1
|
Stewart AG, Chambers DC. Primary graft dysfunction, dysbiosis, and innate immune activation after lung transplantation. J Heart Lung Transplant 2024:S1053-2498(24)02016-3. [PMID: 39672290 DOI: 10.1016/j.healun.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024] Open
Affiliation(s)
- Adam G Stewart
- Transplant and Immunocompromised Host Infectious Diseases, Department of Medicine, Infectious Diseases Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Daniel C Chambers
- The Medical School, Faculty of Medicine, The University of Queensland, Brisbane, Australia; The Prince Charles Hospital, Brisbane, Australia.
| |
Collapse
|
2
|
Medina CK, Aykut B. Gut Microbial Dysbiosis and Implications in Solid Organ Transplantation. Biomedicines 2024; 12:2792. [PMID: 39767699 PMCID: PMC11673786 DOI: 10.3390/biomedicines12122792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome has been shown to play a significant role in solid organ transplantation, potentially influencing graft function and patient outcomes. Dysbiosis, characterized by reduced microbial diversity and an increase in pathogenic taxa, has been linked to higher incidences of allograft rejection, graft dysfunction, and post-transplant mortality. Several studies suggest that the gut microbiome might be able to serve as both a biomarker and a therapeutic target, potentially guiding personalized immunosuppressive therapies and other interventions to improve outcomes after solid organ transplantation. As summarized in this review, clinical studies have shown that specific microbial shifts correlate with adverse outcomes, including acute rejection and chronic allograft dysfunction. As research surrounding the relationship between the gut microbiome and solid organ transplant progresses, the integration of microbial analysis into clinical practice has the potential to revolutionize post-transplant care, offering new avenues to improve graft survival and patient quality of life. This review aims to provide a comprehensive overview of the relationship between gut microbial dysbiosis and transplantation outcomes, emphasizing the impact on kidney, liver, lung, and heart transplant recipients.
Collapse
Affiliation(s)
| | - Berk Aykut
- Department of Surgery, Duke University, Durham, NC 27710, USA
| |
Collapse
|
3
|
Grando L, Boada M, Faner R, Gómez-Ollés S, Ruiz V, Bohils M, Albiol J, Marrero R, Rosell L, Salinas I, Ruiz D, Ruiz Á, Rodríguez-Villar C, Ureña A, Paredes-Zapata D, Guirao Á, Sánchez-Etayo G, Molins L, Quiroga N, Gómez-Brey A, Michavila X, Sandiumenge A, Agustí À, Ramos R, Bello I. The impact of the EVLP on the lung microbiome and its inflammatory reaction. Transpl Int 2024; 37:12979. [PMID: 39588198 PMCID: PMC11587763 DOI: 10.3389/ti.2024.12979] [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: 03/12/2024] [Accepted: 10/01/2024] [Indexed: 11/27/2024]
Abstract
The pulmonary microbiome has emerged as a significant factor in respiratory health and diseases. Despite the sterile conditions maintained during ex vivo lung perfusion (EVLP), the use of antibiotics in the perfuse liquid can lead to dynamic changes in the lung microbiome. Here, we present the design of a study that aims to investigate the hypothesis that EVLP alters the lung microbiome and induces tissue inflammation. This pilot, prospective, controlled study will be conducted in two Spanish donor centers and will include seven organ donors after brain death or after controlled cardiac death. After standardized retrieval, the left lung will be preserved in cold storage and the right lung will be perfused with EVLP. Samples from bronchoalveolar lavage, perfusion and preservation solutions, and lung biopsies will be collected from both lungs and changes in lung microbiome and inflammatory response will be compared.
Collapse
Affiliation(s)
- Leandro Grando
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Marc Boada
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Rosa Faner
- Universitat de Barcelona, Catedra Salut Respiratoria, Barcelona, Catalunya, Spain
- CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Institut d’Investigacions BIomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Universitat de Barcelona, Biomedicine Department, Barcelona, Catalunya, Spain
| | - Susana Gómez-Ollés
- CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Pneumology, Vall d’Hebron Institut de Recerca, Barcelona, Spain
| | - Victoria Ruiz
- Department of Pneumology, Vall d’Hebron Institut de Recerca, Barcelona, Spain
| | - Marc Bohils
- Department of Donor and Transplant Coordination, Hospital Clínic, Barcelona, Spain
| | - Joaquim Albiol
- Department of Donor and Transplant Coordination, Hospital Clínic, Barcelona, Spain
| | - Ramses Marrero
- Department of Anaesthesia and Perioperative Care, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Laia Rosell
- Surgical Area, Hospital Clínic, Barcelona, Spain
| | - Ivan Salinas
- Surgical Area, Hospital Clínic, Barcelona, Spain
| | - Daniel Ruiz
- Surgical Area, Hospital Clínic, Barcelona, Spain
| | - Ángel Ruiz
- Department of Donor and Transplant Coordination, Hospital Clínic, Barcelona, Spain
| | | | - Anna Ureña
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - David Paredes-Zapata
- Department of Donor and Transplant Coordination, Hospital Clínic, Barcelona, Spain
| | - Ángela Guirao
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Gerard Sánchez-Etayo
- Department of Donor and Transplant Coordination, Hospital Clínic, Barcelona, Spain
| | - Laureano Molins
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Néstor Quiroga
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Aroa Gómez-Brey
- Department of Donor and Transplant Coordination, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Xavier Michavila
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Alberto Sandiumenge
- Department of Donor and Transplant Coordination, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Àlvar Agustí
