1
|
Chen Y, Chen Z, Liang L, Li J, Meng L, Yuan W, Xie B, Zhang X, Feng L, Jia Y, Fu Z, Su P, Tong Z, Zhong J, Liu X. Multi-kingdom gut microbiota dysbiosis is associated with the development of pulmonary arterial hypertension. EBioMedicine 2025; 115:105686. [PMID: 40220715 PMCID: PMC12013117 DOI: 10.1016/j.ebiom.2025.105686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Gut microbiota dysbiosis has been implicated in pulmonary arterial hypertension (PAH). However, the exact roles and underlying mechanisms of multi-kingdom gut microbiota, including bacteria, archaea, and fungi, in PAH remain largely unclear. METHODS The shotgun metagenomics was used to analyse multi-kingdom gut microbial communities in patients with idiopathic PAH (IPAH) and healthy controls. Furthermore, fecal microbiota transplantation (FMT) was performed to transfer gut microbiota from IPAH patients or monocrotaline (MCT)-PAH rats to normal rats and from normal rats to MCT-PAH rats. FINDINGS Gut microbiota analysis revealed substantial alterations in the bacterial, archaeal, and fungal communities in patients with IPAH compared with healthy controls. Notably, FMT from IPAH patients or MCT-PAH rats induced PAH phenotypes in recipient rats. More intriguingly, FMT from normal rats to MCT-PAH rats significantly ameliorated PAH symptoms; restored gut bacteria, archaea, and fungi composition; and shifted the plasma metabolite profiles of MCT-PAH rats toward those of normal rats. In parallel, RNA-sequencing analysis demonstrated the expression of genes involved in key signalling pathways related to PAH. A panel of multi-kingdom markers exhibited superior diagnostic accuracy compared with single-kingdom panels for IPAH. INTERPRETATION Our findings established an association between multi-kingdom gut microbiota dysbiosis and PAH, thereby indicating the therapeutic potential of FMT in PAH. More importantly, apart from gut bacteria, gut archaea and fungi were also significantly associated with PAH pathogenesis, highlighting their indispensable role in PAH. FUNDING This work was supported by Noncommunicable Chronic Diseases-National Science and Technology Major Projects No. 2024ZD0531200, No. 2024ZD0531201 (Research on Prevention and Treatment of Cancer, Cardiovascular and Cerebrovascular Diseases, Respiratory Diseases, and Metabolic Diseases), the National Natural Science Foundation of China of China (No. 82170302, 82370432), Financial Budgeting Project of Beijing Institute of Respiratory Medicine (Ysbz2025004, Ysbz2025007), National clinical key speciality construction project Cardiovascular Surgery, Reform and Development Program of Beijing Institute of Respiratory Medicine (Ggyfz202417, Ggyfz202501), Clinical Research Incubation Program of Beijing Chaoyang Hospital Affiliated to Capital Medical University (CYFH202209).
Collapse
Affiliation(s)
- Yihang Chen
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhenzhen Chen
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lirong Liang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 100020, Beijing, China
| | - Jifeng Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Liukun Meng
- Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Wen Yuan
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Boqia Xie
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xun Zhang
- Department of Pharmacy, Chinese PLA General Hospital, 100039, Beijing, China
| | - Lin Feng
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 100020, Beijing, China
| | - Yanxiong Jia
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhou Fu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Pixiong Su
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Xiaoyan Liu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
2
|
Su L, Wang X, Lin Y, Zhang Y, Yao D, Pan T, Huang X. Exploring the Causal Relationship Between Gut Microbiota and Pulmonary Artery Hypertension: Insights From Mendelian Randomization. J Am Heart Assoc 2025; 14:e038150. [PMID: 40079338 DOI: 10.1161/jaha.124.038150] [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: 08/06/2024] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Research into the "gut-lung" axis links gut microbiota to pulmonary artery hypertension (PAH). However, the mechanisms by which gut microbiota influence PAH remain unclear. We aimed to investigate the causal relationship between the gut microbiota and PAH using Mendelian randomization analysis, identify key microbiota and metabolites, and explore the regulatory role of associated genes in PAH pathogenesis. METHODS AND RESULTS We examined the association between gut microbiota taxa and PAH using inverse variance weighted 2-sample Mendelian randomization analysis, Mendelian randomization-Egger, weighted median, and weighted mode methods. Additionally, we identified PAH-regulating genes in the intestinal microbiome using bioinformatics tools and validated their expression levels in the lung tissue of hypoxia-induced PAH mice models by quantitative reverse transcription polymerase chain reaction. Eleven gut microbiota taxa were associated with PAH. The order Clostridiales and genera Eubacterium fissicatena group, Lachnospiraceae UCG004, and Ruminococcaceae UCG002 were positively associated with PAH, whereas the order Bifidobacteriales; family Bifidobacteriaceae; and genera Eubacterium eligens group, Sutterella, Methanobrevibacter, Sellimonas, and Tyzzerella3 were negatively associated with PAH, with some exhibiting bidirectional causality. These microbiota modulate 24 metabolites, including palmitoylcholine, oleoylcholine, and 3,7-dimethylurate, to influence PAH. Hypoxia-induced PAH mice had significantly downregulated 1,4,5-trisphosphate receptor type 2, degrading enzyme, nuclear receptor-interacting protein 1, and growth factor-binding protein 1 in lung tissues, indicating their potential role in PAH regulation. CONCLUSIONS These findings suggest that gut microbiota composition and associated metabolites contribute to PAH development by regulating lung tissue gene expression. Our findings have implications for advancing gut microbiota-based PAH diagnostic technologies and targeted therapies.
Collapse
Affiliation(s)
- Lihuang Su
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Xinghong Wang
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Ya Lin
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Yiying Zhang
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Dan Yao
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Tongtong Pan
- Department of Gerontology The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| |
Collapse
|
3
|
Zhang C, Zhong B, Jiang Q, Lu W, Wu H, Xing Y, Wu X, Zhang Z, Zheng Y, Li P, Li Z, Lin Z, Chen Y, Hong C, Zhao Z, Zhang T, Liang W, Zhang Y, Zhang C, Yuan JXJ, Liu C, Wang J, Yang K. Distinct airway mycobiome signature in patients with pulmonary hypertension and subgroups. BMC Med 2025; 23:148. [PMID: 40059138 PMCID: PMC11892250 DOI: 10.1186/s12916-025-03982-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 03/03/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND The association between lung microbiome and pulmonary hypertension (PH) remain unknown. This study aims to define the airway mycobiome signature and its potential correlation with clinical parameters of PH. METHODS Overall, 244 patients with PH and 120 healthy controls (CON) were recruited from three independent centers. The PH group was divided into subgroups not using antibiotics or corticosteroids (non-ANT/CORT), and those using ANT, CORT, or ANT + CORT within 1 month, and clinical classification (Groups 1, 3, and 4), World Health Organization functional class (I-IV), and disease severity based on mean pulmonary artery pressure or pulmonary vascular resistance levels for in-depth comparison. RESULTS Distinct airway mycobiome profiles were observed in PH, CON, and PH subgroups. Linear discriminant analysis effect size analysis showed increased Purpureocillium, Issatchenkia, and Cyberlindnera and decreased Peroneutypa, Simplicillium, and Metarhizium in patients with PH (non-ANT/CORT, ANT, CORT, and ANT + CORT) than in CON. Receiver operating characteristic analysis indicated a strong prediction of the two fungal genera sets in distinguishing PH and its subgroups from CON. The two major fungal phyla, Ascomycota and Basidiomycota, correlated differently with major clinical factors. Increased connections among the top fungal phyla or genera were observed in the PH than in the CON group. Dominant enrichment (Purpureocillium, Issatchenkia, and Cyberlindnera) and diminishment (Peroneutypa, Simplicillium, and Metarhizium) of fungal genera consistently and strongly predicted PH without being influenced by different PH subgroups. CONCLUSIONS This study provides the first description of the unique airway mycobiome signature in PH and among different PH subgroups.
