1
|
Wang Z, Gao X, Ji H, Shao M, Ni B, Fei S, Sun L, Chen H, Tan R, Du M, Gu M. Characterization of gut microbiota and metabolites in renal transplant recipients during COVID-19 and prediction of one-year allograft function. J Transl Med 2025; 23:420. [PMID: 40211390 PMCID: PMC11987245 DOI: 10.1186/s12967-025-06090-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND The gut-lung-kidney axis is pivotal in immune-related kidney diseases, with gut dysbiosis potentially exacerbating the severity of Coronavirus disease 2019 (COVID-19) in recipients of kidney transplant. This study aimed to characterize the gut microbiome and metabolome in renal transplant recipients with COVID-19 pneumonia over a one-year follow-up period. METHODS A total of 30 renal transplant recipients were enrolled, comprising 17 with COVID-19 pneumonia, six with mild COVID-19, and seven without COVID-19. Fecal samples were collected at the onset of infection for gut microbiome and metabolome analysis. Generalized Estimating Equations (GEE) model and Latent Class Growth Mixed Model (LCGMM) were employed to dissect the relationships among clinical characteristics, laboratory tests, and gut microbiota and metabolites. RESULTS Four microbial phyla (Deferribacteres, TM7, Fusobacteria, and Gemmatimonadetes) and 13 genera were significantly enriched across three recipients groups, correlating with baseline inflammatory response and allograft function. Additionally, 52 differentially expressed metabolites were identified, with seven significantly correlating with eight altered microbiota genera. LCGMM revealed two distinct classes of recipients, with those suffering from COVID-19 pneumonia exhibiting significantly elevated serum creatinine (Scr) trajectories over the one-year period. GEE further identified 12 genera and 181 metabolites closely associated with these trajectories; a multivariable model incorporating gut metabolites of 1-Caffeoylquinic Acid and PMK was found to effectively predict one-year allograft function. CONCLUSIONS Our study indicates a possible interaction between the composition of the gut microbiota and metabolites community and COVID-19 in renal transplant recipients, particularly in relation to disease severity and the prediction of one-year allograft function.
Collapse
Affiliation(s)
- Zijie Wang
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
| | - Xiang Gao
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
| | - Hongsheng Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Departments of Environmental Genomics and Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ming Shao
- Departments of Environmental Genomics and Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Bin Ni
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
| | - Shuang Fei
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Li Sun
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Chen
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ruoyun Tan
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Mulong Du
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China.
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Departments of Environmental Genomics and Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Min Gu
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China.
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
2
|
Bhunyakarnjanarat T, Udompornpitak K, Wannigama DL, Ruchusatsawat A, Aphiboon P, Sripong T, Thim-Uam A, Leelahavanichkul A. Intratracheal Candida administration induced lung dysbiosis, activated neutrophils, and worsened lung hemorrhage in pristane-induced lupus mice. Sci Rep 2025; 15:9768. [PMID: 40118938 PMCID: PMC11928548 DOI: 10.1038/s41598-025-94632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Because the innate immunity might and fungi in the lungs might enhance the severity of lupus-induced diffuse alveolar hemorrhage (DAH), intraperitoneal pristane injection was performed in C57BL6 mice with intratracheal administration by Candida albicans or phosphate buffer solution (PBS). Despite the similar pristane-induced lupus (proteinuria, serum creatinine, and serum anti-dsDNA) at 5 weeks of the model, Candida administration worsened several characteristics, including mortality, body weight, serum cytokines (TNF-α and IL-6), and lung hemorrhage score, and cytokines in the lung tissue (TNF-α, IL-6, and IL-10), but not gut permeability (FITC-dextran assay), serum IL-10, immune cells in the spleens (flow cytometry analysis), and activities of peritoneal macrophages (polymerase-chain reaction). Although Candida administration reduced proteobacterial abundance and altered alpha and beta diversity compared with PBS control, lung microbiota was not different between Candida administration in pristane- and non-pristane-administered mice. Because of the prominent Gram-negative bacteria in lung microbiota and the role of neutrophils in DAH, lipopolysaccharide (LPS) with and without heat-killed Candida preparation was tested. Indeed, Candida preparation with LPS induced more severe pro-inflammatory neutrophils than LPS stimulation alone as indicated by the expression of several genes (TNF-α, IL-6, IL-1β, IL-10, Dectin-1, and NF-κB). In conclusion, the intratracheal Candida worsened pristane-induced lung hemorrhage partly through the enhanced neutrophil responses against bacteria and fungi. More studies on Candida colonization in sputum from patients with lupus-induced DAH are interesting.