- Universitat de Barcelona, Catedra Salut Respiratoria, Barcelona, Catalunya, Spain
- CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Pneumology, Respiratory Institute, Hospital Clínic, Barcelona, Spain
| | - Ricard Ramos
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions BIomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Irene Bello
- Department of Thoracic Surgery, Respiratory Institute, Hospital Clínic, Barcelona, Spain
- Institut d’Investigacions BIomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
4
|
Garaci E, Pariano M, Nunzi E, Costantini C, Bellet MM, Antognelli C, Russo MA, Romani L. Bacteria and fungi of the lung: allies or enemies? Front Pharmacol 2024; 15:1497173. [PMID: 39584143 PMCID: PMC11584946 DOI: 10.3389/fphar.2024.1497173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024] Open
Abstract
Moving from the earlier periods in which the lungs were believed to represent sterile environments, our knowledge on the lung microbiota has dramatically increased, from the first descriptions of the microbial communities inhabiting the healthy lungs and the definition of the ecological rules that regulate its composition, to the identification of the changes that occur in pathological conditions. Despite the limitations of lung as a microbiome reservoir due to the low microbial biomass and abundance, defining its microbial composition and function in the upper and lower airways may help understanding the impact on local homeostasis and its disruption in lung diseases. In particular, the understanding of the metabolic and immune significance of microbes, their presence or lack thereof, in health and disease states could be valuable in development of novel druggable targets in disease treatments. Next-generation sequencing has identified intricate inter-microbe association networks that comprise true mutualistic or antagonistic direct or indirect relationships in the respiratory tract. In this review, the tripartite interaction of bacteria, fungi and the mammalian host is addressed to provide an integrated view of the microbial-host cross-talk in lung health and diseases from an immune and metabolic perspective.
Collapse
Affiliation(s)
- Enrico Garaci
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luigina Romani
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
5
|
Martin C, Mahan KS, Wiggen TD, Gilbertsen AJ, Hertz MI, Hunter RC, Quinn RA. Microbiome and metabolome patterns after lung transplantation reflect underlying disease and chronic lung allograft dysfunction. MICROBIOME 2024; 12:196. [PMID: 39385282 PMCID: PMC11462767 DOI: 10.1186/s40168-024-01893-y] [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: 05/24/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Progression of chronic lung disease may lead to the requirement for lung transplant (LTx). Despite improvements in short-term survival after LTx, chronic lung allograft dysfunction (CLAD) remains a critical challenge for long-term survival. This study investigates the molecular and microbial relationships between underlying lung disease and the development of CLAD in bronchoalveolar lavage fluid (BALF) from subjects post-LTx, which is crucial for tailoring treatment strategies specific to allograft dysfunctions. METHODS Paired 16S rRNA gene amplicon sequencing and untargeted LC-MS/MS metabolomics were performed on 856 BALF samples collected over 10 years from LTx recipients (n = 195) with alpha-1-antitrypsin disease (AATD, n = 23), cystic fibrosis (CF, n = 47), chronic obstructive pulmonary disease (COPD, n = 78), or pulmonary fibrosis (PF, n = 47). Data were analyzed using random forest (RF) machine learning and multivariate statistics for associations with underlying disease and CLAD development. RESULTS The BALF microbiome and metabolome after LTx differed significantly according to the underlying disease state (PERMANOVA, p = 0.001), with CF and AATD demonstrating distinct microbiome and metabolome profiles, respectively. Uniqueness in CF was mainly driven by Pseudomonas abundance and its metabolites, whereas AATD had elevated levels of phenylalanine and a lack of shared metabolites with the other underlying diseases. BALF microbiome and metabolome composition were also distinct between those who did or did not develop CLAD during the sample collection period (PERMANOVA, p = 0.001). An increase in the average abundance of Veillonella (AATD, COPD) and Streptococcus (CF, PF) was associated with CLAD development, and decreases in the abundance of phenylalanine-derivative alkaloids (CF, COPD) and glycerophosphorylcholines (CF, COPD, PF) were signatures of the CLAD metabolome. Although the relative abundance of Pseudomonas was not associated with CLAD, the abundance of its virulence metabolites, including siderophores, quorum-sensing quinolones, and phenazines, were elevated in those with CF who developed CLAD. There was a positive correlation between the abundance of these molecules and the abundance of Pseudomonas in the microbiome, but there was no correlation between their abundance and the time in which BALF samples were collected post-LTx. CONCLUSIONS The BALF microbiome and metabolome after LTx are particularly distinct in those with underlying CF and AATD. These data reflect those who developed CLAD, with increased virulence metabolite production from Pseudomonas, an aspect of CF CLAD cases. These findings shed light on disease-specific microbial and metabolic signatures in LTx recipients, offering valuable insights into the underlying causes of allograft rejection. Video Abstract.
Collapse
Affiliation(s)
- Christian Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Kathleen S Mahan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Talia D Wiggen
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Adam J Gilbertsen
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Marshall I Hertz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Ryan C Hunter
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14051, USA.