Collapse
Affiliation(s)
- Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Bihua Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University), Guangdong, Foshan, China
| | - Qian Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Hongkai Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Yue Xing
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Xuefen Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Zizhou Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Yulin Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Peiwen Li
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University), Guangdong, Foshan, China
| | - Zhenxiang Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziying Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Cheng Hong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China
| | - Zhuxiang Zhao
- Department of Pulmonary and Critical Care Medicine, Infectious Diseases, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangdong, Guangzhou, China
| | - Tingting Zhang
- Department of Intensive Care Unit, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Weiquan Liang
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University), Guangdong, Foshan, China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Caojin Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Chunli Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China.
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital, Guangzhou Medical University, 195 West Dong Feng Road, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
4
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
5
|
Zhang Z, Liang Y, Mo S, Zhao M, Li Y, Zhang C, Shan X, Liu S, Liao J, Luo X, Zhu J, Wang C, Jiang Q, Hou C, Hong W, Lai N, Chen Y, Xu L, Lu W, Wang J, Wang Z, Yang K. Oral administration of pioglitazone inhibits pulmonary hypertension by regulating the gut microbiome and plasma metabolome in male rats. Physiol Rep 2025; 13:e70174. [PMID: 39739369 DOI: 10.14814/phy2.70174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
The oral administrated thiazolidinediones (TZDs) have been widely reported to alleviate experimental pulmonary hypertension (PH). However, previous studies mainly focused on their beneficial effects on the cardiopulmonary vascular system but failed to determine their potential roles on gut microenvironment. This study aims to investigate the effects of pioglitazone, an oral TZD drug, on gut microbiome in classic PH rat models induced by hypoxia (HPH) or SU5416/hypoxia (SuHx-PH) and evaluate the therapeutic potential of supplementation of selective probiotics for experimental PH. Pioglitazone remarkably inhibited the PH pathogenesis in both models and reshaped the gut microbiome and plasma metabolome. Correlation analyses represented strong and unique association between the protective metabolites and bacteria genera (Roseburia, Lactobacillus, and Streptococcus) that were positively stimulated by pioglitazone. Supplementation of selective probiotics Roseburia intestinalis (R. intestinalis) partially attenuated SuHx-PH and rebuilt a novel gut microbiome and host metabolome. This study reports for the first time that oral administration of pioglitazone protects PH by regulating the gut microbiome and host metabolome, providing novel insights for the TZD drugs. The data also supports that modulation of gut microbiota by supplementation of selective probiotics could be a novel effective therapeutic strategy for the treatment of PH.
Collapse
Affiliation(s)
- Zizhou Zhang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yaru Liang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Shaocong Mo
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingming Zhao
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yi Li
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Diseases of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Liao
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiaoyun Luo
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Junqi Zhu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chen Wang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qian Jiang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chi Hou
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ning Lai
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Zhongfang Wang
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Kai Yang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
- State Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Thenappan T, Weir EK. Gut Microbiome and Pulmonary Arterial Hypertension - A Novel and Evolving Paradigm. Physiol Res 2024; 73:S477-S485. [PMID: 39589297 PMCID: PMC11627261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 11/27/2024] Open
Abstract
Pulmonary arterial hypertension is characterized by perivascular and systemic inflammation. The gut microbiome influences the host immune system. Here we review the emerging preclinical and clinical evidence that strongly suggests that alterations in the gut microbiome may either initiate or facilitate progression of established pulmonary arterial hypertension by modifying the systemic immune responses. We also briefly review the relationship between the gut microbiome and preeclampsia, a vascular disease also characterized by inflammation. Key words: Dysbiosis, Right ventricle, Inflammation.
Collapse
Affiliation(s)
- T Thenappan
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.
| | | |
Collapse
|
7
|
Alswat AS. The Influence of the Gut Microbiota on Host Health: A Focus on the Gut-Lung Axis and Therapeutic Approaches. Life (Basel) 2024; 14:1279. [PMID: 39459579 PMCID: PMC11509314 DOI: 10.3390/life14101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
The human gut microbiota is a complex ecosystem harboring thousands of microbial strains that play a crucial role in maintaining the overall well-being of its host. The composition of the gut microbiota varies with age from infancy to adulthood and is influenced by dietary habits, environment, and genetic disposition. Recent advances in culture-independent techniques and nucleic acid sequencing have improved our understanding of the diversity of the gut microbiota. The microbial species present in the gut release short-chain fatty acids (SCFAs), which have anti-inflammatory properties. The gut microbiota also plays a substantial role in modulating the host's immune system, promoting immune tolerance, and maintaining homeostasis. The impact of the gut microbiota on the health of the host is quite evident, as gut dysbiosis has been linked to various diseases, including metabolic disorders, autoimmune diseases, allergies, and inflammatory bowel diseases. The gut microbiota has bidirectional communication with the respiratory system, creating the gut-lung axis, which has been associated with different respiratory diseases. Therapeutic approaches targeting the gut microbiota, such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation (FMT), aim to restore microbial balance and promote the growth of beneficial strains in the gut. Nonetheless, gaining knowledge of the complex interactions between the gut microbiota and the host is necessary to develop personalized medicine approaches and microbiota-based therapies for various conditions. This review summarizes studies related to the gut-lung axis with particular emphasis on the role of the microbiota. Future research directions are also discussed.
Collapse
Affiliation(s)
- Amal S Alswat
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
8
|
Abudukeremu A, Aikemu A, Yang T, Fang L, Aihemaitituoheti A, Zhang Y, Shanahaiti D, Nijiati Y. Effects of the ACE2-Ang-(1-7)-Mas axis on gut flora diversity and intestinal metabolites in SuHx mice. Front Microbiol 2024; 15:1412502. [PMID: 39247700 PMCID: PMC11380154 DOI: 10.3389/fmicb.2024.1412502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024] Open
Abstract
Objective Pulmonary artery hypertension (PAH) poses a significant challenge due to its limited therapeutic options and high mortality rates. The ACE2-Ang-(1-7)-Mas axis plays a pivotal role in regulating blood pressure and inhibiting myocardial remodeling. However, the precise mechanistic links between the ACE2-Ang-(1-7)-Mas axis and PAH remain poorly understood. This study aimed to elucidate the involvement of the ACE2-Ang-(1-7)-Mas axis in the development of PAH. Methods PAH was induced in mice using Sugen5416/hypoxia, PAAT/PET ratio and PA were detected using cardiac ultrasound; inflammation related factors such as MCP-1, TNF, IL-10and IL-12p70 were detected in intestines using cytometric bead array (CBA) kits; histopathological and morphological changes in lung and intestinal tissues were assessed via HE staining and Masson staining to evaluate the progression of PAH. Immunohistochemistry and western blotting were employed to determine the expression levels of two tight junction proteins, occludin and ZO-1, in intestinal tissues. Additionally, 16rRNA sequencing and non-targeted metabolomics by LC-MS/MS techniques were utilized to investigate the impact of the ACE2-Ang-(1-7)-Mas axis on microbial diversity and metabolomics of intestinal contents. Results Activation of the ACE2-Ang-(1-7)-Mas axis improves heart function, reduces intestines inflammatory factors and ameliorates pathological and histological alterations in SuHx mice. This activation notably upregulated the expression of occludin and ZO-1 proteins in intestinal tissues and promoted the proliferation of SCFA-producing bacteria genera, such as g_Candidatus_Saccharimonas. Furthermore, it enhanced the abundance of beneficial metabolites, including tryptophan and butyric acid. Conclusion The findings suggest that modulation of the ACE2-Ang-(1-7)-Mas axis can alleviate PAH by regulating intestinal microbes and metabolites. These results highlight the potential of the ACE2-Ang-(1-7)-Mas axis as a promising therapeutic target for clinical management of PAH.
