, Yao Y, Xu X, Yang Y, Ran C, Zhang Y, Li S, Niemann B, Guan LL, Zhang Z, Zhou Z. Activation of Gut Microbiota-Hypoxia-Inducible Factor α Axis Effectively Restores Resistance to Aeromonas veronii Caused by Improper Administration of AiiO-AIO6. J Nutr 2025:S0022-3166(25)00157-9. [PMID: 40064423 DOI: 10.1016/j.tjnut.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Feeding adult zebrafish a diet supplemented with quenching enzyme AiiO-AIO6 for 3 wk improved the growth performance and disease resistance. However, when the feeding period was extended to 8 wk, zebrafish's disease resistance to Aeromonas veronii decreased. OBJECTIVES We investigated the mechanisms of the reduced disease resistance of zebrafish induced by feeding on an AiiO-AIO6 supplemented diet for a long term (8 wk) and assessed the effectiveness of feed additives in restoring the low disease resistance. METHODS One-month-old (adult) zebrafish were fed with a basal diet and the basal diet supplemented with AiiO-AIO6 (10 U/g) for 8 wk (experiment 1). Furthermore, the zebrafish larvae model (experiment 2) was developed and used to study the mechanisms of how AiiO-AIO6 affected disease resistance (experiment 3). We also investigated the effectiveness of selected prebiotic tributyrin, β-glucan or mannan in activating gut microbiota- hypoxia-inducible factor α (hif1α) to restore the low disease resistance of adult zebrafish fed with AiiO-AIO6 for 8 wk (experiment 4). Lastly, the effects of Bacillus subtilis in activating the gut microbiota-hif1α and improving the low disease resistance of zebrafish larvae induced by AiiO-AIO6 were examined (experiment 5). RESULTS Feeding adult zebrafish with AiiO-AIO6 for 8 wk promoted growth but disordered the gut microbiota and reduced disease resistance. The zebrafish larvae model confirmed that feeding AiiO-AIO6 for 2 d increased disease resistance, whereas 7 d decreased the resistance by suppressing hif1α. Using a germ-free zebrafish larvae model, we also demonstrated that AiiO-AIO6-induced gut microbiota mediated inhibition of hif1α. Furthermore, zebrafish fed on the AiiO-AIO6-containing diet supplement with tributyrin, β-glucan, mannan, or Bacillus subtilis activated the gut microbiota-hif1α axis to reverse the low resistance caused by AiiO-AIO6. CONCLUSIONS Activating the gut microbiota-hif1α axis has a vital role in improving intestinal health and restores the low resistance to Aeromonas veronii caused by improper administration of dietary AiiO-AIO6 in zebrafish.
Collapse
Affiliation(s)
- Hongwei Yang
- Institute of Feed Research, China-Norway Joint Lab on Fish Gut Microbiota, Chinese Academy of Agricultural Sciences, Beijing, China; Institute of Marine Sciences, Shantou University, Shantou, China
| | - Rui Xia
- Institute of Feed Research, China-Norway Joint Lab on Fish Gut Microbiota, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tsegay Teame
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China; Tigray Agricultural Research Institute (TARI), Mekelle, Tigray, Ethiopia
| | - Delong Meng
- Institute of Feed Research, China-Norway Joint Lab on Fish Gut Microbiota, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shenghui Li
- Institute of Feed Research, China-Norway Joint Lab on Fish Gut Microbiota, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tiantian Wang
- Institute of Feed Research, China-Norway Joint Lab on Fish Gut Microbiota, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qianwen Ding
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuanyuan Yao
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoqing Xu
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalin Yang
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chao Ran
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yaqing Zhang
- Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Shengkang Li
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Benjamin Niemann
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada
| | - Le Luo Guan
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada
| | - Zhen Zhang
- Institute of Feed Research, Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China; Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada.
| | - Zhigang Zhou
- Institute of Feed Research, China-Norway Joint Lab on Fish Gut Microbiota, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
8
|
Buehler A, Brown EL, Eckstein M, Thoma OM, Wachter F, Mandelbaum H, Ludwig P, Claßen M, Oraiopoulou ME, Rother U, Neurath MF, Woelfle J, Waldner MJ, Friedrich O, Knieling F, Bohndiek SE, Regensburger AP. Guided Multispectral Optoacoustic Tomography for 3D Imaging of the Murine Colon. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413434. [PMID: 39836529 PMCID: PMC11905093 DOI: 10.1002/advs.202413434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/07/2025] [Indexed: 01/23/2025]
Abstract
Multispectral optoacoustic tomography is a promising medical imaging modality that combines light and sound to provide molecular imaging information at depths of several centimeters, based on the optical absorption of endogenous chromophores, such as hemoglobin. Assessment of inflammatory bowel disease has emerged as a promising clinical application of optoacoustic tomography. In this context, preclinical studies in animal models are essential to identify novel disease-specific imaging biomarkers and understand findings from emerging clinical pilot studies, however to-date, these studies have been limited by the precise identification of the bowel wall. Herein, a transrectal-absorber guide is applied, serving as a high-contrast landmark for 3D optoacoustic tomography of the colon. This study shows that guided multispectral optoacoustic tomography is able to measure changes in blood oxygenation status over the course of acute, chemically-induced colitis in mice and correlates with standard disease activity scores. This novel approach depicts intestinal hemoglobin composition non-invasively during murine inflammation. These results underscore the potential for optoacoustic imaging in translational inflammatory bowel disease research.
Collapse
Affiliation(s)
- Adrian Buehler
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Emma L Brown
- Department of Physics and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Oana-Maria Thoma
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie DZI, University Hospital Erlangen, 91054, Erlangen, Germany
| | - Felix Wachter
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Henriette Mandelbaum
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Petra Ludwig
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Merle Claßen
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Mariam-Eleni Oraiopoulou
- Department of Physics and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Ulrich Rother
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie DZI, University Hospital Erlangen, 91054, Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Maximilian J Waldner
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie DZI, University Hospital Erlangen, 91054, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Ferdinand Knieling
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Sarah E Bohndiek
- Department of Physics and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Adrian P Regensburger
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| |
Collapse
|
9
|
Yang Z, Lin Z, You Y, Zhang M, Gao N, Wang X, Peng J, Wei H. Gut Microbiota-Derived Hyocholic Acid Enhances Type 3 Immunity and Protects Against Salmonella enterica Serovar Typhimurium in Neonatal Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412071. [PMID: 39737849 PMCID: PMC11905087 DOI: 10.1002/advs.202412071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/18/2024] [Indexed: 01/01/2025]
Abstract
This study investigates how microbiome colonization influences the development of intestinal type 3 immunity in neonates. The results showed that reduced oxygen levels in the small intestine of neonatal rats induced by Saccharomyces boulardii accelerated microbiome colonization and type 3 immunity development, which protected against Salmonella enterica serovar Typhimurium infection. Microbiome maturation increased the abundance of microbiome-encoded bile salt hydrolase (BSH) genes and hyocholic acid (HCA) levels. Furthermore, reducing oxygen levels in the intestine increased the abundance of Limosilactobacillus reuteri, a bacterium encoding BSH, and promoted intestinal type 3 immunity. However, inhibition of BSH blocked the L. reuteri-induced development of intestinal type 3 immunity. Mechanistically, HCA promoted the development of gamma-delta T cells and type 3 innate lymphoid cells by stabilizing the mRNA expression of RAR-related orphan receptor C via the farnesoid X receptor-WT1-associated protein-N6-methyl-adenosine axis. These results reveal that gut microbiota-derived HCA plays a crucial role in promoting the development of intestinal type 3 immunity in neonates. This discovery introduces potential therapeutic avenues for strengthening intestinal immunity in early life or treating bacterial infections by targeting microbial metabolites.
Collapse
Affiliation(s)
- Zhipeng Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhiyuan Lin
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yaojie You
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mei Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ning Gao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xinru Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| |
Collapse
|
10
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
11
|
Soranno DE, Coopersmith CM, Brinkworth JF, Factora FNF, Muntean JH, Mythen MG, Raphael J, Shaw AD, Vachharajani V, Messer JS. A review of gut failure as a cause and consequence of critical illness. Crit Care 2025; 29:91. [PMID: 40011975 DOI: 10.1186/s13054-025-05309-7] [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: 10/03/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence factors appear. Intestinal permeability increases, allowing for translocation of bacteria and/or bacterial products. Epithelial function is altered at a cellular level and homeostasis of the epithelial monolayer is compromised by increased intestinal epithelial cell death and decreased proliferation. Gut immunity is impaired with simultaneous activation of maladaptive pro- and anti-inflammatory signals leading to both tissue damage and susceptibility to infections. Additionally, splanchnic vasoconstriction leads to decreased blood flow with local ischemic changes. Together, these interrelated elements of gastrointestinal dysfunction drive and then perpetuate multi-organ dysfunction syndrome. Despite the clear importance of maintaining gut homeostasis, there are very few reliable measures of gut function in critical illness. Further, while multiple therapeutic strategies have been proposed, most have not been shown to conclusively demonstrate benefit, and care is still largely supportive. The key role of the gut in critical illness was the subject of the tenth Perioperative Quality Initiative meeting, a conference to summarize the current state of the literature and identify key knowledge gaps for future study. This review is the product of that conference.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Jessica F Brinkworth
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Faith N F Factora
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Julia H Muntean
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Monty G Mythen
- Perioperative Medicine, University College London, London, England
| | - Jacob Raphael
- Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew D Shaw
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Vidula Vachharajani
- Department of Pulmonary and Critical Care, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jeannette S Messer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
12
|
Gregory AL, Bussan HE, Topf MA, Hryckowian AJ. Impacts of perR on oxygen sensitivity, gene expression, and murine infection in Clostridioides difficile 630∆ erm. J Bacteriol 2025; 207:e0046824. [PMID: 39846733 PMCID: PMC11841134 DOI: 10.1128/jb.00468-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Clostridioides difficile infection (CDI), characterized by colitis and diarrhea, afflicts approximately half a million people in the USA every year, burdening both individuals and the healthcare system. C. difficile 630Δerm is an erythromycin-sensitive variant of the clinical isolate C. difficile 630 and is commonly used in the C. difficile research community due to its genetic tractability. 630Δerm possesses a point mutation in perR, an autoregulated transcriptional repressor that regulates oxidative stress resistance genes. This point mutation results in a constitutively de-repressed PerR operon in 630Δerm. To address the impacts of perR on phenotypes relevant for oxygen tolerance and relevant to a murine model of CDI, we corrected the point mutant to restore PerR function in 630∆erm (herein, 630∆erm perRWT). We demonstrate that there is no difference in growth between 630Δerm and 630Δerm perRWT under anaerobic conditions or when exposed to concentrations of O2 that mimic those found near the surface of the colonic epithelium. However, 630∆erm perRWT is more sensitive to ambient oxygen than 630∆erm, which coincides with alterations in expression of a variety of perR-dependent and perR-independent genes. Finally, we show that 630∆erm and 630∆erm perRWT do not differ in their ability to infect and cause disease in a well-established murine model of CDI. Together, these data support the hypothesis that the perR mutation in 630∆erm arose as a result of exposure to ambient oxygen and that the perR mutation in 630∆erm is unlikely to impact CDI-relevant phenotypes in laboratory studies.IMPORTANCEClostridioides difficile is a diarrheal pathogen and a major public health concern. To improve humans' understanding of C. difficile, a variety of C. difficile isolates are used in research, including C. difficile 630Δerm. 630Δerm is a derivative of the clinical isolate 630 and is commonly studied because it is genetically manipulable. Previous work showed that a mutation in perR in 630Δerm results in a dysregulated oxidative stress response, but no work has been done to characterize perR-dependent effects on the transcriptome or to determine impacts of perR during infection. Here, we identify transcriptomic differences between 630∆erm and 630∆erm perRWT exposed to ambient oxygen and demonstrate that there is no strain-based difference in burdens in murine C. difficile infection.