Collapse
Affiliation(s)
- Thansita Bhunyakarnjanarat
- Department of Microbiology, Faculty of Medicine, Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, UK
| | - Alisa Ruchusatsawat
- Department of Microbiology, Faculty of Medicine, Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, 10330, Thailand
- Engineering Science Classroom, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pranpan Aphiboon
- Department of Microbiology, Faculty of Medicine, Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanadech Sripong
- Department of Microbiology, Faculty of Medicine, Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Arthid Thim-Uam
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand.
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, 10330, Thailand.
- Faculty of Medicine, Center of Excellence in Systems Biology, Research Affairs, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
3
|
Stankovic MM. Lung Microbiota: From Healthy Lungs to Development of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2025; 26:1403. [PMID: 40003871 PMCID: PMC11854937 DOI: 10.3390/ijms26041403] [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: 12/11/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Lung health is dependent on a complex picture of the lung microbiota composed of bacteriobiota, mycobiota, and virome. The studies have demonstrated that the lung microbiota has a crucial role in host protection by regulating innate and adaptive lung immunity. Chronic obstructive pulmonary disease (COPD) is an inflammatory lung disease featuring changed microbiota composition and diversity, known as a dysbiosis. The lung dysbiosis increases with the progress of COPD and during exacerbation. Two models of dysbiosis have been proposed: dysbiosis and inflammation cycles and the disturbance of bacterial interactome. Still, it is unknown if the driving factor of the pathogenesis of COPD belongs to the host or microbiota. Recently, host-microbiota and microbe-microbe interactions have been highlighted in COPD, but the mechanisms behind these interactions need further exploration. The function of the gut-lung axis is crucial for the maintenance of lung health and is affected in COPD. The application of probiotics has resulted in host-beneficial effects, and it is likely that future progress in this field will aid in the therapy of COPD. In this review, the composition of the lung microbiota, molecular mechanisms, and clinical aspects relating to host and microbiota in health and COPD are comprehensively provided.
Collapse
Affiliation(s)
- Marija M Stankovic
- Group for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia
| |
Collapse
|
4
|
Fadell F, Saliba R, El-Solh AA. Bacteriology of Aspiration Pneumonia: The Lung Microbiome and the Changing Microbial Etiology. Semin Respir Crit Care Med 2024; 45:626-633. [PMID: 39536943 DOI: 10.1055/s-0044-1792111] [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: 11/16/2024]
Abstract
Aspiration pneumonia refers to the process of alveolar inflammation induced by the inhalation of oropharyngeal secretions into the lower respiratory tract. Predisposing factors comprise swallowing dysfunction, impaired cough reflex, and degenerative neurological diseases. Accumulating evidence projects a fading contribution of anaerobic bacteria in aspiration pneumonia at the expense of Gram-negative bacilli, with Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa, becoming the predominant organisms recovered from respiratory specimens. Aspiration of oropharyngeal secretions colonized with respiratory pathogens induces a profound disequilibrium of the lung microbiota resulting in a state of dysbiosis. Understanding this complex temporal variability between microbiome-host associations was only made possible with the introduction of metagenomic sequencing. In this narrative review, we summarize existing knowledge and elaborate on the evolving microbiology of aspiration pneumonia including the link between oral microbiome and pulmonary aspiration. We also highlight the progress and challenges in instituting microbiome-targeted strategies for preventing and treating the sequelae of aspiration pneumonia.