Collapse
Affiliation(s)
- Asimuguli Abudukeremu
- Central Laboratory of Xinjiang Medical University, Urumqi, China
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Ainiwaer Aikemu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
- Department of Pharmacy, College of Xinjiang Uyghur Medicine, Hetian, China
- Xinjiang Key Laboratory of Hetian Characteristic Chinese Traditional Medicine Research, Hetian, China
- Engineering Research Center for Quality Control of Uyghur Medicinal Materials and Preparations, Hetian, China
| | - Tao Yang
- Central Laboratory of Xinjiang Medical University, Urumqi, China
| | - Lei Fang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | | | - Yupeng Zhang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | | | - Yiliyaer Nijiati
- Central Laboratory of Xinjiang Medical University, Urumqi, China
- Key Laboratory of High Incidence Disease Research in Xinjiang (Xinjiang Medical University), Ministry of Education, Urumqi, China
| |
Collapse
|
9
|
Cao W, Wang L, Mo Q, Peng F, Hong W, Zhou Y, Sun R, Li H, Liang C, Zhao D, Zheng M, Li B, Peng G. Disease-associated gut microbiome and metabolome changes in rats with chronic hypoxia-induced pulmonary hypertension. Front Cell Dev Biol 2024; 12:1022181. [PMID: 39071798 PMCID: PMC11272533 DOI: 10.3389/fcell.2024.1022181] [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: 08/18/2022] [Accepted: 06/19/2024] [Indexed: 07/30/2024] Open
Abstract
Background Pulmonary hypertension (PH) is a progressive disease affecting the lung vasculature that is characterized by sustained vasoconstriction and leads to vascular remodeling. The lung microbiome contributes to PH progression, but the function of the gut microbiome and the correlation between the gut microbiome and metabolome remain unclear. We have analyzed whether chronic hypoxia-induced PH alters the rat fecal microbiota. Purpose We explored hypoxia-induced pulmonary hypertension model rats to find out the characteristic changes of intestinal microorganisms and metabolites of hypoxia-induced pulmonary hypertension, and provide a theoretical basis for clinical treatment. Methods In the current study, a chronic hypoxia-induced PH rat model was used to investigate the role of the gut microbiome and metabolome as a potential mechanism contributing to the occurrence and development of PH. 16S ribosomal ribonucleic acid (16S rRNA), short-chain fatty acid (SCFA) measurements, mass spectrometry (MS) metabolomics analysis and metatranscriptome were performed to analyze stool samples. The datasets were analyzed individually and integrated for combined analysis using bioinformatics approaches. Results Our results suggest that the gut microbiome and metabolome of chronic hypoxia-induced PH rats are distinct from those of normoxic rats and may thus aid in the search for new therapeutic or diagnostic paradigms for PH. Conclusion The gut microbiome and metabolome are altered as a result of chronic hypoxia-induced PH. This imbalanced bacterial ecosystem might play a pathophysiological role in PH by altering homeostasis.
Collapse
Affiliation(s)
- Weitao Cao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Luyao Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiudi Mo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Respiratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fang Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Hong
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yumin Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruiting Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiqing Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunxiao Liang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Thoracic Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Guangzhou, China
| | - Dongxing Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengning Zheng
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Bing Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gongyong Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Yang Y, Zhang H, Wang Y, Xu J, Shu S, Wang P, Ding S, Huang Y, Zheng L, Yang Y, Xiong C. Promising dawn in the management of pulmonary hypertension: The mystery veil of gut microbiota. IMETA 2024; 3:e159. [PMID: 38882495 PMCID: PMC11170974 DOI: 10.1002/imt2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 11/25/2023] [Indexed: 06/18/2024]
Abstract
The gut microbiota is a complex community of microorganisms inhabiting the intestinal tract, which plays a vital role in human health. It is intricately involved in the metabolism, and it also affects diverse physiological processes. The gut-lung axis is a bidirectional pathway between the gastrointestinal tract and the lungs. Recent research has shown that the gut microbiome plays a crucial role in immune response regulation in the lungs and the development of lung diseases. In this review, we present the interrelated factors concerning gut microbiota and the associated metabolites in pulmonary hypertension (PH), a lethal disease characterized by elevated pulmonary vascular pressure and resistance. Our research team explored the role of gut-microbiota-derived metabolites in cardiovascular diseases and established the correlation between metabolites such as putrescine, succinate, trimethylamine N-oxide (TMAO), and N, N, N-trimethyl-5-aminovaleric acid with the diseases. Furthermore, we found that specific metabolites, such as TMAO and betaine, have significant clinical value in PH, suggesting their potential as biomarkers in disease management. In detailing the interplay between the gut microbiota, their metabolites, and PH, we underscored the potential therapeutic approaches modulating this microbiota. Ultimately, we endeavor to alleviate the substantial socioeconomic burden associated with this disease. This review presents a unique exploratory analysis of the link between gut microbiota and PH, intending to propel further investigations in the gut-lung axis.
Collapse
Affiliation(s)
- Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Hanwen Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yaoyao Wang
- State Key Laboratory of Cardiovascular Disease, Department of Nephrology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Department of Genetics University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Songren Shu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Peizhi Wang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Center for Molecular Cardiology University of Zurich Zurich Switzerland
| | - Shusi Ding
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
| | - Yuan Huang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
- Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, School of Basic Medical Sciences, Health Science Center The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, Peking University Beijing China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Changming Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
11
|
Meng X, Du L, Xu S, Zhou L, Chen B, Li Y, Chen C, Ye H, Zhang J, Tian G, Bai X, Dong T, Lin W, Sun M, Zhou K, Liu Y, Zhang W, Duan S. Periodontitis exacerbates pulmonary hypertension by promoting IFNγ + T cell infiltration in mice. Int J Oral Sci 2024; 16:27. [PMID: 38548721 PMCID: PMC10978940 DOI: 10.1038/s41368-024-00291-2] [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: 12/06/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 04/01/2024] Open
Abstract
Uncovering the risk factors of pulmonary hypertension and its mechanisms is crucial for the prevention and treatment of the disease. In the current study, we showed that experimental periodontitis, which was established by ligation of molars followed by orally smearing subgingival plaques from patients with periodontitis, exacerbated hypoxia-induced pulmonary hypertension in mice. Mechanistically, periodontitis dysregulated the pulmonary microbiota by promoting ectopic colonization and enrichment of oral bacteria in the lungs, contributing to pulmonary infiltration of interferon gamma positive (IFNγ+) T cells and aggravating the progression of pulmonary hypertension. In addition, we identified Prevotella zoogleoformans as the critical periodontitis-associated bacterium driving the exacerbation of pulmonary hypertension by periodontitis, and the exacerbation was potently ameliorated by both cervical lymph node excision and IFNγ neutralizing antibodies. Our study suggests a proof of concept that the combined prevention and treatment of periodontitis and pulmonary hypertension are necessary.
Collapse
Affiliation(s)
- Xiaoqian Meng
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Linjuan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shuo Xu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lujun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Boyan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yulin Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Chumao Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huilin Ye
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jun Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Guocai Tian
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xuebing Bai
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ting Dong
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenzhen Lin
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Mengjun Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kecong Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wuchang Zhang
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China.
| | - Shengzhong Duan
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China.