Collapse
Affiliation(s)
- Anna L. Gregory
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hailey E. Bussan
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Madeline A. Topf
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
13
|
Weis JA, Rauh JL, Ellison MA, Cruz-Diaz N, Yamaleyeva LM, Welch CD, Zeller KA, Weis VG. Photoacoustic imaging for non-invasive assessment of biomarkers of intestinal injury in experimental necrotizing enterocolitis. Pediatr Res 2025; 97:169-177. [PMID: 38914761 PMCID: PMC11666804 DOI: 10.1038/s41390-024-03358-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/24/2024] [Accepted: 05/27/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is an often-lethal disease of the premature infant intestinal tract, exacerbated by significant diagnostic difficulties. In NEC, the intestine exhibits hypoperfusion and dysmotility, contributing to disease pathogenesis. However, these features cannot be accurately and quantitively assessed with current imaging modalities. We have previously demonstrated the ability of photoacoustic imaging (PAI) to non-invasively assess intestinal tissue oxygenation and motility in a healthy neonatal rat model. METHODS In this first-in-disease application, we evaluated NEC using PAI to assess intestinal health biomarkers in an experimental model of NEC. NEC was induced in neonatal rats from birth to 4-days. Healthy breastfed (BF) and NEC rat pups were imaged at 2- and 4-days. RESULTS Intestinal tissue oxygen saturation was measured with PAI, and NEC pups showed significant decreases at 2- and 4-days. Ultrasound and PAI cine recordings were used to capture intestinal peristalsis and contrast agent transit within the intestine. Intestinal motility, assessed using computational intestinal deformation analysis, demonstrated significant reductions in both early and established NEC. NEC damage was confirmed with histology and dysmotility was confirmed by small intestinal transit assay. CONCLUSION This preclinical study presents PAI as an emerging diagnostic imaging modality for intestinal disease assessment in premature infants. IMPACT Necrotizing enterocolitis (NEC) is a devastating intestinal disease affecting premature infants with significant mortality. NEC presents significant clinical diagnostic difficulties, with limited diagnostic confidence complicating timely and effective interventional efforts. This study is an important foundational first-in-disease preclinical study that establishes the utility for PAI to detect changes in intestinal tissue oxygenation and intestinal motility with NEC disease induction and progression. This study demonstrates the feasibility and exceptional promise for the use of PAI to non-invasively assess oxygenation and motility in the healthy and diseased infant intestine.
Collapse
Affiliation(s)
- Jared A Weis
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Comprehensive Cancer Center, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA.
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, USA.
| | - Jessica L Rauh
- Section of Pediatric Surgery, Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - Maryssa A Ellison
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Nildris Cruz-Diaz
- Department of Surgery/Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Liliya M Yamaleyeva
- Department of Surgery/Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Cherrie D Welch
- Division of Neonatology, Department of Pediatrics, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - Kristen A Zeller
- Section of Pediatric Surgery, Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - Victoria G Weis
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA.
- Department of Pediatrics, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA.
| |
Collapse
|
14
|
Srivastav S, Biswas A, Anand A. Interplay of niche and respiratory network in shaping bacterial colonization. J Biol Chem 2025; 301:108052. [PMID: 39662826 PMCID: PMC11742581 DOI: 10.1016/j.jbc.2024.108052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024] Open
Abstract
The human body is an intricate ensemble of prokaryotic and eukaryotic cells, and this coexistence relies on the interplay of many biotic and abiotic factors. The inhabiting microbial population has to maintain its physiological homeostasis under highly dynamic and often hostile host environments. While bacterial colonization primarily relies on the metabolic suitability for the niche, there are reports of active remodeling of niche microenvironments to create favorable habitats, especially in the context of pathogenic settlement. Such physiological plasticity requires a robust metabolic system, often dependent on an adaptable energy metabolism. This review focuses on the respiratory electron transport system and its adaptive consequences within the host environment. We provide an overview of respiratory chain plasticity, which allows pathogenic bacteria to niche-specify, niche-diversify, mitigate inflammatory stress, and outcompete the resident microbiota. We have reviewed existing and emerging knowledge about the role of respiratory chain components responsible for the entry and exit of electrons in influencing the pathogenic outcomes.
Collapse
Affiliation(s)
- Stuti Srivastav
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Arpita Biswas
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Amitesh Anand
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India.
| |
Collapse
|
15
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [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: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
16
|
Zhang Y, Yan M, Yue Y, Cheng Y. Hypoxia-Inducible Factor-1α Modulates the Toll-Like Receptor 4/Nuclear Factor Kappa B Signaling Pathway in Experimental Necrotizing Enterocolitis. Mediators Inflamm 2024; 2024:4811500. [PMID: 39719983 PMCID: PMC11668547 DOI: 10.1155/mi/4811500] [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: 07/16/2024] [Accepted: 11/11/2024] [Indexed: 12/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease observed in premature infants, characterized by intestinal ischemia and inflammation. Hypoxia-inducible factor-1 alpha (HIF-1α), a master regulator of the cellular response to hypoxia and ischemia, plays a critical role in NEC pathogenesis. However, the precise mechanisms by which HIF-1α influences the intestines in NEC remain poorly understood. Herein, we aimed to explore the role of HIF-1α in NEC using a transgenic mouse model. We induced NEC in neonatal mice from postnatal day 5 to 9, and various parameters, including intestinal injury, oxidative stress, inflammatory responses, intestinal epithelial cell (IEC) proliferation, and apoptosis, were assessed. The results confirmed that the absence of intestinal epithelial HIF-1α increased the susceptibility of mice to NEC-induced intestinal injury, as evidenced by increased oxidative stress, inflammatory responses, apoptosis, and inhibition of proliferation. Additionally, we observed an upregulation of the Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway specifically in the intestines of mice lacking HIF-1α in IECs (HIF-1αΔIEC) with NEC. These findings provide crucial insights into the role of HIF-1α in regulating intestinal oxidative stress and inflammation to maintain intestinal homeostasis, highlighting its association with the TLR4-NF-κB signaling pathway. Furthermore, these insights might lead to the identification of novel therapeutic targets for the treatment of NEC.
Collapse
Affiliation(s)
- Yunfei Zhang
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Mei Yan
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Yingbin Yue
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Yongfeng Cheng
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| |
Collapse
|
17
|
Fu Y, Ding X, Zhang M, Feng C, Yan Z, Wang F, Xu J, Lin X, Ding X, Wang L, Fan Y, Li T, Yin Y, Liang X, Xu C, Chen S, Pulous FE, Gennert D, Pun FW, Kamya P, Ren F, Aliper A, Zhavoronkov A. Intestinal mucosal barrier repair and immune regulation with an AI-developed gut-restricted PHD inhibitor. Nat Biotechnol 2024:10.1038/s41587-024-02503-w. [PMID: 39663371 DOI: 10.1038/s41587-024-02503-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Hypoxia-inducible factor prolyl hydroxylase (PHD) inhibitors have been approved for treating renal anemia yet have failed clinical testing for inflammatory bowel disease because of a lack of efficacy. Here we used a multimodel multimodal generative artificial intelligence platform to design an orally gut-restricted selective PHD1 and PHD2 inhibitor that exhibits favorable safety and pharmacokinetic profiles in preclinical studies. ISM012-042 restores intestinal barrier function and alleviates gut inflammation in multiple experimental colitis models.
Collapse
Affiliation(s)
- Yanyun Fu
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
- Insilico Medicine Hong Kong, Ltd., Hong Kong, China
- Insilico Medicine AI, Ltd., Abu Dhabi, UAE
| | - Xiao Ding
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
- Insilico Medicine AI, Ltd., Abu Dhabi, UAE
| | - Man Zhang
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
- Insilico Medicine Hong Kong, Ltd., Hong Kong, China
| | - Chunlei Feng
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Ziqi Yan
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Feng Wang
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Jianyu Xu
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Xiaoxia Lin
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Xiaoyu Ding
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Ling Wang
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Yaya Fan
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Taotao Li
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Yushu Yin
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Xing Liang
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Chenxi Xu
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Shan Chen
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
| | - Fadi E Pulous
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine US, Inc., New York, NY, USA
| | - David Gennert
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine US, Inc., New York, NY, USA
| | - Frank W Pun
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Hong Kong, Ltd., Hong Kong, China
| | - Petrina Kamya
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Canada, Inc., Montréal, Québec, Canada
| | - Feng Ren
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine Shanghai, Ltd., Shanghai, China
- Insilico Medicine Hong Kong, Ltd., Hong Kong, China
- Insilico Medicine AI, Ltd., Abu Dhabi, UAE
| | - Alex Aliper
- Insilico Medicine US, Inc., Boston, MA, USA
- Insilico Medicine AI, Ltd., Abu Dhabi, UAE
| | - Alex Zhavoronkov
- Insilico Medicine US, Inc., Boston, MA, USA.
- Insilico Medicine Shanghai, Ltd., Shanghai, China.
- Insilico Medicine Hong Kong, Ltd., Hong Kong, China.