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
6
|
Ponholzer F, Bogensperger C, Krendl FJ, Krapf C, Dumfarth J, Schneeberger S, Augustin F. Beyond the organ: lung microbiome shapes transplant indications and outcomes. Eur J Cardiothorac Surg 2024; 66:ezae338. [PMID: 39288305 PMCID: PMC11466426 DOI: 10.1093/ejcts/ezae338] [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: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 09/19/2024] Open
Abstract
The lung microbiome plays a crucial role in the development of chronic lung diseases, which may ultimately lead to the need for lung transplantation. Also, perioperative results seem to be connected with altered lung microbiomes and its dynamic changes providing a possible target for optimizing short-term outcome after transplantation. A literature review using MEDLINE, PubMed Central and Bookshelf was performed. Chronic lung allograft dysfunction (CLAD) seems to be influenced and partly triggered by changes in the pulmonary microbiome and dysbiosis, e.g. through increased bacterial load or abundance of specific species such as Pseudomonas aeruginosa. Additionally, the specific indications for transplantation, with their very heterogeneous changes and influences on the pulmonary microbiome, influence long-term outcome. Next to composition and measurable bacterial load, dynamic changes in the allografts microbiome also possess the ability to alter long-term outcomes negatively. This review discusses the "new" microbiome after transplantation and the associations with direct postoperative outcome. With the knowledge of these principles the impact of alterations in the pulmonary microbiome in hindsight to CLAD and possible therapeutic implications are described and discussed. The aim of this review is to summarize the current literature regarding pre- and postoperative lung microbiomes and how they influence different lung diseases on their progression to failure of conservative treatment. This review provides a summary of current literature for centres looking for further options in optimizing lung transplant outcomes and highlights possible areas for further research activities investigating the pulmonary microbiome in connection to transplantation.
Collapse
Affiliation(s)
- Florian Ponholzer
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Bogensperger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Julius Krendl
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Krapf
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Dumfarth
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Augustin
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
7
|
Snyder ME, Kitsios GD. Lung Transplant Outcomes Keep BUGging us: Acute Cellular Rejection and the Lung Microbiome. Am J Respir Crit Care Med 2024; 209:1423-1425. [PMID: 38564413 PMCID: PMC11208966 DOI: 10.1164/rccm.202403-0499ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 04/04/2024] Open
Affiliation(s)
- Mark E Snyder
- Department of Medicine University of Pittsburgh Pittsburgh, Pennsylvania
| | - Georgios D Kitsios
- Department of Medicine University of Pittsburgh Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Courjon J, Neofytos D, van Delden C. Bacterial infections in solid organ transplant recipients. Curr Opin Organ Transplant 2024; 29:155-160. [PMID: 38205868 DOI: 10.1097/mot.0000000000001134] [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: 01/12/2024]
Abstract
PURPOSE OF REVIEW Bacteria are the leading cause of infections in solid organ transplant (SOT) recipients, significantly impacting patient outcome. Recently detailed and comprehensive epidemiological data have been published. RECENT FINDING This literature review aims to provide an overview of bacterial infections affecting different types of SOT recipients, emphasizing underlying risk factors and pathophysiological mechanisms. SUMMARY Lung transplantation connects two microbiotas: one derived from the donor's lower respiratory tract with one from the recipient's upper respiratory tract. Similarly, liver transplantation involves a connection to the digestive tract and its microbiota through the bile ducts. For heart transplant recipients, specific factors are related to the management strategies for end-stage heart failure based with different circulatory support tools. Kidney and kidney-pancreas transplant recipients commonly experience asymptomatic bacteriuria, but recent studies have suggested the absence of benefice of routine treatment. Bloodstream infections (BSI) are frequent and affect all SOT recipients. Nonorgan-related risk factors as age, comorbidity index score, and leukopenia contribute to BSI development. Bacterial opportunistic infections have become rare in the presence of efficient prophylaxis. Understanding the epidemiology, risk factors, and pathophysiology of bacterial infections in SOT recipients is crucial for effective management and improved patient outcomes.
Collapse
Affiliation(s)
- Johan Courjon
- Transplant Infectious Diseases Unit, Service of Infectious Diseases, University Hospitals Geneva, Geneva, Switzerland
- Université Côte d'Azur, Inserm, C3M, Nice, France
| | - Dionysios Neofytos
- Transplant Infectious Diseases Unit, Service of Infectious Diseases, University Hospitals Geneva, Geneva, Switzerland
| | - Christian van Delden
- Transplant Infectious Diseases Unit, Service of Infectious Diseases, University Hospitals Geneva, Geneva, Switzerland
| |
Collapse
|
9
|
Lian Q, Song X, Yang J, Wang L, Xu P, Wang X, Xu X, Yang B, He J, Ju C. Alterations of lung microbiota in lung transplant recipients with pneumocystis jirovecii pneumonia. Respir Res 2024; 25:125. [PMID: 38486264 PMCID: PMC10941442 DOI: 10.1186/s12931-024-02755-9] [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: 01/21/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Increasing evidence revealed that lung microbiota dysbiosis was associated with pulmonary infection in lung transplant recipients (LTRs). Pneumocystis jirovecii (P. jirovecii) is an opportunistic fungal pathogen that frequently causes lethal pneumonia in LTRs. However, the lung microbiota in LTRs with P. jirovecii pneumonia (PJP) remains unknow. METHODS In this prospective observational study, we performed metagenomic next-generation sequencing (mNGS) on 72 bronchoalveolar lavage fluid (BALF) samples from 61 LTRs (20 with PJP, 22 with PJC, 19 time-matched stable LTRs, and 11 from LTRs after PJP recovery). We compared the lung microbiota composition of LTRs with and without P. jirovecii, and analyzed the related clinical variables. RESULTS BALFs collected at the episode of PJP showed a more discrete distribution with a lower species diversity, and microbiota composition differed significantly compared to P. jirovecii colonization (PJC) and control group. Human gammaherpesvirus 4, Phreatobacter oligotrophus, and Pseudomonas balearica were the differential microbiota species between the PJP and the other two groups. The network analysis revealed that most species had a positive correlation, while P. jirovecii was correlated negatively with 10 species including Acinetobacter venetianus, Pseudomonas guariconensis, Paracandidimonas soli, Acinetobacter colistiniresistens, and Castellaniella defragrans, which were enriched in the control group. The microbiota composition and diversity of BALF after PJP recovery were also different from the PJP and control groups, while the main components of the PJP recovery similar to control group. Clinical variables including age, creatinine, total protein, albumin, IgG, neutrophil, lymphocyte, CD3+CD45+, CD3+CD4+ and CD3+CD8+ T cells were deeply implicated in the alterations of lung microbiota in LTRs. CONCLUSIONS This study suggests that LTRs with PJP had altered lung microbiota compared to PJC, control, and after recovery groups. Furthermore, lung microbiota is related to age, renal function, nutritional and immune status in LTRs.