Collapse
Affiliation(s)
- Francois Fadell
- VA Western New York Healthcare System, Research and Development, Buffalo, New York
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University at Buffalo, Buffalo, New York
| | - Ranime Saliba
- VA Western New York Healthcare System, Research and Development, Buffalo, New York
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University at Buffalo, Buffalo, New York
| | - Ali A El-Solh
- VA Western New York Healthcare System, Research and Development, Buffalo, New York
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University at Buffalo, Buffalo, New York
- Department of Anesthesiology, Jacobs School of Medicine, University at Buffalo, Buffalo, New York
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| |
Collapse
|
5
|
Filiz Y, Esposito A, De Maria C, Vozzi G, Yesil-Celiktas O. A comprehensive review on organ-on-chips as powerful preclinical models to study tissue barriers. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 6:042001. [PMID: 39655848 DOI: 10.1088/2516-1091/ad776c] [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: 10/27/2023] [Accepted: 09/04/2024] [Indexed: 12/18/2024]
Abstract
In the preclinical stage of drug development, 2D and 3D cell cultures under static conditions followed by animal models are utilized. However, these models are insufficient to recapitulate the complexity of human physiology. With the developing organ-on-chip (OoC) technology in recent years, human physiology and pathophysiology can be modeled better than traditional models. In this review, the need for OoC platforms is discussed and evaluated from both biological and engineering perspectives. The cellular and extracellular matrix components are discussed from a biological perspective, whereas the technical aspects such as the intricate working principles of these systems, the pivotal role played by flow dynamics and sensor integration within OoCs are elucidated from an engineering perspective. Combining these two perspectives, bioengineering applications are critically discussed with a focus on tissue barriers such as blood-brain barrier, ocular barrier, nasal barrier, pulmonary barrier and gastrointestinal barrier, featuring recent examples from the literature. Furthermore, this review offers insights into the practical utility of OoC platforms for modeling tissue barriers, showcasing their potential and drawbacks while providing future projections for innovative technologies.
Collapse
Affiliation(s)
- Yagmur Filiz
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 8500 Kortrijk, Belgium
| | - Alessio Esposito
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- EgeSAM-Ege University Translational Pulmonary Research Center, Bornova, Izmir, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| |
Collapse
|
6
|
Jameie M, Ahli B, Ghadir S, Azami M, Amanollahi M, Ebadi R, Rafati A, Naser Moghadasi A. The hidden link: How oral and respiratory microbiomes affect multiple sclerosis. Mult Scler Relat Disord 2024; 88:105742. [PMID: 38964239 DOI: 10.1016/j.msard.2024.105742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Extensive research has explored the role of gut microbiota in multiple sclerosis (MS). However, the impact of microbial communities in the oral cavity and respiratory tract on MS is an emerging area of investigation. PURPOSE We aimed to review the current literature related to the nasal, oral, and lung microbiota in people with MS (PwMS). METHODS We conducted a narrative review of clinical and preclinical original studies on PubMed that explored the relationship between the bacterial or viral composition of the nasal, lung, and oral microbiota and MS. Additionally, to find relevant studies not retrieved initially, we also searched for references in related review papers, as well as the references cited within the included studies. RESULTS AND CONCLUSIONS Thirteen studies were meticulously reviewed in three sections; oral microbiota (n = 8), nasal microbiota (n = 3), and lung microbiota (n = 2), highlighting considerable alterations in the oral and respiratory microbiome of PwMS compared to healthy controls (HCs). Genera like Aggregatibacter and Streptococcus were less abundant in the oral microbiota of PwMS compared to HCs, while Staphylococcus, Leptotrichia, Fusobacterium, and Bacteroides showed increased abundance in PwMS. Additionally, the presence of specific bacteria, including Streptococcus sanguinis, within the oral microbiota was suggested to influence Epstein-Barr virus reactivation, a well-established risk factor for MS. Studies related to the nasal microbiome indicated elevated levels of specific Staphylococcus aureus toxins, as well as nasal glial cell infection with human herpes virus (HHV)-6 in PwMS. Emerging research on lung microbiome in animal models demonstrated that manipulating the lung microbiome towards lipopolysaccharide-producing bacteria might suppress MS symptoms. These findings open avenues for potential therapeutic strategies. However, further research is crucial to fully understand the complex interactions between the microbiome and MS. This will help identify the most effective timing, bacterial strains, and modulation techniques.