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
12
|
Singh N, Al-Naamani N, Brown MB, Long GM, Thenappan T, Umar S, Ventetuolo CE, Lahm T. Extrapulmonary manifestations of pulmonary arterial hypertension. Expert Rev Respir Med 2024; 18:189-205. [PMID: 38801029 PMCID: PMC11713041 DOI: 10.1080/17476348.2024.2361037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Extrapulmonary manifestations of pulmonary arterial hypertension (PAH) may play a critical pathobiological role and a deeper understanding will advance insight into mechanisms and novel therapeutic targets. This manuscript reviews our understanding of extrapulmonary manifestations of PAH. AREAS COVERED A group of experts was assembled and a complimentary PubMed search performed (October 2023 - March 2024). Inflammation is observed throughout the central nervous system and attempts at manipulation are an encouraging step toward novel therapeutics. Retinal vascular imaging holds promise as a noninvasive method of detecting early disease and monitoring treatment responses. PAH patients have gut flora alterations and dysbiosis likely plays a role in systemic inflammation. Despite inconsistent observations, the roles of obesity, insulin resistance and dysregulated metabolism may be illuminated by deep phenotyping of body composition. Skeletal muscle dysfunction is perpetuated by metabolic dysfunction, inflammation, and hypoperfusion, but exercise training shows benefit. Renal, hepatic, and bone marrow abnormalities are observed in PAH and may represent both end-organ damage and disease modifiers. EXPERT OPINION Insights into systemic manifestations of PAH will illuminate disease mechanisms and novel therapeutic targets. Additional study is needed to understand whether extrapulmonary manifestations are a cause or effect of PAH and how manipulation may affect outcomes.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
| | - Nadine Al-Naamani
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington School of Medicine, Seattle, WA
| | - Gary Marshall Long
- Department of Kinesiology, Health and Sport Sciences, University of Indianapolis, Indianapolis, IN
| | - Thenappan Thenappan
- Section of Advanced Heart Failure and Pulmonary Hypertension, Cardiovascular Division, University of Minnesota, Minneapolis, MN
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Corey E. Ventetuolo
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
- Department of Health Services, Policy and Practice, Brown University, Providence, RI
| | - Tim Lahm
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Medicine, University of Colorado, Aurora, CO
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| |
Collapse
|
13
|
Mason T, Mukherjee B, Marino P. Pulmonary Hypertension and the Gut Microbiome. Biomedicines 2024; 12:169. [PMID: 38255274 PMCID: PMC10813515 DOI: 10.3390/biomedicines12010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
The gut microbiome and its associated metabolites are integral to the maintenance of gut integrity and function. There is increasing evidence that its alteration, referred to as dysbiosis, is involved in the development of a systemic conditions such as cardiovascular disease (e.g., systemic hypertension, atherosclerosis). Pulmonary hypertension (PH) is a condition characterised by progressive remodelling and vasoconstriction of the pulmonary circulation, ultimately leading to right ventricular failure and premature mortality if untreated. Initial studies have suggested a possible association between dysbiosis of the microbiome and the development of PH. The aim of this article is to review the current experimental and clinical data with respect to the potential interaction between the gut microbiome and the pathophysiology of pulmonary hypertension. It will also highlight possible new therapeutic targets that may provide future therapies.
Collapse
Affiliation(s)
- Thomas Mason
- Lane Fox Respiratory Service, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London SE1 7EH, UK
| | - Bhashkar Mukherjee
- Lane Fox Respiratory Service, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London SE1 7EH, UK
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London SW3 6NP, UK
| | - Philip Marino
- Lane Fox Respiratory Service, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London SE1 7EH, UK
| |
Collapse
|
14
|
Suswał K, Tomaszewski M, Romaniuk A, Świechowska-Starek P, Zygmunt W, Styczeń A, Romaniuk-Suswał M. Gut-Lung Axis in Focus: Deciphering the Impact of Gut Microbiota on Pulmonary Arterial Hypertension. J Pers Med 2023; 14:8. [PMID: 38276223 PMCID: PMC10817474 DOI: 10.3390/jpm14010008] [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: 11/09/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Recent advancements in the understanding of pulmonary arterial hypertension (PAH) have highlighted the significant role of the gut microbiota (GM) in its pathogenesis. This comprehensive review delves into the intricate relationship between the GM and PAH, emphasizing the influence of gut microbial composition and the critical metabolites produced. We particularly focus on the dynamic interaction between the gut and lung, examining how microbial dysbiosis contributes to PAH development through inflammation, altered immune responses, and changes in the gut-lung axis. Noteworthy findings include variations in the ratios of key bacterial groups such as Firmicutes and Bacteroidetes in PAH and the pivotal roles of metabolites like trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and serotonin in the disease's progression. Additionally, the review elucidates potential diagnostic biomarkers and novel therapeutic approaches, including the use of probiotics and fecal microbiota transplantation, which leverage the gut microbiota for managing PAH. This review encapsulates the current state of research in this field, offering insights into the potential of gut microbiota modulation as a promising strategy in PAH diagnosing and treatment.
Collapse
Affiliation(s)
- Konrad Suswał
- Department of Pulmonology, Alergollogy and Oncology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Michał Tomaszewski
- Department of Cardiology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Aleksandra Romaniuk
- Cardiology Student Scientific Circle, Academy of Silesia, 40-555 Katowice, Poland;
| | | | - Wojciech Zygmunt
- Department of Pulmonology, Alergollogy and Oncology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Agnieszka Styczeń
- Department of Cardiology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Małgorzata Romaniuk-Suswał
- Department of Psychiatry, Psychotheraphy and Early Intervention, Medical University of Lublin, 20-954 Lublin, Poland
| |
Collapse
|
15
|
Bao ZY, Li HM, Zhang SB, Fei YQ, Yao MF, Li LJ. Administration of A. muciniphila ameliorates pulmonary arterial hypertension by targeting miR-208a-3p/NOVA1 axis. Acta Pharmacol Sin 2023; 44:2201-2215. [PMID: 37433872 PMCID: PMC10618511 DOI: 10.1038/s41401-023-01126-2] [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: 12/22/2022] [Accepted: 06/08/2023] [Indexed: 07/13/2023]
Abstract
Pulmonary arterial hypertension (PH) is a chronic disease induced by a progressive increase in pulmonary vascular resistance and failure of the right heart function. A number of studies show that the development of PH is closely related to the gut microbiota, and lung-gut axis might be a potential therapeutic target in the PH treatment. A. muciniphila has been reported to play a critical role in treating cardiovascular disorders. In this study we evaluated the therapeutic effects of A. muciniphila against hypoxia-induced PH and the underlying mechanisms. Mice were pretreated with A. muciniphila suspension (2 × 108 CFU in 200 μL sterile anaerobic PBS, i.g.) every day for 3 weeks, and then exposed to hypoxia (9% O2) for another 4 weeks to induce PH. We showed that A. muciniphila pretreatment significantly facilitated the restoration of the hemodynamics and structure of the cardiopulmonary system, reversed the pathological progression of hypoxia-induced PH. Moreover, A. muciniphila pretreatment significantly modulated the gut microbiota in hypoxia-induced PH mice. miRNA sequencing analysis reveals that miR-208a-3p, a commensal gut bacteria-regulated miRNA, was markedly downregulated in lung tissues exposed to hypoxia, which was restored by A. muciniphila pretreatment. We showed that transfection with miR-208a-3p mimic reversed hypoxia-induced abnormal proliferation of human pulmonary artery smooth muscle cells (hPASMCs) via regulating the cell cycle, whereas knockdown of miR-208a-3p abolished the beneficial effects of A. muciniphila pretreatment in hypoxia-induced PH mice. We demonstrated that miR-208a-3p bound to the 3'-untranslated region of NOVA1 mRNA; the expression of NOVA1 was upregulated in lung tissues exposed to hypoxia, which was reversed by A. muciniphila pretreatment. Furthermore, silencing of NOVA1 reversed hypoxia-induced abnormal proliferation of hPASMCs through cell cycle modulation. Our results demonstrate that A. muciniphila could modulate PH through the miR-208a-3p/NOVA1 axis, providing a new theoretical basis for PH treatment.