Collapse
Affiliation(s)
- Qiaoyan Lian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China
| | - Xiuling Song
- Vision Medicals Co., Ltd, 510700, Guangzhou, Guangdong, P.R. China
| | - Juhua Yang
- Vision Medicals Co., Ltd, 510700, Guangzhou, Guangdong, P.R. China
| | - Lulin Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China
| | - Peihang Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China
| | - Xiaohua Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China
| | - Xin Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China
- Department of Thoracic Surgery, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China
| | - Bin Yang
- Vision Medicals Co., Ltd, 510700, Guangzhou, Guangdong, P.R. China
| | - Jianxing He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China.
- Department of Thoracic Surgery, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China.
| | - Chunrong Ju
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ transplantation, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 510120, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
10
|
Yu Y, Kim YH, Cho WH, Kim D, So MW, Son BS, Yeo HJ. Unique Changes in the Lung Microbiome following the Development of Chronic Lung Allograft Dysfunction. Microorganisms 2024; 12:287. [PMID: 38399691 PMCID: PMC10893466 DOI: 10.3390/microorganisms12020287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The importance of lung microbiome changes in developing chronic lung allograft dysfunction (CLAD) after lung transplantation is poorly understood. The lung microbiome-immune interaction may be critical in developing CLAD. In this context, examining alterations in the microbiome and immune cells of the lungs following CLAD, in comparison to the lung condition immediately after transplantation, can offer valuable insights. Four adult patients who underwent lung retransplantation between January 2019 and June 2020 were included in this study. Lung tissues were collected from the same four individuals at two different time points: at the time of the first transplant and at the time of the explantation of CLAD lungs at retransplantation due to CLAD. We analyzed whole-genome sequencing using the Kraken2 algorithm and quantified the cell fractionation from the bulk tissue gene expression profile for each lung tissue. Finally, we compared the differences in lung microbiome and immune cells between the lung tissues of these two time points. The median age of the recipients was 57 years, and most (75%) had undergone lung transplants for idiopathic pulmonary fibrosis. All patients were administered basiliximab for induction therapy and were maintained on three immunosuppressants. The median CLAD-free survival term was 693.5 days, and the median time to redo the lung transplant was 843.5 days. Bacterial diversity was significantly lower in the CLAD lungs than at transplantation. Bacterial diversity tended to decrease according to the severity of the CLAD. Aerococcus, Caldiericum, Croceibacter, Leptolyngbya, and Pulveribacter genera were uniquely identified in CLAD, whereas no taxa were identified in lungs at transplantation. In particular, six taxa, including Croceibacter atlanticus, Caldiserium exile, Dolichospermum compactum, Stappia sp. ES.058, Kinetoplastibacterium sorsogonicusi, and Pulveribacter suum were uniquely detected in CLAD. Among immune cells, CD8+ T cells were significantly increased, while neutrophils were decreased in the CLAD lung. In conclusion, unique changes in lung microbiome and immune cell composition were confirmed in lung tissue after CLAD compared to at transplantation.
Collapse
Affiliation(s)
- Yeuni Yu
- Biomedical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Yun Hak Kim
- Department of Anatomy and Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Woo Hyun Cho
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Dohyung Kim
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Min Wook So
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Bong Soo Son
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Hye Ju Yeo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| |
Collapse
|
11
|
Wang P, Wang J, Wang L, Lv J, Shao Y, He D. High throughput sequencing technology reveals alteration of lower respiratory tract microbiome in severe aspiration pneumonia and its association with inflammation. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 116:105533. [PMID: 37995886 DOI: 10.1016/j.meegid.2023.105533] [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: 08/05/2023] [Revised: 10/18/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Aspiration pneumonia is a common and severe clinical condition. The microbiome present in the lower respiratory tract plays a crucial role in regulating human inflammatory response. However, the relationship between the altered lower respiratory tract microbiome and inflammation in aspiration pneumonia remains inadequately explored. PURPOSE To investigate the alteration of the lower respiratory tract microbiome in severe aspiration pneumonia patients and explore the potential correlation between microbiome components and inflammatory response. METHOD Patients in the severe aspiration pneumonia group and control group were enrolled from the intensive care unit of Jinshan Hospital, Fudan University between December 31, 2020 and August 19, 2021. Sputum specimens were collected from all participants and subsequently subjected to 16S rDNA high throughput sequencing technology. The concentration of inflammatory cytokines in serum was measured using enzyme-linked immunosorbent assay (ELISA) kits, and collected data including patients' demographic information, clinical data, and laboratory examination results were recorded for further analysis. RESULTS Alteration in the lower respiratory tract microbiome was observed in severe aspiration pneumonia. Compared to the control group, a significant decrease in the relative abundance of Firmicutes was found at the phylum level (P < 0.01). At the family level, the relative abundance of Corynebacteriaceae, Enterobacteriaceae and Enterococcaceae increased significantly (P < 0.001, P < 0.05, P < 0.01). There were no significant differences in community diversity of the lower respiratory tract between the two groups. Patients in the severe aspiration pneumonia group exhibited significantly higher levels of inflammation compared to those in the control group. Correlation analysis showed that the relative abundance of Corynebacteriaceae was positively correlated with the expression level of IL-1β and IL-18 (P = 0.002, P = 0.02); the relative abundance of Enterobacteriaceae was negatively correlated with IL-4 (P = 0.011); no other significant correlations have been identified between microbiome and inflammatory indicators thus far (P > 0.05). CONCLUSIONS Alteration of the lower respiratory tract microbiome is critically involved in inflammation and disease progression in severe cases of aspiration pneumonia. The potential inflammation regulation properties of the microbiome hold promising value for developing novel therapeutic approaches aimed at mitigating the severity of the disease.