Collapse
Affiliation(s)
- Melika Jameie
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran; Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Ahli
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Ghadir
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mobin Azami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mobina Amanollahi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Ebadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Rafati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abdorreza Naser Moghadasi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Garmendia J, Cebollero‐Rivas P. Environmental exposures, the oral-lung axis and respiratory health: The airway microbiome goes on stage for the personalized management of human lung function. Microb Biotechnol 2024; 17:e14506. [PMID: 38881505 PMCID: PMC11180993 DOI: 10.1111/1751-7915.14506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 06/18/2024] Open
Abstract
The human respiratory system is constantly exposed to environmental stimuli, sometimes including toxicants, which can trigger dysregulated lung immune responses that lead to respiratory symptoms, impaired lung function and airway diseases. Evidence supports that the microbiome in the lungs has an indispensable role in respiratory health and disease, acting as a local gatekeeper that mediates the interaction between the environmental cues and respiratory health. Moreover, the microbiome in the lungs is intimately intertwined with the oral microbiome through the oral-lung axis. Here, we discuss the intricate three-way relationship between (i) cigarette smoking, which has strong effects on the microbial community structure of the lung; (ii) microbiome dysbiosis and disease in the oral cavity; and (iii) microbiome dysbiosis in the lung and its causal role in patients suffering chronic obstructive pulmonary disease (COPD), a leading cause of morbidity and mortality worldwide. We highlight exciting outcomes arising from recently established interactions in the airway between environmental exposures, microbiome, metabolites-functional attributes and the host, as well as how these associations have the potential to predict the respiratory health status of the host through an airway microbiome health index. For completion, we argue that incorporating (synthetic) microbial community ecology in our contemporary understanding of lung disease presents challenges and also rises novel opportunities to exploit the oral-lung axis and its microbiome towards innovative airway disease diagnostics, prognostics, patient stratification and microbiota-targeted clinical interventions in the context of current therapies.
Collapse
Affiliation(s)
- Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de NavarraMutilvaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Pilar Cebollero‐Rivas
- Servicio de NeumologíaHospital Universitario de NavarraNavarraSpain
- Universidad Pública de Navarra (UPNa)NavarraSpain
| |
Collapse
|
8
|
Marrella V, Nicchiotti F, Cassani B. Microbiota and Immunity during Respiratory Infections: Lung and Gut Affair. Int J Mol Sci 2024; 25:4051. [PMID: 38612860 PMCID: PMC11012346 DOI: 10.3390/ijms25074051] [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: 02/02/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Bacterial and viral respiratory tract infections are the most common infectious diseases, leading to worldwide morbidity and mortality. In the past 10 years, the importance of lung microbiota emerged in the context of pulmonary diseases, although the mechanisms by which it impacts the intestinal environment have not yet been fully identified. On the contrary, gut microbial dysbiosis is associated with disease etiology or/and development in the lung. In this review, we present an overview of the lung microbiome modifications occurring during respiratory infections, namely, reduced community diversity and increased microbial burden, and of the downstream consequences on host-pathogen interaction, inflammatory signals, and cytokines production, in turn affecting the disease progression and outcome. Particularly, we focus on the role of the gut-lung bidirectional communication in shaping inflammation and immunity in this context, resuming both animal and human studies. Moreover, we discuss the challenges and possibilities related to novel microbial-based (probiotics and dietary supplementation) and microbial-targeted therapies (antibacterial monoclonal antibodies and bacteriophages), aimed to remodel the composition of resident microbial communities and restore health. Finally, we propose an outlook of some relevant questions in the field to be answered with future research, which may have translational relevance for the prevention and control of respiratory infections.
Collapse
Affiliation(s)
- Veronica Marrella
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, 20138 Milan, Italy;
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Federico Nicchiotti
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20089 Milan, Italy;
| | - Barbara Cassani
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20089 Milan, Italy;
| |
Collapse
|
9
|
Alrezaihi A, Penrice-Randal R, Dong X, Prince T, Randle N, Semple MG, Openshaw PJM, MacGill T, Myers T, Orr R, Zakotnik S, Suljič A, Avšič-Županc T, Petrovec M, Korva M, AlJabr W, Hiscox JA. Enrichment of SARS-CoV-2 sequence from nasopharyngeal swabs whilst identifying the nasal microbiome. J Clin Virol 2024; 171:105620. [PMID: 38237303 DOI: 10.1016/j.jcv.2023.105620] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/06/2023] [Accepted: 11/18/2023] [Indexed: 03/17/2024]
Abstract
Simultaneously characterising the genomic information of coronaviruses and the underlying nasal microbiome from a single clinical sample would help characterise infection and disease. Metatranscriptomic approaches can be used to sequence SARS-CoV-2 (and other coronaviruses) and identify mRNAs associated with active transcription in the nasal microbiome. However, given the large sequence background, unenriched metatranscriptomic approaches often do not sequence SARS-CoV-2 to sufficient read and coverage depth to obtain a consensus genome, especially with moderate and low viral loads from clinical samples. In this study, various enrichment methods were assessed to detect SARS-CoV-2, identify lineages and define the nasal microbiome. The methods were underpinned by Oxford Nanopore long-read sequencing and variations of sequence independent single primer amplification (SISPA). The utility of the method(s) was also validated on samples from patients infected seasonal coronaviruses. The feasibility of profiling the nasal microbiome using these enrichment methods was explored. The findings shed light on the performance of different enrichment strategies and their applicability in characterising the composition of the nasal microbiome.