Collapse
Affiliation(s)
- Zheng-Yi Bao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Hui-Min Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Shuo-Bo Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Yi-Qiu Fei
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Ming-Fei Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China.
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100010, China.
| | - Lan-Juan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China.
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100010, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China.
| |
Collapse
|
16
|
Wang J, Zhang H, Wu L, Lu D. Sacubitril/valsartan mitigated intermittent hypoxia related intestinal microbiota alteration and aortic injury. Sleep Breath 2023; 27:1769-1777. [PMID: 36719525 DOI: 10.1007/s11325-023-02781-9] [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: 06/21/2022] [Revised: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To investigate the influence of sacubitril valsartan sodium (SVS) on chronic intermittent hypoxia (IH) related gut microbiome composition alteration and aortic injury. METHODS Experiments were performed using SD rats, which were divided into three groups: control, IH, and SVS group. O2 concentration was decreased to 7-8% at nadir approximately every 3 min in IH group (8 h per day for 6 weeks) or was left unchanged in control group. Rats in SVS group were orally gavaged with SVS at the dosage of 30 mg/kg/day (2 weeks after chronic IH exposure). At week 6, fecal and aortic samples were harvested for 16 s rDNA analysis and histological analysis, respectively. RESULTS Principal coordinate analysis and non-metric multidimensional scaling analysis indicated that the bacterial community was altered by chronic IH exposure, while SVS treatment restored the intestinal microbial communities. Further analysis showed that IH decreased the relative abundance of Lactobacillus and Prevotella, while rats treated with SVS was enriched with Firmicutes, Bacilli, Prevotellaceae, and Lactobacillus, which was similar to control rats. Immunohistochemical staining showed that SVS prevented the upregulation of transforming growth factor-β1 and tumor necrosis factor-alpha in the aorta. CONCLUSION SVS prevented aortic adverse response to IH, possibly through modulating intestinal microbiota.
Collapse
Affiliation(s)
- Jinfeng Wang
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Hongxiang Zhang
- Department of Cardiology, The Second Affiliated Hospital of Wannan Medical College, 10# Kangfu Road, Wuhu, 241000, Anhui Province, China
- Vascular Diseases Research Center of Wannan Medical College, Wuhu, China
| | - LiJuan Wu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Dasheng Lu
- Department of Cardiology, The Second Affiliated Hospital of Wannan Medical College, 10# Kangfu Road, Wuhu, 241000, Anhui Province, China.
- Translational Medicine Center of the Second Hospital Affiliated Wannan Medical College & Pathogens Detection Engineering Center of Wuhu, Wuhu, China.
| |
Collapse
|
17
|
Liu Y, Deng J, Zhao Y, Liu K, Zhang W, Wang Q, Wang J, Piao C. Outcomes of pregnancy in mice with pulmonary hypertension induced by Hypoxia/SU5416. Biochem Biophys Res Commun 2023; 669:128-133. [PMID: 37269595 DOI: 10.1016/j.bbrc.2023.05.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/26/2023] [Accepted: 05/15/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) seriously affects the health of patients. We have found in clinical studies that PH has adverse effects on both maternal and offspring. OBJECTIVE To establish a animal model of PH induced by hypoxia/SU5416 and observe the effects of PH on pregnant mice and their fetuses. METHODS Twenty-four C57 mice aged 7-9 weeks were selected and divided into 4 groups with 6 mice in each group. ① Female mice with normal oxygen; ② Female mice with hypoxia/SU5416; ③ Pregnant mice with normal oxygen; ④ Pregnant mice with hypoxia/SU5416. After 19 days, weight, right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) were compared in each group. Lung tissue and right ventricular blood were collected. The number and weight of fetal mice were also compared between the two pregnant groups. RESULTS There was no significant difference in RVSP and RVHI between female and pregnant mice under the same condition. Compared with normal oxygen condition, two groups of mice in hypoxia/SU5416 had poor development, RVSP and RVHI were significantly increased, the number of fetal mice was small, hypoplasia, degeneration and even abortion. CONCLUSION The model of mice PH was successfully established. PH affects the development and health of female and pregnant mice, and seriously affects the fetuses.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital Affiliated to Capital Medical University, Chaoyang District, Beijing, China
| | - Jing Deng
- School of Basic Medical Sciences, Yanbian University, Yanji, 133000, China
| | - Yichen Zhao
- Department of Valvular Cardiac Surgery Center, Beijing Anzhen Hospital Affiliated to Capital Medical University, Chaoyang District, Beijing, China
| | - Kemin Liu
- Department of Valvular Cardiac Surgery Center, Beijing Anzhen Hospital Affiliated to Capital Medical University, Chaoyang District, Beijing, China
| | - Wenbo Zhang
- Department of Valvular Cardiac Surgery Center, Beijing Anzhen Hospital Affiliated to Capital Medical University, Chaoyang District, Beijing, China
| | - Qiang Wang
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital Affiliated to Capital Medical University, Chaoyang District, Beijing, China.
| | - Jiangang Wang
- Department of Valvular Cardiac Surgery Center, Beijing Anzhen Hospital Affiliated to Capital Medical University, Chaoyang District, Beijing, China.
| | - Chunmei Piao
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Chaoyang District, Beijing, China.
| |
Collapse
|
18
|
Sanada TJ, Hosomi K, Park J, Naito A, Sakao S, Tanabe N, Kunisawa J, Tatsumi K, Suzuki T. Partially hydrolyzed guar gum suppresses the progression of pulmonary arterial hypertension in a SU5416/hypoxia rat model. Pulm Circ 2023; 13:e12266. [PMID: 37448440 PMCID: PMC10336776 DOI: 10.1002/pul2.12266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
This study investigated the effects of partially hydrolyzed guar gum (PHGG) on the development of pulmonary arterial hypertension using a SU5416/hypoxia rat model. Our results demonstrated that PHGG treatment suppressed the development of pulmonary hypertension and vascular remodeling with an altered gut microbiota composition.