Collapse
Affiliation(s)
- Pengfei Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Junming Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Lina Wang
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Jiang Lv
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Yiru Shao
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Daikun He
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai 201508, China; Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China.
| |
Collapse
|
12
|
Wu Y, Huang L, Li M, Cui X, Zhan Q, Wang C. The role of lung microbiota in primary graft dysfunction in lung transplant recipients. Clin Transplant 2023; 37:e15152. [PMID: 37788167 DOI: 10.1111/ctr.15152] [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: 06/21/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND Recent studies have shown that the lung microbiota is altered in critically ill patients and predicts clinical outcomes. Primary graft dysfunction (PGD) is a common complication and a leading cause of death within 1 month of lung transplantation, but the clinical significance of changes in the lung bacterial community during PGD is unclear. The aim of this study was to determine the contribution of the lung microbiota to the development and course of severe PGD. METHODS We conducted a retrospective study to characterize the lung microbiota of 32 lung transplant patients with combined PGD using next-generation sequencing of bronchoalveolar lavage samples. The relationship between lung flora dysbiosis and lung immunity in PGD was assessed by quantification of alveolar cytokines. The contribution of microbiota characteristics to patient outcomes was assessed by estimating overall survival. RESULTS Patients diagnosed with PGD grade 3 showed a reduction in alpha diversity, driven by a significant increase in the abundance of the genera Modestobacter, Scardovia and Selenomonas, and a reduction in the proportion of the genera Klebsiella and Oribacterium. Alpha diversity of the lung microbiota in PGD3 patients was negatively correlated with BALF interleukin (IL)-2 (r = -.752, p < .05). In addition, bacterial diversity in the lung microbiota of non-survivors was lower than that of survivors (p = .041). CONCLUSIONS There is variation in the lung microbiota of PGD grade 3 patients and dysbiosis of the lung microbiota is associated with lung immunity. The lung microbiota has potential in the diagnosis and treatment of PGD grade 3.
Collapse
Affiliation(s)
- Yuhan Wu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Harbin Medical University, Harbin, China
- Harbin Medical University, Harbin, China
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Linna Huang
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Min Li
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoyang Cui
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qingyuan Zhan
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Chen Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Harbin Medical University, Harbin, China
- Harbin Medical University, Harbin, China
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
13
|
Abstract
Patients with chronic lung disease and lung transplantation have high rates of colonization and infection from multidrug-resistant (MDR) organisms. This article summarizes the current state of knowledge regarding phage therapy in the setting of lung transplantation. Phage therapy has been used in several lung transplant candidates and recipients on a compassionate use basis targeting mostly MDR gram-negative infections and atypical mycobacterial infections with demonstrated clinical safety. Phage biodistribution given intravenously or via nebulization has not been extensively studied, though preliminary data are presented. Phage interacts with both the innate and adaptive immune system; current literature demonstrates the development of serum neutralization in some cases of phage therapy, although the clinical impact seems variable. A summary of current clinical trials involving patients with chronic lung disease is presented, though none are specifically targeting lung transplant candidates or recipients. In addition to treatment of active infections, a variety of clinical scenarios may benefit from phage therapy, and well-designed clinical trials involving this vulnerable patient population are needed: pre- or peritransplantation use of phage in the setting of MDR organism colonization may lead to waitlisting of candidates currently declined by many centers, along with potential reduction of waitlist mortality rates and posttransplant infections; phage may be used for biofilm-related bronchial stent infections; and, finally, there is a possibility that phage use can affect allograft function and chronic rejection.
Collapse
Affiliation(s)
- Saima Aslam
- Center for Innovative Phage Applications and Therapeutics, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
14
|
Goldstein DR. My time to say goodbye to JHLT. J Heart Lung Transplant 2023; 42:1319-1320. [PMID: 37410000 DOI: 10.1016/j.healun.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 07/07/2023] Open
Affiliation(s)
- Daniel R Goldstein
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
15
|
Yang RZ, Liang M, Lin S, Weng J, Hu JM, Lin SZ, Wu XD, Zeng K. General anesthesia alters the diversity and composition of the lung microbiota in rat. Biomed Pharmacother 2023; 166:115381. [PMID: 37639744 DOI: 10.1016/j.biopha.2023.115381] [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: 07/04/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND The lung microbiome plays a crucial role in human health and disease. Extensive studies have demonstrated that the disturbance of the lung microbiome influences immune response, cognition, and behavior. The goal of this study was to investigate the effect of general anesthetics on lung microbiome. METHODS Eight-week-old male SD rats received a continuous intravenous infusion of propofol or inhalation of isoflurane for 4 h. 16S rRNA gene amplification from BALF samples was used to investigate the changes in the lung microbiome after interventions. We further performed neurobehavioral assessments to find the differential strains' association with behavior disorder after isoflurane anesthesia. RESULTS The absolute and relative quantitation of 16S rRNA sequencing data showed that isoflurane altered the diversity and abundance of the lung microbiome in rats more than propofol. Elusimicrobia increased significantly in the isoflurane group. Both EPM and OFT results showed that rats exhibited depression-like behaviors after inhalation of isoflurane. In addition, significant differences were found in the COG/KO/MetaCyc/KEGG pathway enrichment analyses among the groups. CONCLUSION Continuous inhalation of isoflurane changed the diversity and composition of the lung microbiota in rats, resulting in post-anesthesia depression.