- Insilico Medicine US, Inc., New York, NY, USA.
| |
Collapse
|
18
|
Su L, Huang S, Huang Y, Bai X, Zhang R, Lei Y, Wang X. Effects of Eimeria challenge on growth performance, intestine integrity, and cecal microbial diversity and composition of yellow broilers. Poult Sci 2024; 103:104470. [PMID: 39504824 PMCID: PMC11570961 DOI: 10.1016/j.psj.2024.104470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
The invasion of Eimeria causes damage to the intestinal barrier, nutrient leakage, and microbial imbalance in poultry. We aimed to investigate the effects of Eimeria infection on growth performance, intestinal integrity, and cecal microbial diversity and composition of yellow broilers. A total of 180 male yellow broilers were randomly divided into an unchallenged control and an Eimeria challenge treatment group within 18 floor pens (10 chicks/pen, 9 replicate pens/group). On day 10, 90 chicks received a cocktail of E. maxima, E. acervulina, and E. tenella oocysts (105/chick) to induce coccidial infection, and the other 90 received an aliquot of PBS. The Eimeria challenge resulted in increased bird feed consumption and FCR from day 11 to 21 (all P < 0.01). Higher fecal Eimeria counts, duodenal, jejunal, and cecal lesions were observed in the challenge group on day 12, 15, 15, 18 respectively (all P < 0.05). Furthermore, the infected birds had larger livers and small intestines, deeper villus crypt, and decreased expression of Claudin-1 on day 21 (all P < 0.05). The 16S rRNA sequencing indicated that alpha diversity (Sobs, Shannon, Simpson, Ace, or Chao) of cecal microbials was not affected by Eimeria challenge (all P > 0.05). However, the PCoA and LEfSe analyses indicated that the Eimeria challenge altered microbial distribution by decreasing the abundance of Firmicutes and enriching the abundance of Proteobacteria at the phylum level. At the genus level, Clostridia vadin BB60 and Lachnospiraceae NK4A136 group were reduced, while Escherichia-Shigella were enriched in the challenged yellow broilers (all P < 0.05). Correlation analyses demonstrated that the birds with higher Lachonospiraceae NK4A136 group and Clostridia vadin BB60, and lower Escherichia-Shigella in their cecal content gained more BW and reached a lower FCR from day 11 to 21 (all P < 0.05). In conclusion, Eimeria infection compromised feed efficiency of yellow broilers by damaging intestinal barrier and shifting cecal microbiota towards colonizers associated with poor performance. Restoring the dysbiotic microbiome could be a potential strategy for improving feed efficiency in yellow broilers under coccidial challenge.
Collapse
Affiliation(s)
- Linjie Su
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China, 610041
| | - Shuping Huang
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China, 610041
| | - Yanling Huang
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China, 610041; Key Laboratory of Qinghai Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Xue Bai
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China, 610041; Key Laboratory of Qinghai Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Runhui Zhang
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China, 610041
| | - Yan Lei
- SiXie Enterprise Management Consulting Co. LTD, Chengdu 610074, China
| | - Xi Wang
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China, 610041; Key Laboratory of Qinghai Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
19
|
He X, Ji J, Liu C, Luo Z, Tang J, Yan H, Guo L. Body mass index and weight loss as risk factors for poor outcomes in patients with idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Ann Med 2024; 56:2311845. [PMID: 38301276 PMCID: PMC10836485 DOI: 10.1080/07853890.2024.2311845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/24/2024] [Indexed: 02/03/2024] Open
Abstract
OBJECTIVE The association between nutritional status and prognosis of idiopathic pulmonary fibrosis (IPF) remains unclear. This systematic review and meta-analysis aimed to explore the effect of body mass index (BMI) and weight loss on the prognosis of IPF patients. METHODS We accumulated studies on IPF, BMI, and weight loss from databases including PubMed, Embase, Web of science, Scopus, Ovid and Cochrane Library up to 4 August 2023. Using Cox proportional hazard regression model for subgroup analysis, hazard ratio (HR) and 95% confidence intervals (CI) for BMI in relation to mortality, acute exacerbation (AE), and hospitalization in IPF patients were calculated, and HR, odds ratio (OR), and 95% CI for weight loss corresponding to IPF patient mortality were assessed. Sensitivity analysis was peformed by eliminating every study one by one, and publication bias was judged by Egger's test and trim-and-fill method. RESULTS A total of 34 eligible studies involving 18,343 IPF patients were included in the meta-analysis. The pooled results by univariate Cox regression analysis showed that baseline BMI was a predictive factor for IPF mortality (HR = 0.93, 95%CI = [0.91, 0.94]). Furthermore, the results by the multivariable regression model indicated that baseline BMI was an independent risk factor for predicting IPF mortality (HR = 0.94, 95%CI = [0.91, 0.98]). Weight loss was identified as a risk factor for IPF mortality (HR = 2.74, 95% CI = [2.12, 3.54]; OR = 4.51, 95% CI = [1.72, 11.82]) and there was no predictive value of BMI for acute exacerbation (HR = 1.00, 95% CI= [0.93, 1.07]) or hospitalization (HR = 0.95, 95% CI = [0.89, 1.02]). CONCLUSION Low baseline BMI and weight loss in the course of IPF may indicate a high risk of mortality in patients with IPF, so it is meaningful to monitor and manage the nutritional status of IPF patients, and early intervention should be conducted for low BMI and weight loss.
Collapse
Affiliation(s)
- Xing He
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Department of Pulmonary and Critical Care Medicine, Cheng Du Qing Cheng Mt. Hospital, Chongzhou City, Chengdu, Sichuan Province, China
| | - Jiaqi Ji
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Chi Liu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Zeli Luo
- Department of Critical Care Medicine, Wenjiang District People’s Hospital, Chengdu, Sichuan Province, China
| | - Jialong Tang
- Department of Respiratory and Critical Care Medicine, Jiange County People’s Hospital, Guangyuan, Sichuan Province, China
| | - Haiying Yan
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Department of Pulmonary and Critical Care Medicine, Cheng Du Qing Cheng Mt. Hospital, Chongzhou City, Chengdu, Sichuan Province, China
| | - Lu Guo
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| |
Collapse
|
20
|
Di Maso AM, Ruiz C. Physiological Effects of TolC-Dependent Multidrug Efflux Pumps in Escherichia coli: Impact on Motility and Growth Under Stress Conditions. Microbiologyopen 2024; 13:e70006. [PMID: 39529380 PMCID: PMC11554990 DOI: 10.1002/mbo3.70006] [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: 07/27/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Enterobacteriaceae possess eight TolC-dependent multidrug efflux pumps: AcrAB-TolC, AcrAD-TolC, AcrEF-TolC, MdtEF-TolC, MdtABC-TolC, EmrAB-TolC, EmrYK-TolC, and MacAB-TolC, which efflux bile salts, antibiotics, metabolites, or other compounds. However, our understanding of their physiological roles remains limited, especially for less-studied pumps like EmrYK-TolC. In this study, we tested the effects on swimming motility and growth under stress conditions of Escherichia coli mutants individually deleted for each inner-membrane transporter component of all eight TolC-dependent pumps, a mutant deleted for the AcrB-accessory protein AcrZ, and a mutant simultaneously deleted for all eight pumps (ΔtolC). We found that all mutants tested, except the ΔemrY and ΔacrZ mutants, displayed increased swimming motility. Additionally, the loss of each individual TolC-dependent pump or AcrZ did not reduce growth and sometimes even enhanced it compared to the parental strain under various growth conditions: temperature (LB at 25, 30, 37, and 42°C), pH (LB at pH 6.0, 7.4, and 9.0; and LB buffered to pH 6.0, 7.4, and 8.25), LB with limited air exchange, and nutritional stress (M9-glucose or M9-glycerol). In contrast, the ΔtolC mutant grew significantly slower than the parental strain under all conditions tested except in LB-TRIS pH 7.4 and LB with limited air exchange. Overall, these findings indicate that while individual TolC-dependent pumps are generally dispensable for growth under many stress conditions in the absence of antimicrobials, possibly due to their partially overlapping substrate profiles, TolC-dependent efflux is required for maximal growth under most conditions.
Collapse
Affiliation(s)
- Amanda M. Di Maso
- Department of BiologyCalifornia State University NorthridgeNorthridgeCaliforniaUSA
| | - Cristian Ruiz
- Department of BiologyCalifornia State University NorthridgeNorthridgeCaliforniaUSA
| |
Collapse
|
21
|
Zhang HY, Shu YQ, Li Y, Hu YL, Wu ZH, Li ZP, Deng Y, Zheng ZJ, Zhang XJ, Gong LF, Luo Y, Wang XY, Li HP, Liao XP, Li G, Ren H, Qiu W, Sun J. Metabolic disruption exacerbates intestinal damage during sleep deprivation by abolishing HIF1α-mediated repair. Cell Rep 2024; 43:114915. [PMID: 39527478 DOI: 10.1016/j.celrep.2024.114915] [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: 02/26/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Sleep deprivation (SD) has been reported to induce intestinal damage by several mechanisms, yet its role in modulating epithelial repair remains unclear. In this study, we find that chronic SD leads to colonic damage through continuous hypoxia. However, HIF1α, which generally responds to hypoxia to modulate barrier integrity, was paradoxically dysregulated in the colon. Further investigation revealed that a metabolic disruption during SD causes accumulation of α-ketoglutarate in the colon. The excessive α-ketoglutarate degrades HIF1α protein through PHD2 (prolyl hydroxylase 2) to abolish the intestinal repair functions of HIF1α. Collectively, these findings provide insights into how SD can exacerbate intestinal damage by fine-tuning metabolism to abolish HIF1α-mediated repair.
Collapse
Affiliation(s)
- Hai-Yi Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ya-Qing Shu
- The Third Affiliated Hospital of Sun Yat-sen University, Department of Neurology, Guangzhou, China
| | - Yan Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ya-Lin Hu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhi-Hong Wu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhi-Peng Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yao Deng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zi-Jian Zheng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Liu-Fei Gong
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yang Luo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Yu Wang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | | | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Gong Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wei Qiu
- The Third Affiliated Hospital of Sun Yat-sen University, Department of Neurology, Guangzhou, China.
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
22
|
Nair PP, Annapure US. Fermentation dynamics of bile salt hydrolase production in Heyndrickxia coagulans ATCC 7050 and Lactiplantibacillus plantarum ATCC 10012: Addressing ninhydrin assay limitations with a novel HPTLC-MS method. J Microbiol Methods 2024; 226:107050. [PMID: 39353547 DOI: 10.1016/j.mimet.2024.107050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Bile salt hydrolase (BSH), a pivotal enzyme in cholesterol management, holds significant promise in both human and animal subjects. This study investigated the effect of fermentation dynamics in Heyndrickxia coagulans ATCC 7050 and Lactiplantibacillus plantarum ATCC 10012 to enhance BSH production. Cultivation of cultures in MRS and M17 media revealed that MRS medium enhanced BSH production by 235.98 % in H. coagulans ATCC 7050 and 147.37 % in L. plantarum ATCC 10012, compared to M 17 medium. Additionally, varying oxygen concentration levels indicated that H. coagulans ATCC 7050 exhibited its minimum doubling time of 79.8 ± 0.64 min in anaerobic conditions, whereas L.plantarum ATCC 10012 demonstrated its minimum doubling time of 85.5 ± 1.2 min under microaerophilic conditions. However, their highest BSH activity was observed during the stationary phase under anaerobic conditions, yielding 17.14 ± 0.78 U/mL by H. coagulans ATCC 7050 and 19.04 ± 0.81 U/mL by L.plantarum ATCC 10012. Furthermore, it was observed that both organisms did not retain BSH within their cells. BSH activity was assessed using ninhydrin assay that detected free taurine liberated from sodium taurocholate. However, ninhydrin can yield false-positive results owing to its interaction with other free amino acids. To subjugate this limitation, the study introduced a novel and sensitive HPTLC-MS method capable of accurately detecting taurine. By comprehending fermentation dynamics and selecting appropriate conditions, BSH production increased 2.1-fold in both organisms. These findings illuminate critical insights, offering a pathway for novel strategies to enhance the BSH-producing capabilities of these LAB strains.