Collapse
Affiliation(s)
| | | | | | | | | | - Malcolm G Semple
- University of Liverpool, Liverpool, UK; NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK; Alder Hey Children's Hospital, Liverpool, UK
| | | | - Tracy MacGill
- Office of Counterterrorism and Emerging Threats, U.S. Food and Drug Administration, Silver Spring, USA
| | - Todd Myers
- Office of Counterterrorism and Emerging Threats, U.S. Food and Drug Administration, Silver Spring, USA
| | - Robert Orr
- Office of Counterterrorism and Emerging Threats, U.S. Food and Drug Administration, Silver Spring, USA
| | | | - Alen Suljič
- University of Ljubljana, Ljubljana, Slovenia
| | | | | | - Miša Korva
- University of Ljubljana, Ljubljana, Slovenia
| | - Waleed AlJabr
- University of Liverpool, Liverpool, UK; King Fahad Medical City, Riyadh, Saudi Arabia
| | - Julian A Hiscox
- University of Liverpool, Liverpool, UK; NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK; Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
10
|
Zheng L, Liu C, Wang H, Zhang J, Mao L, Dong X, Hu S, Li N, Pi D, Qiu J, Xu F, Chen C, Zou Z. Intact lung tissue and bronchoalveolar lavage fluid are both suitable for the evaluation of murine lung microbiome in acute lung injury. MICROBIOME 2024; 12:56. [PMID: 38494479 PMCID: PMC10946114 DOI: 10.1186/s40168-024-01772-6] [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: 06/07/2023] [Accepted: 01/30/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Accumulating clinical evidence suggests that lung microbiome is closely linked to the progression of pulmonary diseases; however, it is still controversial which specimen type is preferred for the evaluation of lung microbiome. METHODS AND RESULTS To address this issue, we established a classical acute lung injury (ALI) mice model by intratracheal instillation of lipopolysaccharides (LPS). We found that the bacterial DNA obtained from the bronchoalveolar lavage fluid (BALF), intact lung tissue [Lung(i)], lung tissue after perfused [Lung(p)], and feces of one mouse were enough for 16S rRNA sequencing, except the BALF of mice treated with phosphate buffer saline (PBS), which might be due to the biomass of lung microbiome in the BALF were upregulated in the mice treated with LPS. Although the alpha diversity among the three specimens from lungs had minimal differences, Lung(p) had higher sample-to-sample variation compared with BALF and Lung(i). Consistently, PCoA analysis at phylum level indicated that BALF was similar to Lung(i), but not Lung(p), in the lungs of mice treated with LPS, suggesting that BALF and Lung(i) were suitable for the evaluation of lung microbiome in ALI. Importantly, Actinobacteria and Firmicutes were identified as the mostly changed phyla in the lungs and might be important factors involved in the gut-lung axis in ALI mice. Moreover, Actinobacteria and Proteobacteria might play indicative roles in the severity of lung injury. CONCLUSION This study shows both Lung(i) and BALF are suitable for the evaluation of murine lung microbiome in ALI, and several bacterial phyla, such as Actinobacteria, may serve as potential biomarkers for the severity of ALI. Video Abstract.