Collapse
Affiliation(s)
- Rui-Zhi Yang
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Min Liang
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Song Lin
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jing Weng
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jia-Min Hu
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shi-Zhu Lin
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiao-Dan Wu
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou, China.
| | - Kai Zeng
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
16
|
Traunero A, Peri F, Badina L, Amaddeo A, Zuliani E, Maschio M, Barbi E, Ghirardo S. Hematopoietic Stem Cells Transplant (HSCT)-Related Chronic Pulmonary Diseases: An Overview. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1535. [PMID: 37761496 PMCID: PMC10530143 DOI: 10.3390/children10091535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
Recipients of HSCT have a high risk of infective and non-infective pulmonary diseases. Most patients with pulmonary involvement present multiple pathogenetic mechanisms simultaneously with complex interactions. Therefore, it can be difficult to distinguish the contributions of each one and to perform studies on this subject. In this opinion article, we discuss only chronic pulmonary manifestations, focusing on LONIPCs (late-onset non-infectious pulmonary complications). This term embraces drug-related toxicity, allergies, and chronic pulmonary graft versus host disease (GvHD) in all its recently identified clinical variants. Among LONIPCs, GvHD represents the most critical in terms of morbidity and mortality, despite the rapid development of new treatment options. A recently emerging perspective suggests that pulmonary lung rejection in transplant patients shares striking similarities with the pathogenesis of GvHD. In a pulmonary transplant, the donor organ is damaged by the host immune system, whereas in GvHD, the donor immune system damages the host organs. It constitutes the most significant breakthrough in recent years and is highly promising for both hematologists and thoracic transplant surgeons. The number of patients with LONIPCs is scarce, with heterogenous clinical characteristics often involving several pathogenetic mechanisms, making it challenging to conduct randomized controlled trials. Therefore, the body of evidence in this field is scarce and generally of low quality, leading to jeopardized choices in terms of immunosuppressive treatment. Moreover, it risks being outdated by common practice due to the quick evolution of knowledge about the diagnosis and treatment of LONIPCs. The literature is even more pitiful for children with pulmonary involvement related to HSCT.
Collapse
Affiliation(s)
- Arianna Traunero
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34126 Trieste, Italy
| | - Francesca Peri
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34126 Trieste, Italy
| | - Laura Badina
- Department of Pediatrics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Alessandro Amaddeo
- Emergency Department, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Elettra Zuliani
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34126 Trieste, Italy
| | - Massimo Maschio
- Department of Pediatrics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Egidio Barbi
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34126 Trieste, Italy
- Department of Pediatrics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Sergio Ghirardo
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34126 Trieste, Italy
| |
Collapse
|
17
|
Wu J, Li C, Gao P, Zhang C, Zhang P, Zhang L, Dai C, Zhang K, Shi B, Liu M, Zheng J, Pan B, Chen Z, Zhang C, Liao W, Pan W, Fang W, Chen C. Intestinal microbiota links to allograft stability after lung transplantation: a prospective cohort study. Signal Transduct Target Ther 2023; 8:326. [PMID: 37652953 PMCID: PMC10471611 DOI: 10.1038/s41392-023-01515-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 05/17/2023] [Accepted: 05/28/2023] [Indexed: 09/02/2023] Open
Abstract
Whether the alternated microbiota in the gut contribute to the risk of allograft rejection (AR) and pulmonary infection (PI) in the setting of lung transplant recipients (LTRs) remains unexplored. A prospective multicenter cohort of LTRs was identified in the four lung transplant centers. Paired fecal and serum specimens were collected and divided into AR, PI, and event-free (EF) groups according to the diagnosis at sampling. Fecal samples were determined by metagenomic sequencing. And metabolites and cytokines were detected in the paired serum to analyze the potential effect of the altered microbiota community. In total, we analyzed 146 paired samples (AR = 25, PI = 43, and EF = 78). Notably, we found that the gut microbiome of AR followed a major depletion pattern with decreased 487 species and compositional diversity. Further multi-omics analysis showed depleted serum metabolites and increased inflammatory cytokines in AR and PI. Bacteroides uniformis, which declined in AR (2.4% vs 0.6%) and was negatively associated with serum IL-1β and IL-12, was identified as a driven specie in the network of gut microbiome of EF. Functionally, the EF specimens were abundant in probiotics related to mannose and cationic antimicrobial peptide metabolism. Furthermore, a support-vector machine classifier based on microbiome, metabolome, and clinical parameters highly predicted AR (AUPRC = 0.801) and PI (AUPRC = 0.855), whereby the microbiome dataset showed a particularly high diagnostic power. In conclusion, a disruptive gut microbiota showed a significant association with allograft rejection and infection and with systemic cytokines and metabolites in LTRs.