Collapse
Affiliation(s)
- Takayuki J. Sanada
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN)OsakaJapan
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN)OsakaJapan
| | - Jonguk Park
- Artificial Intelligence Center for Health and Biomedical ResearchNational Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)OsakaJapan
| | - Akira Naito
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
- Department of RespirologyChibaken Saiseikai Narashino HospitalNarashinoJapan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN)OsakaJapan
- Artificial Intelligence Center for Health and Biomedical ResearchNational Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)OsakaJapan
- Graduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
- Graduate School of MedicineOsaka UniversityOsakaJapan
- Graduate School of DentistryOsaka UniversityOsakaJapan
- Graduate School of ScienceOsaka UniversityOsakaJapan
- Department of Microbiology and Immunology, Graduate School of MedicineKobe UniversityHyogoJapan
- International Vaccine Design Center, Institute of Medical ScienceUniversity of TokyoTokyoJapan
- Research Organization for Nano and Life InnovationWaseda UniversityTokyoJapan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| |
Collapse
|
19
|
Moutsoglou DM, Tatah J, Prisco SZ, Prins KW, Staley C, Lopez S, Blake M, Teigen L, Kazmirczak F, Weir EK, Kabage AJ, Guan W, Khoruts A, Thenappan T. Pulmonary Arterial Hypertension Patients Have a Proinflammatory Gut Microbiome and Altered Circulating Microbial Metabolites. Am J Respir Crit Care Med 2023; 207:740-756. [PMID: 36343281 PMCID: PMC10037487 DOI: 10.1164/rccm.202203-0490oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Rationale: Inflammation drives pulmonary arterial hypertension (PAH). Gut dysbiosis causes immune dysregulation and systemic inflammation by altering circulating microbial metabolites; however, little is known about gut dysbiosis and microbial metabolites in PAH. Objectives: To characterize the gut microbiome and microbial metabolites in patients with PAH. Methods: We performed 16S ribosomal RNA gene and shotgun metagenomics sequencing on stool from patients with PAH, family control subjects, and healthy control subjects. We measured markers of inflammation, gut permeability, and microbial metabolites in plasma from patients with PAH, family control subjects, and healthy control subjects. Measurements and Main Results: The gut microbiome was less diverse in patients with PAH. Shannon diversity index correlated with measures of pulmonary vascular disease but not with right ventricular function. Patients with PAH had a distinct gut microbial signature at the phylogenetic level, with fewer copies of gut microbial genes that produce antiinflammatory short-chain fatty acids (SCFAs) and secondary bile acids and lower relative abundances of species encoding these genes. Consistent with the gut microbial changes, patients with PAH had relatively lower plasma concentrations of SCFAs and secondary bile acids. Patients with PAH also had enrichment of species with the microbial genes that encoded the proinflammatory microbial metabolite trimethylamine. The changes in the gut microbiome and circulating microbial metabolites between patients with PAH and family control subjects were not as substantial as the differences between patients with PAH and healthy control subjects. Conclusions: Patients with PAH have proinflammatory gut dysbiosis, in which lower circulating SCFAs and secondary bile acids may facilitate pulmonary vascular disease. These findings support investigating modulation of the gut microbiome as a potential treatment for PAH.
Collapse
Affiliation(s)
| | - Jasmine Tatah
- Division of Cardiovascular Medicine, Department of Medicine
| | | | - Kurt W. Prins
- Division of Cardiovascular Medicine, Department of Medicine
| | - Christopher Staley
- Division of Basic and Translational Research, Department of Surgery, and
| | - Sharon Lopez
- Division of Gastroenterology, Hepatology, and Nutrition
| | - Madelyn Blake
- Division of Cardiovascular Medicine, Department of Medicine
| | - Levi Teigen
- Division of Gastroenterology, Hepatology, and Nutrition
| | | | | | | | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | | | | |
Collapse
|
20
|
Mock community as an in situ positive control for amplicon sequencing of microbiotas from the same ecosystem. Sci Rep 2023; 13:4056. [PMID: 36906688 PMCID: PMC10008532 DOI: 10.1038/s41598-023-30916-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 03/03/2023] [Indexed: 03/13/2023] Open
Abstract
Metataxonomy has become the standard for characterizing the diversity and composition of microbial communities associated with multicellular organisms and their environment. Currently available protocols for metataxonomy assume a uniform DNA extraction, amplification and sequencing efficiency for all sample types and taxa. It has been suggested that the addition of a mock community (MC) to biological samples before the DNA extraction step could aid identification of technical biases during processing and support direct comparisons of microbiota composition, but the impact of MC on diversity estimates of samples is unknown. Here, large and small aliquots of pulverized bovine fecal samples were extracted with no, low or high doses of MC, characterized using standard Illumina technology for metataxonomics, and analysed with custom bioinformatic pipelines. We demonstrated that sample diversity estimates were distorted only if MC dose was high compared to sample mass (i.e. when MC > 10% of sample reads). We also showed that MC was an informative in situ positive control, permitting an estimation of the sample 16S copy number, and detecting sample outliers. We tested this approach on a range of sample types from a terrestrial ecosystem, including rhizosphere soil, whole invertebrates, and wild vertebrate fecal samples, and discuss possible clinical applications.
Collapse
|
21
|
Gut Microbiota and Metabolome Changes in Three Pulmonary Hypertension Rat Models. Microorganisms 2023; 11:microorganisms11020472. [PMID: 36838437 PMCID: PMC9959815 DOI: 10.3390/microorganisms11020472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Dysbiosis of the gut microbiota and metabolites is found in both pulmonary hypertension patients and pulmonary hypertension rodent models. However, the exact changes in gut microbiota during the development of pulmonary hypertension is unclear. The function of the gut microbiota is also ambiguous. Here, this study showed that the gut microbiota was disrupted in rats with hypoxia (Hyp)-, hypoxia/Sugen5416 (HySu)-, and monocrotaline (MCT)-induced pulmonary hypertension. The gut microbiota is dynamically changed during the development of Hyp-, HySu-, and MCT-induced rat pulmonary hypertension. The variation in the α diversity of the gut microbiota in Hyp-induced pulmonary hypertension rats was similar to that in rats with MCT-induced pulmonary hypertension and different from that in rats with HySu-induced pulmonary hypertension. In addition, six plasma biomarkers, His, Ala, Ser, ADMA, 2-hydroxybutyric acid, and cystathionine, were identified in Hyp-induced pulmonary hypertension rats. Furthermore, a disease-associated network connecting Streptococcus with Hyp-induced pulmonary hypertension-associated metabolites was described here, including trimethylamine N-oxide, Asp, Asn, Lys, His, Ser, Pro, and Ile.
Collapse
|
22
|
Wang T, Xing Y, Peng B, Yang K, Zhang C, Chen Y, Geng G, Li Q, Fu J, Li M, Luo Z, Fu Z, Wang J. Respiratory Microbiome Profile of Pediatric Pulmonary Hypertension Patients Associated With Congenital Heart Disease. Hypertension 2023; 80:214-226. [PMID: 36353996 PMCID: PMC9722361 DOI: 10.1161/hypertensionaha.122.19182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Pulmonary hypertension (PH) associated with congenital heart disease (CHD) is the most common type of PH in pediatric patients. The airway microbiome profile in CHD-PH patients remains rarely studied. METHODS A total of 158 children were recruited for collection of oropharyngeal swabs to sequence the 16S ribosomal RNA (16S rRNA) V3-V4 region of respiratory microbiome, to establish a correlation between these bacterial groups and echocardiography indicators in CHD-PH patients. RESULTS Bacterial α- and β-diversity of the airway microbiome indicated a significantly lower richness in the CHD-PH group and compositional differences associated with the specific taxa and their relative abundances in the upper respiratory tract. Principal coordinate analysis showed that the pharynx microbiota composition in the CHD-PH group varied from that in the CHD or control group. The linear discriminant analysis effect size also highlighted an increased presence of Streptococcus and Rothia in pediatric CHD-PH patients. Comparison of microbial composition between pediatric and adult PH patients showed significant differences and separation of microbiota. The correlation between bacterial abundance and transthoracic echocardiography indexes in CHD-associated PH indicated that different groups of microbiomes may be related to different PH grades. CONCLUSIONS In summary, our study reported the systematic definition and divergent profile of the upper respiratory tract microbiota in pediatric PH patients, CHD and reference subjects, as well as between pediatric and adult PH patients.
Collapse
Affiliation(s)
- Ting Wang
- Department of Respiratory Children’s Hospital of Chongqing Medical University (T.W., B.P., G.G., Q.L., Z.L., Z.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Yue Xing
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (Y.X., K.Y., C.Z., Y.C., J.W.)
| | - Bingming Peng
- Department of Respiratory Children’s Hospital of Chongqing Medical University (T.W., B.P., G.G., Q.L., Z.L., Z.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (Y.X., K.Y., C.Z., Y.C., J.W.)
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (Y.X., K.Y., C.Z., Y.C., J.W.)