Collapse
Affiliation(s)
- Lijun Zheng
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Chengjun Liu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Hongjing Wang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing, 400016, People's Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaomei Dong
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Siyao Hu
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Na Li
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dandan Pi
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing, 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Research Center for Environment and Human Health, School of Public Health, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
11
|
Yang W, Xi C, Yao H, Yuan Q, Zhang J, Chen Q, Wu G, Hu J. Oral administration of lysozyme protects against injury of ileum via modulating gut microbiota dysbiosis after severe traumatic brain injury. Front Cell Infect Microbiol 2024; 14:1304218. [PMID: 38352055 PMCID: PMC10861676 DOI: 10.3389/fcimb.2024.1304218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
Objective The current study sought to clarify the role of lysozyme-regulated gut microbiota and explored the potential therapeutic effects of lysozyme on ileum injury induced by severe traumatic brain injury (sTBI) and bacterial pneumonia in vivo and in vitro experiments. Methods Male 6-8-week-old specific pathogen-free (SPF) C57BL/6 mice were randomly divided into Normal group (N), Sham group (S), sTBI group (T), sTBI + or Lysozyme-treated group (L), Normal + Lysozyme group (NL) and Sham group + Lysozyme group (SL). At the day 7 after establishment of the model, mice were anesthetized and the samples were collected. The microbiota in lungs and fresh contents of the ileocecum were analyzed. Lungs and distal ileum were used to detect the degree of injury. The number of Paneth cells and the expression level of lysozyme were assessed. The bacterial translocation was determined. Intestinal organoids culture and co-coculture system was used to test whether lysozyme remodels the intestinal barrier through the gut microbiota. Results After oral administration of lysozyme, the intestinal microbiota is rebalanced, the composition of lung microbiota is restored, and translocation of intestinal bacteria is mitigated. Lysozyme administration reinstates lysozyme expression in Paneth cells, thereby reducing intestinal permeability, pathological score, apoptosis rate, and inflammation levels. The gut microbiota, including Oscillospira, Ruminococcus, Alistipes, Butyricicoccus, and Lactobacillus, play a crucial role in regulating and improving intestinal barrier damage and modulating Paneth cells in lysozyme-treated mice. A co-culture system comprising intestinal organoids and brain-derived proteins (BP), which demonstrated that the BP effectively downregulated the expression of lysozyme in intestinal organoids. However, supplementation of lysozyme to this co-culture system failed to restore its expression in intestinal organoids. Conclusion The present study unveiled a virtuous cycle whereby oral administration of lysozyme restores Paneth cell's function, mitigates intestinal injury and bacterial translocation through the remodeling of gut microbiota.
Collapse
Affiliation(s)
- Weijian Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Caihua Xi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haijun Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Yuan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jun Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Qifang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gang Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| |
Collapse
|
12
|
Zolnikova O, Dzhakhaya N, Bueverova E, Sedova A, Kurbatova A, Kryuchkova K, Butkova T, Izotov A, Kulikova L, Yurku K, Chekulaev P, Zaborova V. The Contribution of the Intestinal Microbiota to the Celiac Disease Pathogenesis along with the Effectiveness of Probiotic Therapy. Microorganisms 2023; 11:2848. [PMID: 38137992 PMCID: PMC10745538 DOI: 10.3390/microorganisms11122848] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
The development of many human disorders, including celiac disease (CD), is thought to be influenced by the microbiota of the gastrointestinal tract and its metabolites, according to current research. This study's goal was to provide a concise summary of the information on the contribution of the intestinal microbiota to the CD pathogenesis, which was actively addressed while examining the reported pathogenesis of celiac disease (CD). We assumed that a change in gluten tolerance is formed under the influence of a number of different factors, including genetic predisposition and environmental factors. In related investigations, researchers have paid increasing attention to the study of disturbances in the composition of the intestinal microbiota and its functional activity in CD. A key finding of our review is that the intestinal microbiota has gluten-degrading properties, which, in turn, may have a protective effect on the development of CD. The intestinal microbiota contributes to maintaining the integrity of the intestinal barrier, preventing the formation of a "leaky" intestine. On the contrary, a change in the composition of the microbiota can act as a significant link in the pathogenesis of gluten intolerance and exacerbate the course of the disease. The possibility of modulating the composition of the microbiota by prescribing probiotic preparations is being considered. The effectiveness of the use of probiotics containing Lactobacillus and Bifidobacterium bacteria in experimental and clinical studies as a preventive and therapeutic agent has been documented.
Collapse
Affiliation(s)
- Oxana Zolnikova
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| | - Natiya Dzhakhaya
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| | - Elena Bueverova
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| | - Alla Sedova
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| | - Anastasia Kurbatova
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| | - Kira Kryuchkova
- Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Tatyana Butkova
- Institute of Biomedical Chemistry, Biobanking Group, 109028 Moscow, Russia; (T.B.); (A.I.); (L.K.)
| | - Alexander Izotov
- Institute of Biomedical Chemistry, Biobanking Group, 109028 Moscow, Russia; (T.B.); (A.I.); (L.K.)
| | - Ludmila Kulikova
- Institute of Biomedical Chemistry, Biobanking Group, 109028 Moscow, Russia; (T.B.); (A.I.); (L.K.)
- Institute of Mathematical Problems of Biology RAS—The Branch of Keldysh Institute of Applied Mathematics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Kseniya Yurku
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia;
| | - Pavel Chekulaev
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| | - Victoria Zaborova
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (O.Z.); (N.D.); (E.B.); (A.S.); (A.K.); (P.C.)
| |
Collapse
|