Collapse
Affiliation(s)
- Pratisha P Nair
- Department of Food Engineering and Technology, Institute of Chemical Technology, Matunga, Mumbai, Maharashtra, India
| | - Uday S Annapure
- Department of Food Engineering and Technology, Institute of Chemical Technology, Matunga, Mumbai, Maharashtra, India.
| |
Collapse
|
23
|
Nguyen HN, Huynh U, Zastrow ML. Fluorescent protein-based Zn 2+ sensors reveal distinct responses of aerobic and anaerobic Escherichia coli cultures to excess Zn 2. J Biol Chem 2024; 300:107840. [PMID: 39357830 PMCID: PMC11550654 DOI: 10.1016/j.jbc.2024.107840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Zinc ions are required by all known organisms. Maintaining zinc homeostasis by preventing toxic overload while ensuring sufficient acquisition for cellular functions is crucial for survival and growth of bacteria. Bacteria, however, frequently encounter and must survive in various environments. During infection in host animals, for example, bacteria are exposed to acidic conditions in the stomach and anaerobic conditions in the intestines, but the effects of oxygen on zinc homeostasis in Escherichia coli have not been well-studied. Previously, we reported a flavin-binding fluorescent protein-based zinc sensor, CreiLOVN41C, which can respond to changes in labile Zn2+ levels in bacteria under both aerobic and anaerobic conditions. Here, we combined the use of CreiLOVN41C with established oxygen-dependent fluorescent protein-based sensors, inductively coupled plasma-mass spectrometry, and growth curves to evaluate how oxygen levels affect zinc uptake in E. coli. Inductively coupled plasma-mass spectrometry results showed that cells grown aerobically with added zinc acquired more zinc, but no additional zinc was accumulated when cells were grown anaerobically. Using oxygen-independent CreiLOVN41C and the oxygen-dependent ZapCY series of sensors, intracellular labile zinc was detected in E. coli grown with varied zinc under varied conditions. Although little to no endogenous zinc was detected by any sensor in E. coli cells grown with up to 2 mM added zinc, CreiLOVN41C revealed that when Zn2+ was added and detected by cells in real-time, anaerobic cells required more Zn2+ to similarly saturate the sensor. Overall, this work reveals that zinc uptake in E. coli is impacted by oxygen levels during cell growth.
Collapse
Affiliation(s)
- Hazel N Nguyen
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Uyen Huynh
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Melissa L Zastrow
- Department of Chemistry, University of Houston, Houston, Texas, United States.
| |
Collapse
|
24
|
Gregory AL, Bussan HE, Topf MA, Hryckowian AJ. Impacts of perR on oxygen sensitivity, gene expression, and murine infection in Clostridioides difficile 630Δ erm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621113. [PMID: 39554039 PMCID: PMC11565800 DOI: 10.1101/2024.10.30.621113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Clostridioides difficile infection (CDI), characterized by colitis and diarrhea, afflicts approximately half a million people in the United States every year, burdening both individuals and the healthcare system. C. difficile 630Δerm is an erythromycin-sensitive variant of the clinical isolate C. difficile 630 and is commonly used in the C. difficile research community due to its genetic tractability. 630Δerm possesses a point mutation in perR, an autoregulated transcriptional repressor that regulates oxidative stress resistance genes. This point mutation results in a constitutively de-repressed PerR operon in 630Δerm. To address the impacts of perR on phenotypes relevant for oxygen tolerance and relevant to a murine model of CDI, we corrected the point mutant to restore PerR function in 630Δerm (herein, 630Δerm perR WT). We demonstrate that there is no difference in growth between 630Δerm and a 630Δerm perR WT under anaerobic conditions or when exposed to concentrations of O2 that mimic those found near the surface of the colonic epithelium. However, 630Δerm perR WT is more sensitive to ambient oxygen than 630Δerm, which coincides with alterations in expression of a variety of perR-dependent and perR-independent genes. Finally, we show that 630Δerm and 630Δerm perR WT do not differ in their ability to infect and cause disease in a well-established murine model of CDI. Together, these data support the hypothesis that the perR mutation in 630Δerm arose as a result of exposure to ambient oxygen and that the perR mutation in 630Δerm is unlikely to impact CDI-relevant phenotypes in laboratory studies.
Collapse
Affiliation(s)
- Anna L. Gregory
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Hailey E. Bussan
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Madeline A. Topf
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Andrew J. Hryckowian
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
25
|
La Vecchia M, Sala G, Sculco M, Aspesi A, Dianzani I. Genetics, diet, microbiota, and metabolome: partners in crime for colon carcinogenesis. Clin Exp Med 2024; 24:248. [PMID: 39470880 PMCID: PMC11522171 DOI: 10.1007/s10238-024-01505-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
Colorectal cancer (CRC) ranks among the most prevalent malignant tumors worldwide, with a multifactorial etiology encompassing genetic, environmental, and life-style factors, as well as the intestinal microbiota and its metabolome. These risk factors often work together in specific groups of patients, influencing how CRC develops and progresses. Importantly, alterations in the gut microbiota act as a critical nexus in this interplay, significantly affecting susceptibility to CRC. This review highlights recent insights into unmodifiable and modifiable risk factors for CRC and how they might interact with the gut microbiota and its metabolome. Understanding the mechanisms of these interactions will help us develop targeted, precision-medicine strategies that can adjust the composition of the gut microbiota to meet individual health needs, preventing or treating CRC more effectively.
Collapse
Affiliation(s)
- Marta La Vecchia
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Gloria Sala
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Marika Sculco
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Anna Aspesi
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Irma Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy.
| |
Collapse
|
26
|
Liu PY, Liaw J, Soutter F, Ortiz JJ, Tomley FM, Werling D, Gundogdu O, Blake DP, Xia D. Multi-omics analysis reveals regime shifts in the gastrointestinal ecosystem in chickens following anticoccidial vaccination and Eimeria tenella challenge. mSystems 2024; 9:e0094724. [PMID: 39287379 PMCID: PMC11494932 DOI: 10.1128/msystems.00947-24] [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: 08/14/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Coccidiosis, caused by Eimeria parasites, significantly impacts poultry farm economics and animal welfare. Beyond its direct impact on health, Eimeria infection disrupts enteric microbial populations leading to dysbiosis and increases vulnerability to secondary diseases such as necrotic enteritis, caused by Clostridium perfringens. The impact of Eimeria infection or anticoccidial vaccination on host gastrointestinal phenotypes and enteric microbiota remains understudied. In this study, the metabolomic profiles and microbiota composition of chicken caecal tissue and contents were evaluated concurrently during a controlled experimental vaccination and challenge trial. Cobb500 broilers were vaccinated with a Saccharomyces cerevisiae-vectored anticoccidial vaccine and challenged with 15,000 Eimeria tenella oocysts. Assessment of caecal pathology and quantification of parasite load revealed correlations with alterations to caecal microbiota and caecal metabolome linked to infection and vaccination status. Infection heightened microbiota richness with increases in potentially pathogenic species, while vaccination elevated beneficial Bifidobacterium. Using a multi-omics factor analysis, data on caecal microbiota and metabolome were integrated and distinct profiles for healthy, infected, and recovering chickens were identified. Healthy and recovering chickens exhibited higher vitamin B metabolism linked to short-chain fatty acid-producing bacteria, whereas essential amino acid and cell membrane lipid metabolisms were prominent in infected and vaccinated chickens. Notably, vaccinated chickens showed distinct metabolites related to the enrichment of sphingolipids, important components of nerve cells and cell membranes. Our integrated multi-omics model revealed latent biomarkers indicative of vaccination and infection status, offering potential tools for diagnosing infection, monitoring vaccination efficacy, and guiding the development of novel treatments or controls.IMPORTANCEAdvances in anticoccidial vaccines have garnered significant attention in poultry health management. However, the intricacies of vaccine-induced alterations in the chicken gut microbiome and its subsequent impact on host metabolism remain inadequately explored. This study delves into the metabolic and microbiotic shifts in chickens post-vaccination, employing a multi-omics integration analysis. Our findings highlight a notable synergy between the microbiome composition and host-microbe interacted metabolic pathways in vaccinated chickens, differentiating them from infected or non-vaccinated cohorts. These insights pave the way for more targeted and efficient approaches in poultry disease control, enhancing both the efficacy of vaccines and the overall health of poultry populations.
Collapse
Affiliation(s)
- Po-Yu Liu
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Janie Liaw
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - José Jaramillo Ortiz
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, London, United Kingdom
| | - Fiona M. Tomley
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
| | - Dirk Werling
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, London, United Kingdom
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Damer P. Blake
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, London, United Kingdom
| | - Dong Xia
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
27
|
Valiei A, Dickson A, Aminian-Dehkordi J, Mofrad MRK. Metabolic interactions shape emergent biofilm structures in a conceptual model of gut mucosal bacterial communities. NPJ Biofilms Microbiomes 2024; 10:99. [PMID: 39358363 PMCID: PMC11447261 DOI: 10.1038/s41522-024-00572-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
The gut microbiome plays a major role in human health; however, little is known about the structural arrangement of microbes and factors governing their distribution. In this work, we present an in silico agent-based model (ABM) to conceptually simulate the dynamics of gut mucosal bacterial communities. We explored how various types of metabolic interactions, including competition, neutralism, commensalism, and mutualism, affect community structure, through nutrient consumption and metabolite exchange. Results showed that, across scenarios with different initial species abundances, cross-feeding promotes species coexistence. Morphologically, competition and neutralism resulted in segregation, while mutualism and commensalism fostered high intermixing. In addition, cooperative relations resulted in community properties with little sensitivity to the selective uptake of metabolites produced by the host. Moreover, metabolic interactions strongly influenced colonization success following the invasion of newcomer species. These results provide important insights into the utility of ABM in deciphering complex microbiome patterns.
Collapse
Affiliation(s)
- Amin Valiei
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Andrew Dickson
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Javad Aminian-Dehkordi
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA.
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
28
|
Wang T, Wang RX, Colgan SP. Physiologic hypoxia in the intestinal mucosa: a central role for short-chain fatty acids. Am J Physiol Cell Physiol 2024; 327:C1087-C1093. [PMID: 39159391 PMCID: PMC11482044 DOI: 10.1152/ajpcell.00472.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
The intestinal mucosa is a dynamic surface that facilitates interactions between the host and an outside world that includes trillions of microbes, collectively termed the microbiota. This fine balance is regulated by an energetically demanding physical and biochemical barrier that is formed by the intestinal epithelial cells. In addition, this homeostasis exists at an interface between the anaerobic colonic lumen and a highly oxygenated, vascularized lamina propria. The resultant oxygen gradient within the intestine establishes "physiologic hypoxia" as a central metabolic feature of the mucosa. Although oxygen is vital for energy production to meet cellular metabolism needs, the availability of oxygen has far-reaching influences beyond just energy provision. Recent studies have shown that the intestinal mucosa has purposefully adapted to use differential oxygen levels largely through the presence of short-chain fatty acids (SCFAs), particularly butyrate (BA). Intestinal epithelial cells use butyrate for a multitude of functions that promote mucosal homeostasis. In this review, we explore how the physiologic hypoxia profile interfaces with SCFAs to benefit host mucosal tissues.