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (Y.X., K.Y., C.Z., Y.C., J.W.)
| | - Gang Geng
- Department of Respiratory Children’s Hospital of Chongqing Medical University (T.W., B.P., G.G., Q.L., Z.L., Z.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Qubei Li
- Department of Respiratory Children’s Hospital of Chongqing Medical University (T.W., B.P., G.G., Q.L., Z.L., Z.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Jian Fu
- Department of thoracic and Cardiac Surgery Children’s Hospital of Chongqing Medical University (J.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Mi Li
- Department of Cardiovascular Medicine Children’s Hospital of Chongqing Medical University (M.L.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Zhengxiu Luo
- Department of Respiratory Children’s Hospital of Chongqing Medical University (T.W., B.P., G.G., Q.L., Z.L., Z.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Zhou Fu
- Department of Respiratory Children’s Hospital of Chongqing Medical University (T.W., B.P., G.G., Q.L., Z.L., Z.F.), National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (Y.X., K.Y., C.Z., Y.C., J.W.)
| |
Collapse
|
23
|
Chen J, Zhou D, Miao J, Zhang C, Li X, Feng H, Xing Y, Zhang Z, Bao C, Lin Z, Chen Y, Yuan JXJ, Sun D, Yang K, Wang J. Microbiome and metabolome dysbiosis of the gut-lung axis in pulmonary hypertension. Microbiol Res 2022; 265:127205. [DOI: 10.1016/j.micres.2022.127205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022]
|
24
|
Ma PJ, Wang MM, Wang Y. Gut microbiota: A new insight into lung diseases. Biomed Pharmacother 2022; 155:113810. [DOI: 10.1016/j.biopha.2022.113810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 11/02/2022] Open
|
25
|
Li B, Jia G, Wen D, Zhao X, Zhang J, Xu Q, Zhao X, Jiang N, Liu Z, Wang Y. Rumen microbiota of indigenous and introduced ruminants and their adaptation to the Qinghai-Tibetan plateau. Front Microbiol 2022; 13:1027138. [PMID: 36299720 PMCID: PMC9589358 DOI: 10.3389/fmicb.2022.1027138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
The grassland in the Qinghai-Tibetan plateau provide habitat for many indigenous and introduced ruminants which perform important ecological functions that impact the whole Qinghai-Tibetan plateau ecosystem. These indigenous Tibetan ruminants have evolved several adaptive traits to withstand the severe environmental conditions, especially cold, low oxygen partial pressure, high altitude, strong UV radiation, and poor forage availability on the alpine rangelands. Despite the challenges to husbandry associated with the need for enhanced adaptation, several domesticated ruminants have also been successfully introduced to the alpine pasture regions to survive in the harsh environment. For ruminants, these challenging conditions affect not only the host, but also their commensal microbiota, especially the diversity and composition of the rumen microbiota; multiple studies have described tripartite interactions among host-environment-rumen microbiota. Thus, there are significant benefits to understanding the role of rumen microbiota in the indigenous and introduced ruminants of the Qinghai-Tibetan plateau, which has co-evolved with the host to ensure the availability of specific metabolic functions required for host survival, health, growth, and development. In this report, we systemically reviewed the dynamics of rumen microbiota in both indigenous and introduced ruminants (including gut microbiota of wild ruminants) as well as their structure, functions, and interactions with changing environmental conditions, especially low food availability, that enable survival at high altitudes. We summarized that three predominant driving factors including increased VFA production, enhanced fiber degradation, and lower methane production as indicators of higher efficiency energy harvest and nutrient utilization by microbiota that can sustain the host during nutrient deficit. These cumulative studies suggested alteration of rumen microbiota structure and functional taxa with genes that encode cellulolytic enzymes to potentially enhance nutrient and energy harvesting in response to low quality and quantity forage and cold environment. Future progress toward understanding ruminant adaptation to high altitudes will require the integration of phenotypic data with multi-omics analyses to identify host-microbiota co-evolutionary adaptations enabling survival on the Qinghai-Tibetan plateau.
Collapse
Affiliation(s)
- Bin Li
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- Agricultural College, Ningxia University, Yinchuan, China
| | - Gaobin Jia
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- Colleges of Life Science and Technology, Dalian University, Dalian Economic Technological Development Zone, Dalian, China
| | - Dongxu Wen
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Xiuxin Zhao
- Agricultural College, Ningxia University, Yinchuan, China
| | - Junxing Zhang
- Agricultural College, Ningxia University, Yinchuan, China
| | - Qing Xu
- Institute of Life Sciences and Bio-Engineering, Beijing Jiaotong University, Beijing, China
| | - Xialing Zhao
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Nan Jiang
- Colleges of Life Science and Technology, Dalian University, Dalian Economic Technological Development Zone, Dalian, China
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yachun Wang
- Agricultural College, Ningxia University, Yinchuan, China
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
26
|
Ma R, Cheng L, Song Y, Sun Y, Gui W, Deng Y, Xie C, Liu M. Altered Lung Microbiome and Metabolome Profile in Children With Pulmonary Arterial Hypertension Associated With Congenital Heart Disease. Front Med (Lausanne) 2022; 9:940784. [PMID: 35966852 PMCID: PMC9366172 DOI: 10.3389/fmed.2022.940784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022] Open
Abstract
Backgrounds Pulmonary arterial hypertension (PAH) is characterized by progressive pulmonary vascular functional and structural changes, resulting in increased pulmonary vascular resistance and eventually right heart failure and death. Congenital Left-to-Right shunts (LTRS) is one type of congenital heart disease (CHD) and PAH associated with the congenital Left-to-Right shunt (PAH-LTRS) is a severe disease in children. However, changes in the lung microbiome and their potential impact on PAH-LTRS have not been not fully studied. We hypothesized that lung microbiota and their derived metabolites have been disturbed in children with PAH-LTRS, which might contribute to the progression and outcomes of PAH-LTRS. Methods In this study, 68 age- and sex-matched children of three different groups (patients with PAH-LTRS cohort, patients with LTRS but have no pathologic features of PAH cohort, and healthy reference cohort) were enrolled in the current study. Bronchoalveolar lavage fluid samples from these participants were conducted for multi-omics analysis, including 16S rRNA sequencing and metabolomic profiling. Data progressing and integration analysis were performed to identify pulmonary microbial and metabolic characteristics of PAH-LTRS in children. Results We found that microbial community density was not significantly altered in PAH-LTRS based on α-diversity analysis. Microbial composition analysis indicated phylum of Bacteroidetes was that less abundant while Lactobacillus, Alicycliphilus, and Parapusillimonas were significantly altered and might contribute to PAH in children with LTRS. Moreover, metabolome profiling data showed that metabolites involved in Purine metabolism, Glycerophospholipid metabolism, Galactose metabolism, and Pyrimidine metabolism were also significantly disturbed in the PAH-LTRS cohort. Correlation analysis between microbes and metabolites indicated that alterations in the microbial composition from the lung microbiota could eventually result in the disturbance in certain metabolites, and might finally contribute to the pathology of PAH-LTRS. Conclusion Lung microbial density was not significantly altered in patients with PAH-LTRS. Composition analysis results showed that the relative microbiome abundance was different between groups. Metabolome profiling and correlation analysis with microbiota showed that metabolome also altered in children with PAH-LTRS. This study indicated that pulmonary microbes and metabolites disturbed in PAH-LTRS could be potentially effective biomarkers and provides valuable perspectives on clinical diagnosis, treatment, and prognosis of pediatric PAH-LTRS.