Collapse
Affiliation(s)
- Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chongwu Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Peigen Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Pei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Kunpeng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Bowen Shi
- Department of Thoracic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mengyang Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bo Pan
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Chen
- Adfontes (Shanghai) Bio-technology Co., Ltd, Shanghai, China
| | - Chao Zhang
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wanqing Liao
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weihua Pan
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Wenjie Fang
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| |
Collapse
|
18
|
Dallal-York J, Croft K, Anderson A, DiBiase L, Donohue C, Vasilopoulos T, Shahmohammadi A, Pelaez A, Pipkin M, Hegland KW, Machuca TN, Plowman EK. A prospective examination of swallow and cough dysfunction after lung transplantation. Neurogastroenterol Motil 2023; 35:e14458. [PMID: 36168190 DOI: 10.1111/nmo.14458] [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: 05/27/2022] [Revised: 08/08/2022] [Accepted: 08/23/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Swallow and cough dysfunction are possible surgical complications of lung transplantation (LT). We examined voluntary cough strength, sensorimotor reflexive cough integrity, and swallow-related respiratory rate (RR) across swallowing safety and aspiration response groups in recovering LT recipients. METHODS Forty-five LT recipients underwent flexible endoscopic evaluation of swallowing indexed by the validated Penetration Aspiration Scale. RR before and after a 3-ounce water drinking task was measured. Voluntary and reflexive cough screening were performed to index motor and sensory outcomes. T-tests, one-way ANOVAs, and chi-square (odds ratios) were used. RESULTS 60% of patients exhibited laryngeal penetration (n = 27) and 40% demonstrated tracheal aspiration (n = 18); 72% (n = 13) demonstrated silent aspiration. Baseline RR was higher in aspirators versus non-aspirators (26.5 vs. 22.6, p = 0.04) and in silent aspirators compared to non-silent aspirators (27.9 vs. 20.7, p = 0.01). RR change post-swallowing did not differ between aspiration response groups; however, it was significantly higher in aspirators compared to non-aspirators (3 vs. -2, p = 0.02). Compared to non-silent aspirators, silent aspirators demonstrated reduced voluntary cough peak expiratory flow (PEF; 166 vs. 324 L/min, p = 0.01). PEF, motor and urge to cough reflex cough ratings did not differ between aspirators and non-aspirators. Silent aspirators demonstrated a 7.5 times higher odds of failing reflex cough screening compared to non-silent aspirators (p = 0.07). CONCLUSIONS During the acute recovery period, all LT participants demonstrated some degree of unsafe swallowing and reduced voluntary cough strength. Silent aspirators exhibited elevated RR, reduced voluntary cough physiologic capacity to defend the airway, and a clinically distinguishable blunted motor response to reflex cough screening.
Collapse
Affiliation(s)
- Justine Dallal-York
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
| | - Kayla Croft
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
| | - Amber Anderson
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
| | - Lauren DiBiase
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
| | - Cara Donohue
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
- Department of Surgery, Division of Cardiothoracic Surgery, University of Florida, Gainesville, Florida, USA
| | - Terrie Vasilopoulos
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Departments of Anesthesiology and Orthopedics and Rehabilitation, University of Florida, Gainesville, Florida, USA
| | | | - Andres Pelaez
- Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Mauricio Pipkin
- Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Karen W Hegland
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA
| | - Tiago N Machuca
- Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Emily K Plowman
- Aerodigestive Research Core, University of Florida, Gainesville, Florida, USA
- Department of Speech, Language and Hearing Sciences, University of Florida, Gainesville, Florida, USA
- Department of Surgery, Division of Cardiothoracic Surgery, University of Florida, Gainesville, Florida, USA
- Departments of Anesthesiology and Orthopedics and Rehabilitation, University of Florida, Gainesville, Florida, USA
- Department of Neurology, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
Farfour E, Zrounba M, Roux A, Revillet H, Vallée A, Vasse M. Inquilinus limosus Bacteremia in Lung Transplant Recipient after SARS-CoV-2 Infection. Emerg Infect Dis 2023; 29:642-644. [PMID: 36823767 PMCID: PMC9973706 DOI: 10.3201/eid2903.221564] [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: 02/25/2023] Open
Abstract
Inquilinus limosus is an environmental bacterium associated with respiratory tract colonization in cystic fibrosis patients. We report a case of I. limosus bacteremia in a patient in France who received a lung transplant and experienced chronic graft dysfunction and SARS-CoV-2 infection. This case suggests I. limosus displays virulence factors associated with invasion.
Collapse
|
20
|
The Lung Microbiome: A New Frontier for Lung and Brain Disease. Int J Mol Sci 2023; 24:ijms24032170. [PMID: 36768494 PMCID: PMC9916971 DOI: 10.3390/ijms24032170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Due to the limitations of culture techniques, the lung in a healthy state is traditionally considered to be a sterile organ. With the development of non-culture-dependent techniques, the presence of low-biomass microbiomes in the lungs has been identified. The species of the lung microbiome are similar to those of the oral microbiome, suggesting that the microbiome is derived passively within the lungs from the oral cavity via micro-aspiration. Elimination, immigration, and relative growth within its communities all contribute to the composition of the lung microbiome. The lung microbiome is reportedly altered in many lung diseases that have not traditionally been considered infectious or microbial, and potential pathways of microbe-host crosstalk are emerging. Recent studies have shown that the lung microbiome also plays an important role in brain autoimmunity. There is a close relationship between the lungs and the brain, which can be called the lung-brain axis. However, the problem now is that it is not well understood how the lung microbiota plays a role in the disease-specifically, whether there is a causal connection between disease and the lung microbiome. The lung microbiome includes bacteria, archaea, fungi, protozoa, and viruses. However, fungi and viruses have not been fully studied compared to bacteria in the lungs. In this review, we mainly discuss the role of the lung microbiome in chronic lung diseases and, in particular, we summarize the recent progress of the lung microbiome in multiple sclerosis, as well as the lung-brain axis.