Collapse
Affiliation(s)
- Timothy Wang
- Mucosal Inflammation Program, Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Healthcare Studies, University of Texas Dallas, Richardson, Texas, United States
| | - Ruth X Wang
- Mucosal Inflammation Program, Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Dermatology, University of California San Diego, San Diego, California, United States
| | - Sean P Colgan
- Mucosal Inflammation Program, Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, United States
| |
Collapse
|
29
|
Bucking C, Bury NR, Sundh H, Wood CM. Making in vitro conditions more reflective of in vivo conditions for research on the teleost gastrointestinal tract. J Exp Biol 2024; 227:jeb246440. [PMID: 39392112 PMCID: PMC11529878 DOI: 10.1242/jeb.246440] [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] [Indexed: 10/12/2024]
Abstract
To date, the majority of in vitro or ex vivo fish gastrointestinal research has been conducted under unrealistic conditions. In a living fish, ionic conditions, as well as levels of ammonia, pH, HCO3- and PCO2 differ considerably between the different regions of the gastrointestinal tract. These factors also differ from those of the saline often used in gut research. Furthermore, the oxygen gradient from the serosa to the gut lumen is rarely considered: in contrast to the serosa, the lumen is a hypoxic/anoxic environment. In addition, the gut microbiome plays a significant role in gut physiology, increasing the complexity of the in vivo gut, but replicating the microbial community for in vitro studies is exceptionally difficult. However, there are ways in which we can begin to overcome these challenges. Firstly, the luminal chemistry and PO2 in each gut compartment must be carefully considered. Secondly, although microbiological culture techniques are improving, we must learn how to maintain the microbiome diversity seen in vivo. Finally, for ex vivo studies, developing mucosal (luminal) solutions that more closely mimic the in vivo conditions will better replicate physiological processes. Within the field of mammalian gut physiology, great advances in 'gut-on-chip' devices are providing the tools to better replicate in vivo conditions; adopting and adapting this technology may assist in fish gut research initiatives. This Commentary aims to make fish gut physiologists aware of the various issues in replicating the in vivo conditions and identifies solutions as well as those areas that require further improvement.
Collapse
Affiliation(s)
- Carol Bucking
- Department of Biology, Farquharson Life Science Building, York University, Toronto, ON, M3J 1P3, Canada
| | - Nic R. Bury
- School of Ocean and Earth Sciences, University of Southampton, National Oceanographic Centre, Waterfront Campus, Southampton, Hampshire, SO14 3ZH, UK
| | - Henrik Sundh
- Department of Biological & Environmental Sciences, University of Gothenburg, Medicinaregatan 7 B, 41390 Göteborg, Sweden
| | - Chris M. Wood
- Department of Zoology, University of British Columbia, 6270 University Blvd, Vancouver, BC, V6T1Z4, Canada
| |
Collapse
|
30
|
Sharma R, Raza GS, Sodum N, Walkowiak J, Herzig KH. Effect of hypoxia on GLP-1 secretion - an in vitro study using enteroendocrine STC-1 -cells as a model. Pflugers Arch 2024; 476:1613-1621. [PMID: 39075239 PMCID: PMC11381484 DOI: 10.1007/s00424-024-02996-z] [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: 05/22/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
Glucagon-like peptide (GLP)-1 is a hormone released by enteroendocrine L-cells after food ingestion. L-cells express various receptors for nutrient sensing including G protein-coupled receptors (GPRs). Intestinal epithelial cells near the lumen have a lower O2 tension than at the base of the crypts, which leads to hypoxia in L-cells. We hypothesized that hypoxia affects nutrient-stimulated GLP-1 secretion from the enteroendocrine cell line STC-1, the most commonly used model. In this study, we investigated the effect of hypoxia (1% O2) on alpha-linolenic acid (αLA) stimulated GLP-1 secretion and their receptor expressions. STC-1 cells were incubated for 12 h under hypoxia (1% O2) and treated with αLA to stimulate GLP-1 secretion. 12 h of hypoxia did not change basal GLP-1 secretion, but significantly reduced nutrient (αLA) stimulated GLP-1 secretion. In normoxia, αLA (12.5 μM) significantly stimulated (~ 5 times) GLP-1 secretion compared to control, but under hypoxia, GLP-1 secretion was reduced by 45% compared to normoxia. αLA upregulated GPR120, also termed free fatty acid receptor 4 (FFAR4), expressions under normoxia as well as hypoxia. Hypoxia downregulated GPR120 and GPR40 expression by 50% and 60%, respectively, compared to normoxia. These findings demonstrate that hypoxia does not affect the basal GLP-1 secretion but decreases nutrient-stimulated GLP-1 secretion. The decrease in nutrient-stimulated GLP-1 secretion was due to decreased GPR120 and GPR40 receptors expression. Changes in the gut environment and inflammation might contribute to the hypoxia of the epithelial and L-cells.
Collapse
Affiliation(s)
- Ravikant Sharma
- Research Unit of Biomedicine and Internal Medicine, Biocenter of Oulu, Medical Research Center, University of Oulu, Aapistie 5, 90220, Oulu, Finland
| | - Ghulam Shere Raza
- Research Unit of Biomedicine and Internal Medicine, Biocenter of Oulu, Medical Research Center, University of Oulu, Aapistie 5, 90220, Oulu, Finland
| | - Nalini Sodum
- Research Unit of Biomedicine and Internal Medicine, Biocenter of Oulu, Medical Research Center, University of Oulu, Aapistie 5, 90220, Oulu, Finland
| | - Jaroslaw Walkowiak
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, 60572, Poznań, Poland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine and Internal Medicine, Biocenter of Oulu, Medical Research Center, University of Oulu, Aapistie 5, 90220, Oulu, Finland.
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, 60572, Poznań, Poland.
| |
Collapse
|
31
|
Yersin S, Vonaesch P. Small intestinal microbiota: from taxonomic composition to metabolism. Trends Microbiol 2024; 32:970-983. [PMID: 38503579 DOI: 10.1016/j.tim.2024.02.013] [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: 11/30/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/21/2024]
Abstract
The small intestinal microbiota (SIM) is essential for gastrointestinal health, influencing digestion, immune modulation, and nutrient metabolism. Unlike the colonic microbiota, the SIM has been poorly characterized due to sampling challenges and ethical considerations. Current evidence suggests that the SIM consists of five core genera and additional segment-specific taxa. These bacteria closely interact with the human host, regulating nutrient absorption and metabolism. Recent work suggests the presence of two forms of small intestinal bacterial overgrowth, one dominated by oral bacteria (SIOBO) and a second dominated by coliform bacteria. Less invasive sampling techniques, omics approaches, and mechanistic studies will allow a more comprehensive understanding of the SIM, paving the way for interventions engineering the SIM towards better health.
Collapse
Affiliation(s)
- Simon Yersin
- Department of Fundamental Microbiology, Université de Lausanne, Lausanne, Switzerland
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, Université de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
32
|
Ma H, Wang Y, Wei J, Wang X, Yang H, Wang S. Stabilization of hypoxia-inducible factor 1α and regulation of specific gut microbes by EGCG contribute to the alleviation of ileal barrier disorder and obesity. Food Funct 2024; 15:9983-9994. [PMID: 39279449 DOI: 10.1039/d4fo02283a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Tea polyphenols have a regulatory effect on metabolic-related diseases, however, the underlying mechanism remains elusive. Our study aims to explore the dietary intervention effect of Epigallocatechin gallate (EGCG), the major polyphenol in green tea, on obesity and intestinal barrier disorders in mice fed a high-fat diet. By supplementing with 50 mg kg-1 EGCG, we observed a significant amelioration in body weight gain, fat accumulation, and liver dysfunction. Furthermore, EGCG modulated the HFD-induced metabolomic alterations. In particular, EGCG intervention restored the ileal barrier by enhancing the expression of tight junction proteins and antimicrobial peptides. At the mechanistic level, EGCG treatment stabilized hypoxia-inducible factor 1α (HIF1α) both in vitro and in vivo. Meanwhile, EGCG significantly increased the abundance of Dubosiella and Akkermansia, along with the elevated SCFA contents. These findings suggest that the ability of EGCG to stabilize HIF1α and regulate specific gut microbes is pivotal in mitigating ileal barrier dysfunction and obesity. Moreover, serum metabolomics revealed potential biomarkers following EGCG intervention. This study supports the intake of EGCG or green tea in obesity management and offers a novel perspective for investigating the metabolic regulatory mechanism of other dietary polyphenols.
Collapse
Affiliation(s)
- Hui Ma
- College of Food Science, Shanxi Normal University, Taiyuan 030031, Shanxi, China.
| | - Yuanyifei Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China.
| | - Jiayu Wei
- College of Food Science, Shanxi Normal University, Taiyuan 030031, Shanxi, China.
| | - Xiaochi Wang
- College of Food Science, Shanxi Normal University, Taiyuan 030031, Shanxi, China.
| | - Hui Yang
- College of Food Science, Shanxi Normal University, Taiyuan 030031, Shanxi, China.
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
33
|
Ling J, Hryckowian AJ. Re-framing the importance of Group B Streptococcus as a gut-resident pathobiont. Infect Immun 2024; 92:e0047823. [PMID: 38436256 PMCID: PMC11392526 DOI: 10.1128/iai.00478-23] [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] [Indexed: 03/05/2024] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is a Gram-positive bacterial species that causes disease in humans across the lifespan. While antibiotics are used to mitigate GBS infections, it is evident that antibiotics disrupt human microbiomes (which can predispose people to other diseases later in life), and antibiotic resistance in GBS is on the rise. Taken together, these unintended negative impacts of antibiotics highlight the need for precision approaches for minimizing GBS disease. One possible approach involves selectively depleting GBS in its commensal niches before it can cause disease at other body sites or be transmitted to at-risk individuals. One understudied commensal niche of GBS is the adult gastrointestinal (GI) tract, which may predispose colonization at other body sites in individuals at risk for GBS disease. However, a better understanding of the host-, microbiome-, and GBS-determined variables that dictate GBS GI carriage is needed before precise GI decolonization approaches can be developed. In this review, we synthesize current knowledge of the diverse body sites occupied by GBS as a pathogen and as a commensal. We summarize key molecular factors GBS utilizes to colonize different host-associated niches to inform future efforts to study GBS in the GI tract. We also discuss other GI commensals that are pathogenic in other body sites to emphasize the broader utility of precise de-colonization approaches for mitigating infections by GBS and other bacterial pathogens. Finally, we highlight how GBS treatments could be improved with a more holistic understanding of GBS enabled by continued GI-focused study.