Collapse
|
21
|
Glanville AR, Mitchell AB. New Tools for Old Problems: Gastroesophageal Reflux Disease and the Lung Allograft Microbiome. Am J Respir Crit Care Med 2022; 206:1444-1445. [PMID: 35925015 PMCID: PMC9757095 DOI: 10.1164/rccm.202207-1446ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Allan R. Glanville
- The Lung Transplant UnitSt. Vincent’s HospitalSydney, New South Wales, Australia
| | | |
Collapse
|
22
|
Guohui J, Kun W, Dong T, Ji Z, Dong L, Dong W, Jingyu C. Microbiosis in lung allotransplantation and xenotransplantation: State of the art and future perspective. HEALTH CARE SCIENCE 2022; 1:119-128. [PMID: 38938886 PMCID: PMC11080722 DOI: 10.1002/hcs2.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/10/2022] [Accepted: 08/03/2022] [Indexed: 06/29/2024]
Abstract
The respiratory tract is known to harbor a microbial community including bacteria, viruses, and fungi. New techniques contribute enormously to the identification of unknown or culture-independent species and reveal the interaction of the community with the host immune system. The existing respiratory microbiome and substantial equilibrium of the transplanted microbiome from donor lung grafts provide an extreme bloom of dynamic changes in the microenvironment in lung transplantation (LT) recipients. Dysbiosis in grafts are not only related to the modified microbial components but also involve the kinetics of the host-graft "talk," which signifies the destination of graft allograft injury, acute rejection, infection, and chronic allograft dysfunction development in short- and long-term survival. Microbiome-derived factors may contribute to lung xenograft survival when using genetically multimodified pig-derived organs. Here, we review the most advanced knowledge of the dynamics and resilience of microbial communities in transplanted lungs with various pretransplant indications. Conceptual and analytical points of view have been illustrated along the time series, gaining insight into the microbiome and lung grafts. Future endeavors on precise tools, sophisticated models, and novel targeted regimens are needed to improve the long-term survival in these patients.
Collapse
Affiliation(s)
- Jiao Guohui
- Center for Medical Device Evaluation, NMPABeijingChina
| | - Wu Kun
- Center for Medical Device Evaluation, NMPABeijingChina
| | - Tian Dong
- Department of Thoracic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Zhang Ji
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiChina
| | - Liu Dong
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiChina
| | - Wei Dong
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiChina
| | - Chen Jingyu
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiChina
| |
Collapse
|
23
|
Sen T, Thummer RP. The Impact of Human Microbiotas in Hematopoietic Stem Cell and Organ Transplantation. Front Immunol 2022; 13:932228. [PMID: 35874759 PMCID: PMC9300833 DOI: 10.3389/fimmu.2022.932228] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
The human microbiota heavily influences most vital aspects of human physiology including organ transplantation outcomes and transplant rejection risk. A variety of organ transplantation scenarios such as lung and heart transplantation as well as hematopoietic stem cell transplantation is heavily influenced by the human microbiotas. The human microbiota refers to a rich, diverse, and complex ecosystem of bacteria, fungi, archaea, helminths, protozoans, parasites, and viruses. Research accumulating over the past decade has established the existence of complex cross-species, cross-kingdom interactions between the residents of the various human microbiotas and the human body. Since the gut microbiota is the densest, most popular, and most studied human microbiota, the impact of other human microbiotas such as the oral, lung, urinary, and genital microbiotas is often overshadowed. However, these microbiotas also provide critical and unique insights pertaining to transplantation success, rejection risk, and overall host health, across multiple different transplantation scenarios. Organ transplantation as well as the pre-, peri-, and post-transplant pharmacological regimens patients undergo is known to adversely impact the microbiotas, thereby increasing the risk of adverse patient outcomes. Over the past decade, holistic approaches to post-transplant patient care such as the administration of clinical and dietary interventions aiming at restoring deranged microbiota community structures have been gaining momentum. Examples of these include prebiotic and probiotic administration, fecal microbial transplantation, and bacteriophage-mediated multidrug-resistant bacterial decolonization. This review will discuss these perspectives and explore the role of different human microbiotas in the context of various transplantation scenarios.
Collapse
Affiliation(s)
| | - Rajkumar P. Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
24
|
Abstract
The healthy lung was long thought of as sterile, but recent advances using molecular sequencing approaches have detected bacteria at low levels. Healthy lung bacteria largely reflect communities present in the upper respiratory tract that enter the lung via microaspiration, which is balanced by mechanical and immune clearance and likely involves limited local replication. The nature and dynamics of the lung microbiome, therefore, differ from those of ecological niches with robust self-sustaining microbial communities. Aberrant populations (dysbiosis) have been demonstrated in many pulmonary diseases not traditionally considered microbial in origin, and potential pathways of microbe-host crosstalk are emerging. The question now is whether and how dysbiotic microbiota contribute to initiation or perpetuation of injury. The fungal microbiome and virome are less well studied. This Review highlights features of the lung microbiome, unique considerations in studying it, examples of dysbiosis in selected disease, emerging concepts in lung microbiome-host interactions, and critical areas for investigation.
Collapse
|