Collapse
Affiliation(s)
- Joie Ling
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
- Microbiology Doctoral
Training Program, University of
Wisconsin-Madison, Madison,
Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
| |
Collapse
|
34
|
Parente IA, Chiara L, Bertoni S. Exploring the potential of human intestinal organoids: Applications, challenges, and future directions. Life Sci 2024; 352:122875. [PMID: 38942359 DOI: 10.1016/j.lfs.2024.122875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
The complex and dynamic environment of the gastrointestinal tract shapes one of the fastest renewing tissues in the human body, the intestinal epithelium. Considering the lack of human preclinical studies, reliable models that mimic the intestinal environment are increasingly explored. Patient-derived intestinal organoids are powerful tools that recapitulate in vitro many pathophysiological features of the human intestine. In this review, the possible applications of human intestinal organoids in different research fields are highlighted. From physiologically relevant to intestinal disease modeling, regenerative medicine, and toxicology studies, the potential of intestinal organoids will be here presented and discussed. Despite the remarkable opportunities offered, limitations related to ethical concerns, tissue collection, reproducibility, and methodologies may hinder the full exploitation of this cell-based model into high throughput studies and clinical practice. Currently, distinct approaches can be used to overcome the numerous challenges found along the way and to allow the full implementation of this ground-breaking technology.
Collapse
Affiliation(s)
- Inês A Parente
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Linda Chiara
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Simona Bertoni
- Department of Food and Drug, University of Parma, Parma, Italy.
| |
Collapse
|
35
|
Nahid DS, Coffey KA, Bei AK, Cordy RJ. Understanding the significance of oxygen tension on the biology of Plasmodium falciparum blood stages: From the human body to the laboratory. PLoS Pathog 2024; 20:e1012514. [PMID: 39298535 DOI: 10.1371/journal.ppat.1012514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Plasmodium falciparum undergoes sequestration within deep tissues of the human body, spanning multiple organ systems with differing oxygen (O2) concentrations. The parasite is exposed to an even greater range of O2 concentrations as it transitions from the human to the mosquito host, suggesting a high level of plasticity as it navigates these different environments. In this review, we explore factors that may contribute to the parasite's response to different environmental O2 concentrations, recognizing that there are likely multiple pieces to this puzzle. We first review O2-sensing mechanisms, which exist in other apicomplexans such as Toxoplasma gondii and consider whether similar systems could exist in Plasmodium. Next, we review morphological and functional changes in P. falciparum's mitochondrion during the asexual-to-sexual stage transition and discuss how these changes overlap with the parasite's access to O2. We then delve into reactive oxygen species (ROS) as ROS production is influenced by O2 availability and oxidative stress impacts Plasmodium intraerythrocytic development. Lastly, given that the primary role of the red blood cell (RBC) is to deliver O2 throughout the body, we discuss how changes in the oxygenation status of hemoglobin, the RBC's O2-carrying protein and key nutrient for Plasmodium, could also potentially impact the parasite's growth during intraerythrocytic development. This review also highlights studies that have investigated P. falciparum biology under varying O2 concentrations and covers technical aspects related to P. falciparum cultivation in the lab, focusing on sources of technical variation that could alter the amount of dissolved O2 encountered by cells during in vitro experiments. Lastly, we discuss how culture systems can better replicate in vivo heterogeneity with respect to O2 gradients, propose ideas for further research in this area, and consider translational implications related to O2 and malaria.
Collapse
Affiliation(s)
- Dinah S Nahid
- Department of Biology, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - Kevin A Coffey
- Department of Biology, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - Amy K Bei
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Regina Joice Cordy
- Department of Biology, Wake Forest University, Winston-Salem, North Carolina, United States of America
| |
Collapse
|
36
|
Rudolph SE, Bethi E, Iglesias-Ledon L, Kumarasinghe HU, Da'darah O, Salgam GK, Vieira KC, Chen Y, Kaplan DL. Set Up and Utilization of a Three-Dimensional In Vitro Bioreactor System for Human Intestinal Studies and Microbial Co-Cultures. Curr Protoc 2024; 4:e70013. [PMID: 39269316 DOI: 10.1002/cpz1.70013] [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: 09/15/2024]
Abstract
The study of human intestinal physiology and host-microbe interactions is crucial for understanding gastrointestinal health and disease. Traditional two-dimensional cell culture models lack the complexity of the native intestinal environment, limiting their utility in studying intestinal biology. Here, we present a detailed protocol for the set up and utilization of a three-dimensional (3D) in vitro bioreactor system for human intestinal studies and bacterial co-culture. This article outlines the design and assembly of the bioreactor system, scaffold fabrication, bacterial culture techniques, analysis methods, and troubleshooting tips. By providing step-by-step instructions, the goal is to enable other laboratories to utilize physiologically relevant tissue models of the human intestine, incorporating key features, such as nutrient flow, multiple human cell types, 3D architecture, and microbial communities. The incorporation of commensal bacteria into the bioreactor system allows for the investigation of complex host-microbe interactions, providing insight into gastrointestinal health and pathology. This article serves as a comprehensive resource for scientists seeking to advance their understanding of intestinal biology toward the development of novel therapeutic strategies for gastrointestinal disorders. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Scaffold design Basic Protocol 2: Intestinal cell culture: Caco2 cells Basic Protocol 3: Intestinal cell culture: organoids Basic Protocol 4: Bioreactor design and set up Basic Protocol 5: Bacteria in 3D bioreactor set up Basic Protocol 6: Bacteria and drug dosing.
Collapse
Affiliation(s)
- Sara E Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Eesha Bethi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | | | | | - Omar Da'darah
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Gautham K Salgam
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Karolinny C Vieira
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
37
|
Pozzoli C, Martinelli G, Fumagalli M, Di Lorenzo C, Maranta N, Colombo L, Piazza S, Dell’Agli M, Sangiovanni E. Castanea sativa Mill. By-Products: Investigation of Potential Anti-Inflammatory Effects in Human Intestinal Epithelial Cells. Molecules 2024; 29:3951. [PMID: 39203029 PMCID: PMC11357300 DOI: 10.3390/molecules29163951] [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: 07/23/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Castanea sativa Mill. (C. sativa) processing and pruning generate several by-products, including leaves, burs, and shells (inner and outer teguments), which are considered an important source of high-value phytochemicals. Ellagitannins from C. sativa leaf extracts have been described to impair H. pylori viability and inflammation in gastric cells. Furthermore, chestnut shells showed an important anti-inflammatory effect in gastric epithelial cells. Dietary polyphenols, including tannins, have been reported to interfere with targets of inflammation, including the nuclear factor κB (NF-κB). A promising role as a further therapeutical target for gut disorders has been recently proposed for the regulatory subunit of hypoxia-inducible factor (HIF-1α), as a potential stabilizer of intestinal barrier integrity. Therefore, the main objective of this work is the chemical characterization of several chestnut by-products (bud, spiny bur, wood, pericarp and episperm), together with the exploitation of their anti-inflammatory properties in intestinal cells, scavenging capacity, and stability following gastrointestinal digestion. The chemical characterization confirmed the presence of bioactive polyphenols in the extracts, including ellagitannins. In CaCo-2 cells stimulated by an IL-1β-IFN-γ cocktail, nearly all chestnut by-products (50 µg/mL) inhibited the release of proinflammatory mediators (CXCL-10, IL-8, MCP-1, ICAM), along with the NF-κB-driven transcription, and induced the HRE-driven transcription. The stability of the most promising extracts, identified through PCA and cluster analysis, was addressed by in vitro gastrointestinal digestion. Despite the significant reduction in total polyphenol index of chestnut bud and wood after gastric and intestinal digestion, the activity of these extracts on both scavenging and anti-inflammatory parameters remained promising. These data contribute to exploit the potential of chestnut by-products as sources of dietary polyphenols with anti-inflammatory properties at the intestinal level. Moreover, this study could represent an important step to encourage the recycling and valorization of chestnut by-products, promoting the circular economy and reducing the environmental impact related to the management of agriculture waste.
Collapse
Affiliation(s)
- Carola Pozzoli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Giulia Martinelli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Marco Fumagalli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Chiara Di Lorenzo
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Nicole Maranta
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Luca Colombo
- Consorzio Castanicoltori di Brinzio, Orino e Castello Cabiaglio, Società Cooperativa Agricola-Varese, 21100 Varese, Italy;
| | - Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Mario Dell’Agli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Enrico Sangiovanni
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| |
Collapse
|
38
|
Hajjar R, Richard C, Santos MM. The gut barrier as a gatekeeper in colorectal cancer treatment. Oncotarget 2024; 15:562-572. [PMID: 39145528 PMCID: PMC11325587 DOI: 10.18632/oncotarget.28634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024] Open
Abstract
Colorectal cancer (CRC) is highly prevalent and is a major cause of cancer-related deaths worldwide. The incidence rate of CRC remains alarmingly high despite screening measures. The main curative treatment for CRC is a surgical resection of the diseased bowel segment. Postoperative complications usually involve a weakened gut barrier and a dissemination of bacterial proinflammatory lipopolysaccharides. Herein we discuss how gut microbiota and microbial metabolites regulate basal inflammation levels in the gut and the healing process of the bowel after surgery. We further elaborate on the restoration of the gut barrier function in patients with CRC and how this potentially impacts the dissemination and implantation of CRC cells in extracolonic tissues, contributing therefore to worse survival after surgery.
Collapse
Affiliation(s)
- Roy Hajjar
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Department of Surgery, Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Carole Richard
- Department of Surgery, Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
39
|
Haque PS, Kapur N, Barrett TA, Theiss AL. Mitochondrial function and gastrointestinal diseases. Nat Rev Gastroenterol Hepatol 2024; 21:537-555. [PMID: 38740978 DOI: 10.1038/s41575-024-00931-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
Mitochondria are dynamic organelles that function in cellular energy metabolism, intracellular and extracellular signalling, cellular fate and stress responses. Mitochondria of the intestinal epithelium, the cellular interface between self and enteric microbiota, have emerged as crucial in intestinal health. Mitochondrial dysfunction occurs in gastrointestinal diseases, including inflammatory bowel diseases and colorectal cancer. In this Review, we provide an overview of the current understanding of intestinal epithelial cell mitochondrial metabolism, function and signalling to affect tissue homeostasis, including gut microbiota composition. We also discuss mitochondrial-targeted therapeutics for inflammatory bowel diseases and colorectal cancer and the evolving concept of mitochondrial impairment as a consequence versus initiator of the disease.
Collapse
Affiliation(s)
- Parsa S Haque
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Neeraj Kapur
- Department of Medicine, Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Terrence A Barrett
- Department of Medicine, Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY, USA
- Lexington Veterans Affairs Medical Center Kentucky, Lexington, KY, USA
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, USA.
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA.
| |
Collapse
|
40
|
Di Mattia M, Sallese M, Neri M, Lopetuso LR. Hypoxic Functional Regulation Pathways in the GI Tract: Focus on the HIF-1α and Microbiota's Crosstalk. Inflamm Bowel Dis 2024; 30:1406-1418. [PMID: 38484200 DOI: 10.1093/ibd/izae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Indexed: 08/02/2024]
Abstract
Hypoxia is an essential gastrointestinal (GI) tract phenomenon that influences both physiologic and pathologic states. Hypoxia-inducible factors (HIFs), the primary drivers of cell adaptation to low-oxygen environments, have been identified as critical regulators of gut homeostasis: directly, through the induction of different proteins linked to intestinal barrier stabilization (ie, adherent proteins, tight junctions, mucins, integrins, intestinal trefoil factor, and adenosine); and indirectly, through the regulation of several immune cell types and the modulation of autophagy and inflammatory processes. Furthermore, hypoxia and HIF-related sensing pathways influence the delicate relationship existing between bacteria and mammalian host cells. In turn, gut commensals establish and maintain the physiologic hypoxia of the GI tract and HIF-α expression. Based on this premise, the goals of this review are to (1) highlight hypoxic molecular pathways in the GI tract, both in physiologic and pathophysiologic settings, such as inflammatory bowel disease; and (2) discuss a potential strategy for ameliorating gut-related disorders, by targeting HIF signaling, which can alleviate inflammatory processes, restore autophagy correct mechanisms, and benefit the host-microbiota equilibrium.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Matteo Neri
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
41
|
Iacovacci J, Serafini MS, Avuzzi B, Badenchini F, Cicchetti A, Devecchi A, Dispinzieri M, Doldi V, Giandini T, Gioscio E, Mancinelli E, Noris Chiorda B, Orlandi E, Palorini F, Possenti L, Reis Ferreira M, Villa S, Zaffaroni N, De Cecco L, Valdagni R, Rancati T. Intestinal microbiota composition is predictive of radiotherapy-induced acute gastrointestinal toxicity in prostate cancer patients. EBioMedicine 2024; 106:105246. [PMID: 39029427 PMCID: PMC11314862 DOI: 10.1016/j.ebiom.2024.105246] [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: 10/11/2023] [Revised: 06/03/2024] [Accepted: 07/02/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND The search for factors beyond the radiotherapy dose that could identify patients more at risk of developing radio-induced toxicity is essential to establish personalised treatment protocols for improving the quality-of-life of survivors. To investigate the role of the intestinal microbiota in the development of radiotherapy-induced gastrointestinal toxicity, the MicroLearner observational cohort study characterised the intestinal microbiota of 136 (discovery) and 79 (validation) consecutive prostate cancer patients at baseline radiotherapy. METHODS Gastrointestinal toxicity was assessed weekly during RT using CTCAE. An average grade >1.3 over time points was used to identify patients suffering from persistent acute toxicity (endpoint). The microbiota of patients was quantified from the baseline faecal samples using 16S rRNA gene sequencing technology and the Ion Reporter metagenomic pipeline. Statistical techniques and computational and machine learning tools were used to extract, functionally characterise, and predict core features of the bacterial communities of patients who developed acute gastrointestinal toxicity. FINDINGS Analysis of the core bacterial composition in the discovery cohort revealed a cluster of patients significantly enriched for toxicity, displaying a toxicity rate of 60%. Based on selected high-risk microbiota compositional features, we developed a clinical decision tree that could effectively predict the risk of toxicity based on the relative abundance of genera Faecalibacterium, Bacteroides, Parabacteroides, Alistipes, Prevotella and Phascolarctobacterium both in internal and external validation cohorts. INTERPRETATION We provide evidence showing that intestinal bacteria profiling from baseline faecal samples can be effectively used in the clinic to improve the pre-radiotherapy assessment of gastrointestinal toxicity risk in prostate cancer patients. FUNDING Italian Ministry of Health (Promotion of Institutional Research INT-year 2016, 5 × 1000, Ricerca Corrente funds). Fondazione Regionale per la Ricerca Biomedica (ID 2721017). AIRC (IG 21479).
Collapse
Affiliation(s)
- Jacopo Iacovacci
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| | - Mara Serena Serafini
- Unit of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Barbara Avuzzi
- Unit of Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Fabio Badenchini
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Alessandro Cicchetti
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Andrea Devecchi
- Unit of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Michela Dispinzieri
- Unit of Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Valentina Doldi
- Unit of Molecular Pharmacology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Tommaso Giandini
- Unit of Medical Physics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Eliana Gioscio
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elisa Mancinelli
- Unit of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Barbara Noris Chiorda
- Unit of Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Ester Orlandi
- Radiation Oncology Clinical Department, National Center for Oncological Hadron Therapy (CNAO), Pavia, Italy
| | - Federica Palorini
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Luca Possenti
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Miguel Reis Ferreira
- King's College London, London, UK; Guys and St Thomas NHS Foundation Trust, London, UK
| | - Sergio Villa
- Unit of Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Nadia Zaffaroni
- Unit of Molecular Pharmacology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Loris De Cecco
- Unit of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Riccardo Valdagni
- Unit of Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy; Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy; Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Tiziana Rancati
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
42
|
Yang YH, Yan F, Shi PS, Yang LC, Cui DJ. HIF-1α Pathway Orchestration by LCN2: A Key Player in Hypoxia-Mediated Colitis Exacerbation. Inflammation 2024; 47:1491-1519. [PMID: 38819583 DOI: 10.1007/s10753-024-01990-y] [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: 12/04/2023] [Revised: 01/19/2024] [Accepted: 02/09/2024] [Indexed: 06/01/2024]
Abstract
In this study, we investigated the role of hypoxia in the development of chronic inflammatory bowel disease (IBD), focusing on its impact on the HIF-1α signaling pathway through the upregulation of lipocalin 2 (LCN2). Using a murine model of colitis induced by sodium dextran sulfate (DSS) under hypoxic conditions, transcriptome sequencing revealed LCN2 as a key gene involved in hypoxia-mediated exacerbation of colitis. Bioinformatics analysis highlighted the involvement of crucial pathways, including HIF-1α and glycolysis, in the inflammatory process. Immune infiltration analysis demonstrated the polarization of M1 macrophages in response to hypoxic stimulation. In vitro studies using RAW264.7 cells further elucidated the exacerbation of inflammation and its impact on M1 macrophage polarization under hypoxic conditions. LCN2 knockout cells reversed hypoxia-induced inflammatory responses, and the HIF-1α pathway activator dimethyloxaloylglycine (DMOG) confirmed LCN2's role in mediating inflammation via the HIF-1α-induced glycolysis pathway. In a DSS-induced colitis mouse model, oral administration of LCN2-silencing lentivirus and DMOG under hypoxic conditions validated the exacerbation of colitis. Evaluation of colonic tissues revealed altered macrophage polarization, increased levels of inflammatory factors, and activation of the HIF-1α and glycolysis pathways. In conclusion, our findings suggest that hypoxia exacerbates colitis by modulating the HIF-1α pathway through LCN2, influencing M1 macrophage polarization in glycolysis. This study contributes to a better understanding of the mechanisms underlying IBD, providing potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Yun-Han Yang
- Department of Gastroenterology, Guizhou Inflammatory Bowel Disease Research Center, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Fang Yan
- Department of Gastroenterology, Guizhou Inflammatory Bowel Disease Research Center, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Peng-Shuang Shi
- Department of Gastroenterology, Guizhou Inflammatory Bowel Disease Research Center, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Liu-Chan Yang
- Department of Gastroenterology, Guizhou Inflammatory Bowel Disease Research Center, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - De-Jun Cui
- Department of Gastroenterology, Guizhou Inflammatory Bowel Disease Research Center, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China.
| |
Collapse
|
43
|
Ermann Lundberg L, Pallabi Mishra P, Liu P, Forsberg MM, Sverremark-Ekström E, Grompone G, Håkansson S, Linninge C, Roos S. Bifidobacterium longum subsp. longum BG-L47 boosts growth and activity of Limosilactobacillus reuteri DSM 17938 and its extracellular membrane vesicles. Appl Environ Microbiol 2024; 90:e0024724. [PMID: 38888338 PMCID: PMC11267924 DOI: 10.1128/aem.00247-24] [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: 02/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.
Collapse
Affiliation(s)
- Ludwig Ermann Lundberg
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| | - Punya Pallabi Mishra
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Manuel Mata Forsberg
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Sebastian Håkansson
- BioGaia, Stockholm, Sweden
- Division of Applied Microbiology, Department of Chemistry, Lund University, Lund, Sweden
| | - Caroline Linninge
- BioGaia, Stockholm, Sweden
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| |
Collapse
|
44
|
Kalam N, Balasubramaniam VRMT. Crosstalk between COVID-19 and the gut-brain axis: a gut feeling. Postgrad Med J 2024; 100:539-554. [PMID: 38493312 DOI: 10.1093/postmj/qgae030] [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: 12/10/2023] [Accepted: 02/15/2024] [Indexed: 03/18/2024]
Abstract
The microbes in the gut are crucial for maintaining the body's immune system and overall gut health. However, it is not fully understood how an unstable gut environment can lead to more severe cases of SARS-CoV-2 infection. The gut microbiota also plays a role in the gut-brain axis and interacts with the central nervous system through metabolic and neuroendocrine pathways. The interaction between the microbiota and the host's body involves hormonal, immune, and neural pathways, and any disruption in the balance of gut bacteria can lead to dysbiosis, which contributes to pathogen growth. In this context, we discuss how dysbiosis could contribute to comorbidities that increase susceptibility to SARS-CoV-2. Probiotics and fecal microbiota transplantation have successfully treated infectious and non-infectious inflammatory-related diseases, the most common comorbidities. These treatments could be adjuvant therapies for COVID-19 infection by restoring gut homeostasis and balancing the gut microbiota.
Collapse
Affiliation(s)
- Nida Kalam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Malaysia
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Malaysia
| |
Collapse
|
45
|
Dzhalilova D, Silina M, Tsvetkov I, Kosyreva A, Zolotova N, Gantsova E, Kirillov V, Fokichev N, Makarova O. Changes in the Expression of Genes Regulating the Response to Hypoxia, Inflammation, Cell Cycle, Apoptosis, and Epithelial Barrier Functioning during Colitis-Associated Colorectal Cancer Depend on Individual Hypoxia Tolerance. Int J Mol Sci 2024; 25:7801. [PMID: 39063041 PMCID: PMC11276979 DOI: 10.3390/ijms25147801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
One of the factors contributing to colorectal cancer (CRC) development is inflammation, which is mostly hypoxia-associated. This study aimed to characterize the morphological and molecular biological features of colon tumors in mice that were tolerant and susceptible to hypoxia based on colitis-associated CRC (CAC). Hypoxia tolerance was assessed through a gasping time evaluation in a decompression chamber. One month later, the animals were experimentally modeled for colitis-associated CRC by intraperitoneal azoxymethane administration and three dextran sulfate sodium consumption cycles. The incidence of tumor development in the distal colon in the susceptible to hypoxia mice was two times higher and all tumors (100%) were represented by adenocarcinomas, while in the tolerant mice, only 14% were adenocarcinomas and 86% were glandular intraepithelial neoplasia. The tumor area assessed on serially stepped sections was statistically significantly higher in the susceptible animals. The number of macrophages, CD3-CD19+, CD3+CD4+, and NK cells in tumors did not differ between animals; however, the number of CD3+CD8+ and vimentin+ cells was higher in the susceptible mice. Changes in the expression of genes regulating the response to hypoxia, inflammation, cell cycle, apoptosis, and epithelial barrier functioning in tumors and the peritumoral area depended on the initial mouse's hypoxia tolerance, which should be taken into account for new CAC diagnostics and treatment approaches development.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Maria Silina
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Ivan Tsvetkov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Anna Kosyreva
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
- Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Natalia Zolotova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Elena Gantsova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
- Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Vladimir Kirillov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Health of Russian Federation, 117513 Moscow, Russia;
| | - Nikolay Fokichev
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Olga Makarova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| |
Collapse
|
46
|
Mafra D, Borges NA, Baptista BG, Martins LF, Borland G, Shiels PG, Stenvinkel P. What Can the Gut Microbiota of Animals Teach Us about the Relationship between Nutrition and Burden of Lifestyle Diseases? Nutrients 2024; 16:1789. [PMID: 38892721 PMCID: PMC11174762 DOI: 10.3390/nu16111789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
The gut microbiota performs several crucial roles in a holobiont with its host, including immune regulation, nutrient absorption, synthesis, and defense against external pathogens, significantly influencing host physiology. Disruption of the gut microbiota has been linked to various chronic conditions, including cardiovascular, kidney, liver, respiratory, and intestinal diseases. Studying how animals adapt their gut microbiota across their life course at different life stages and under the dynamics of extreme environmental conditions can provide valuable insights from the natural world into how the microbiota modulates host biology, with a view to translating these into treatments or preventative measures for human diseases. By modulating the gut microbiota, opportunities to address many complications associated with chronic diseases appear. Such a biomimetic approach holds promise for exploring new strategies in healthcare and disease management.
Collapse
Affiliation(s)
- Denise Mafra
- Graduate Program in Medical Sciences and Graduate Program in Nutrition Sciences, Federal Fluminense University (UFF), Niterói 24020-141, Brazil;
- Graduate Program in Biological Sciences—Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Natália A. Borges
- Graduate Program in Food, Nutrition, and Health, Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro 21941-909, Brazil;
| | - Beatriz G. Baptista
- Graduate Program in Medical Sciences and Graduate Program in Nutrition Sciences, Federal Fluminense University (UFF), Niterói 24020-141, Brazil;
| | - Layla F. Martins
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo 05508-220, Brazil;
| | - Gillian Borland
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK; (G.B.); (P.G.S.)
| | - Paul G. Shiels
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK; (G.B.); (P.G.S.)
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, 17165 Stockholm, Sweden;
| |
Collapse
|
47
|
Oladele P, Ngo J, Chang T, Johnson TA. Temporal dynamics of fecal microbiota community succession in broiler chickens, calves, and piglets under aerobic exposure. Microbiol Spectr 2024; 12:e0408423. [PMID: 38717193 PMCID: PMC11237419 DOI: 10.1128/spectrum.04084-23] [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: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 06/06/2024] Open
Abstract
Researchers have extensively studied the effect of oxygen on the growth and survival of bacteria. However, the impact of oxygen on bacterial community structure, particularly its ability to select for taxa within the context of a complex microbial community, is still unclear. In a 21-day microcosm experiment, we investigated the effect of aerobic exposure on the fecal community structure and succession pattern in broiler, calf, and piglet feces (n = 10 for each feces type). Bacterial diversity decreased and community structure changed rapidly in the broiler microbiome (P < 0.001), while the fecal community of calves and piglets, which have higher initial diversity, was stable after initial exposure but decreased in diversity after 3 days (P < 0.001). The response to aerobic exposure was host animal specific, but in all three animals, the change in community structure was driven by a decrease in anaerobic species, primarily belonging to Firmicutes and Bacteroidetes (except in broilers where Bacteroidetes increased), along with an increase in aerobic species belonging to Proteobacteria and Actinobacteria. Using random forest regression, we identified microbial features that predict aerobic exposure. In all three animals, host-beneficial Prevotella-related ASVs decreased after exposure, while ASVs belonging to Acinetobacter, Corynbacterium, and Tissierella were increased. The decrease of Prevotella was rapid in broilers but delayed in calves and piglets. Knowing when these pathobionts increase in abundance after aerobic exposure could inform farm sanitation practices and could be important in designing animal experiments that modulate the microbiome.IMPORTANCEThe fecal microbial community is contained within a dynamic ecosystem of interacting microbes that varies in biotic and abiotic components across different animal species. Although oxygen affects bacterial growth, its specific impact on the structure of complex communities, such as those found in feces, and how these effects vary between different animal species are poorly understood. In this study, we demonstrate that the effect of aerobic exposure on the fecal microbiota was host-animal-specific, primarily driven by a decrease in Firmicutes and Bacteroidetes, but accompanied by an increase in Actinobacteria, Proteobacteria, and other pathobionts. Interestingly, we observed that more complex communities from pig and cattle exhibited initial resilience, while a less diverse community from broilers displayed a rapid response to aerobic exposure. Our findings offer insights that can inform farm sanitation practices, as well as experimental design, sample collection, and processing protocols for microbiome studies across various animal species.
Collapse
Affiliation(s)
- Paul Oladele
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Jennifer Ngo
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Tiffany Chang
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Timothy A. Johnson
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
48
|
Arturo Tozzi, Minella R. Dynamics and metabolic effects of intestinal gases in healthy humans. Biochimie 2024; 221:81-90. [PMID: 38325747 DOI: 10.1016/j.biochi.2024.02.001] [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: 11/04/2023] [Revised: 01/06/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
Many living beings use exogenous and/or endogenous gases to attain evolutionary benefits. We make a comprehensive assessment of one of the major gaseous reservoirs in the human body, i.e., the bowel, providing extensive data that may serve as reference for future studies. We assess the intestinal gases in healthy humans, including their volume, composition, source and local distribution in proximal as well as distal gut. We analyse each one of the most abundant intestinal gases including nitrogen, oxygen, nitric oxide, carbon dioxide, methane, hydrogen, hydrogen sulfide, sulfur dioxide and cyanide. For every gas, we describe diffusive patterns, active trans-barrier transport dynamics, chemical properties, intra-/extra-intestinal metabolic effects mediated by intracellular, extracellular, paracrine and distant actions. Further, we highlight the local and systemic roles of gasotransmitters, i.e., signalling gaseous molecules that can freely diffuse through the intestinal cellular membranes. Yet, we provide testable hypotheses concerning the still unknown effects of some intestinal gases on the myenteric and submucosal neurons.
Collapse
Affiliation(s)
- Arturo Tozzi
- Center for Nonlinear Science, Department of Physics, University of North Texas, 1155 Union Circle, #311427, Denton, TX, 76203-5017, USA.
| | | |
Collapse
|
49
|
Vandecruys M, De Smet S, De Beir J, Renier M, Leunis S, Van Criekinge H, Glorieux G, Raes J, Vanden Wyngaert K, Nagler E, Calders P, Monbaliu D, Cornelissen V, Evenepoel P, Van Craenenbroeck AH. Revitalizing the Gut Microbiome in Chronic Kidney Disease: A Comprehensive Exploration of the Therapeutic Potential of Physical Activity. Toxins (Basel) 2024; 16:242. [PMID: 38922137 PMCID: PMC11209503 DOI: 10.3390/toxins16060242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Both physical inactivity and disruptions in the gut microbiome appear to be prevalent in patients with chronic kidney disease (CKD). Engaging in physical activity could present a novel nonpharmacological strategy for enhancing the gut microbiome and mitigating the adverse effects associated with microbial dysbiosis in individuals with CKD. This narrative review explores the underlying mechanisms through which physical activity may favorably modulate microbial health, either through direct impact on the gut or through interorgan crosstalk. Also, the development of microbial dysbiosis and its interplay with physical inactivity in patients with CKD are discussed. Mechanisms and interventions through which physical activity may restore gut homeostasis in individuals with CKD are explored.
Collapse
Affiliation(s)
- Marieke Vandecruys
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
| | - Stefan De Smet
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, 3000 Leuven, Belgium;
| | - Jasmine De Beir
- Department of Rehabilitation Sciences, Ghent University, 9000 Ghent, Belgium; (J.D.B.); (P.C.)
| | - Marie Renier
- Group Rehabilitation for Internal Disorders, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (M.R.); (V.C.)
| | - Sofie Leunis
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
| | - Hanne Van Criekinge
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
| | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute for Medical Research, 3000 Leuven, Belgium;
- VIB-KU Leuven Center for Microbiology, 3000 Leuven, Belgium
| | - Karsten Vanden Wyngaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Evi Nagler
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Patrick Calders
- Department of Rehabilitation Sciences, Ghent University, 9000 Ghent, Belgium; (J.D.B.); (P.C.)
| | - Diethard Monbaliu
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
- Transplantoux Foundation, 3000 Leuven, Belgium
| | - Véronique Cornelissen
- Group Rehabilitation for Internal Disorders, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (M.R.); (V.C.)
| | - Pieter Evenepoel
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
- Department of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Amaryllis H. Van Craenenbroeck
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
- Department of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
50
|
Yan Y, Zheng X, Liu G, Shi G, Li C, Chen H, He X, Lin K, Deng Z, Zhang H, Li WG, Chen H, Tong X, Zhu Z. Gut microbiota-derived cholic acid mediates neonatal brain immaturity and white matter injury under chronic hypoxia. iScience 2024; 27:109633. [PMID: 38638560 PMCID: PMC11025012 DOI: 10.1016/j.isci.2024.109633] [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: 11/21/2023] [Revised: 02/18/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024] Open
Abstract
Chronic hypoxia, common in neonates, disrupts gut microbiota balance, which is crucial for brain development. This study utilized cyanotic congenital heart disease (CCHD) patients and a neonatal hypoxic rat model to explore the association. Both hypoxic rats and CCHD infants exhibited brain immaturity, white matter injury (WMI), brain inflammation, and motor/learning deficits. Through 16s rRNA sequencing and metabolomic analysis, a reduction in B. thetaiotaomicron and P. distasonis was identified, leading to cholic acid accumulation. This accumulation triggered M1 microglial activation and inflammation-induced WMI. Administration of these bacteria rescued cholic acid-induced WMI in hypoxic rats. These findings suggest that gut microbiota-derived cholic acid mediates neonatal WMI and brain inflammation, contributing to brain immaturity under chronic hypoxia. Therapeutic targeting of these bacteria provides a non-invasive intervention for chronic hypoxia patients.
Collapse
Affiliation(s)
- Yichen Yan
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Zheng
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science, Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Liu
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Guocheng Shi
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Li
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongtong Chen
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kana Lin
- Center for Brain Science, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pharmacy, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaohui Deng
- Department of Gastroenterology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Zhang
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guang Li
- Center for Brain Science, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiwen Chen
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Tong
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science, Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongqun Zhu
- Department of Cardiothoracic Surgery, Congenital Heart Center, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|