Collapse
Affiliation(s)
- Runwei Ma
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
- *Correspondence: Runwei Ma
| | - Liming Cheng
- Department of Anesthesiology, Kunming Children's Hospital, Kunming, China
| | - Yi Song
- Department of Extracorporeal Circulation, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| | - Yi Sun
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| | - Wenting Gui
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| | - Yao Deng
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| | - Chao Xie
- Department of Anesthesiology, Kunming Children's Hospital, Kunming, China
| | - Min Liu
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| |
Collapse
|
27
|
Yan D, Sun Y, Zhou X, Si W, Liu J, Li M, Wu M. Regulatory effect of gut microbes on blood pressure. Animal Model Exp Med 2022; 5:513-531. [PMID: 35880388 PMCID: PMC9773315 DOI: 10.1002/ame2.12233] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/25/2022] [Indexed: 12/30/2022] Open
Abstract
Hypertension is an important global public health issue because of its high morbidity as well as the increased risk of other diseases. Recent studies have indicated that the development of hypertension is related to the dysbiosis of the gut microbiota in both animals and humans. In this review, we outline the interaction between gut microbiota and hypertension, including gut microbial changes in hypertension, the effect of microbial dysbiosis on blood pressure (BP), indicators of gut microbial dysbiosis in hypertension, and the microbial genera that affect BP at the taxonomic level. For example, increases in Lactobacillus, Roseburia, Coprococcus, Akkermansia, and Bifidobacterium are associated with reduced BP, while increases in Streptococcus, Blautia, and Prevotella are associated with elevated BP. Furthermore, we describe the potential mechanisms involved in the regulation between gut microbiota and hypertension. Finally, we summarize the commonly used treatments of hypertension that are based on gut microbes, including fecal microbiota transfer, probiotics and prebiotics, antibiotics, and dietary supplements. This review aims to find novel potential genera for improving hypertension and give a direction for future studies on gut microbiota in hypertension.
Collapse
Affiliation(s)
- Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Ye Sun
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Xiaoyue Zhou
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Wenhao Si
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina,Department of Dermatologythe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Jieyu Liu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Minna Wu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
28
|
Huang L, Zhang H, Liu Y, Long Y. The Role of Gut and Airway Microbiota in Pulmonary Arterial Hypertension. Front Microbiol 2022; 13:929752. [PMID: 35910623 PMCID: PMC9326471 DOI: 10.3389/fmicb.2022.929752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe clinical condition that is characterized pathologically by perivascular inflammation and pulmonary vascular remodeling that ultimately leads to right heart failure. However, current treatments focus on controlling vasoconstriction and have little effect on pulmonary vascular remodeling. Better therapies of PAH require a better understanding of its pathogenesis. With advances in sequencing technology, researchers have begun to focus on the role of the human microbiota in disease. Recent studies have shown that the gut and airway microbiota and their metabolites play an important role in the pathogenesis of PAH. In this review, we summarize the current literature on the relationship between the gut and airway microbiota and PAH. We further discuss the key crosstalk between the gut microbiota and the lung associated with PAH, and the potential link between the gut and airway microbiota in the pathogenesis of PAH. In addition, we discuss the potential of using the microbiota as a new target for PAH therapy.
Collapse
Affiliation(s)
- Linlin Huang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Hongdie Zhang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yijun Liu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yang Long
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Yang Long
| |
Collapse
|
29
|
Wu P, Zhu T, Tan Z, Chen S, Fang Z. Role of Gut Microbiota in Pulmonary Arterial Hypertension. Front Cell Infect Microbiol 2022; 12:812303. [PMID: 35601107 PMCID: PMC9121061 DOI: 10.3389/fcimb.2022.812303] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Gut microbiota and its metabolites play an important role in maintaining host homeostasis. Pulmonary arterial hypertension (PAH) is a malignant clinical syndrome with a frightening mortality. Pulmonary vascular remodeling is an important feature of PAH, and its pathogenesis is not well established. With the progress of studies on intestinal microbes in different disease, cumulative evidence indicates that gut microbiota plays a major role in PAH pathophysiology. In this review, we will systematically summarize translational and preclinical data on the correlation between gut dysbiosis and PAH and investigate the role of gut dysbiosis in the causation of PAH. Then, we point out the potential significance of gut dysbiosis in the diagnosis and treatment of PAH as well as several problems that remain to be resolved in the field of gut dysbiosis and PAH. All of this knowledge of gut microbiome might pave the way for the extension of novel pathophysiological mechanisms, diagnosis, and targeted therapies for PAH.
Collapse
|
30
|
Ikubo Y, Sanada TJ, Hosomi K, Park J, Naito A, Shoji H, Misawa T, Suda R, Sekine A, Sugiura T, Shigeta A, Nanri H, Sakao S, Tanabe N, Mizuguchi K, Kunisawa J, Suzuki T, Tatsumi K. Altered gut microbiota and its association with inflammation in patients with chronic thromboembolic pulmonary hypertension: a single-center observational study in Japan. BMC Pulm Med 2022; 22:138. [PMID: 35395844 PMCID: PMC8994357 DOI: 10.1186/s12890-022-01932-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) is considered to be associated with chronic inflammation; however, the underlying mechanism remains unclear. Recently, altered gut microbiota were found in patients with pulmonary arterial hypertension (PAH) and in experimental PAH models. The aim of this study was to characterize the gut microbiota in patients with CTEPH and assess the relationship between gut dysbiosis and inflammation in CTEPH. Methods In this observational study, fecal samples were collected from 11 patients with CTEPH and 22 healthy participants. The abundance of gut microbiota in these fecal samples was assessed using 16S ribosomal ribonucleic acid (rRNA) gene sequencing. Inflammatory cytokine and endotoxin levels were also assessed in patients with CTEPH and control participants. Results The levels of serum tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-8, and macrophage inflammatory protein (MIP)-1α were elevated in patients with CTEPH. Plasma endotoxin levels were significantly increased in patients with CTEPH (P < 0.001), and were positively correlated with TNF-α, IL-6, IL-8, and MIP-1α levels. The 16S rRNA gene sequencing and the principal coordinate analysis revealed the distinction in the gut microbiota between patients with CTEPH (P < 0.01) and control participants as well as the decreased bacterial alpha-diversity in patients with CTEPH. A random forest analysis for predicting the distinction in gut microbiota revealed an accuracy of 80.3%. Conclusion The composition of the gut microbiota in patients with CTEPH was distinct from that of healthy participants, which may be associated with the elevated inflammatory cytokines and endotoxins in CTEPH. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01932-0.
Collapse
Affiliation(s)
- Yumiko Ikubo
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Takayuki Jujo Sanada
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
| | - Jonguk Park
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Hiroki Shoji
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Tomoko Misawa
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Rika Suda
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Respirology, Chibaken Saiseikai Narashino Hospital, Narashino, Japan
| | - Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Toshihiko Sugiura
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Ayako Shigeta
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Hinako Nanri
- Section of Energy Metabolism, Department of Nutrition and Metabolism, National Institute of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Respirology, Chibaken Saiseikai Narashino Hospital, Narashino, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Institute for Protein Research, Osaka University, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| |
Collapse
|
31
|
Aldred MA. Food for Thought: The Emerging Role of Intestinal Microbiota in Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2022; 66:361-362. [PMID: 35143384 PMCID: PMC8990121 DOI: 10.1165/rcmb.2022-0005ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Micheala A. Aldred
- Department of MedicineIndiana University School of MedicineIndianapolis, Indiana
| |
Collapse
|
32
|
Huang Y, Lin F, Tang R, Bao C, Zhou Q, Ye K, Shen Y, Liu C, Hong C, Yang K, Tang H, Wang J, Lu W, Wang T. Gut Microbial Metabolite Trimethylamine N-Oxide Aggravates Pulmonary Hypertension. Am J Respir Cell Mol Biol 2022; 66:452-460. [PMID: 35100519 DOI: 10.1165/rcmb.2021-0414oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Trimethylamine N-oxide (TMAO), a metabolite derived from intestine microbial flora, enhances vascular inflammation in a variety of cardiovascular disease, and the bacterial communities associated with trimethylamine N-oxide (TMAO) metabolism is higher in pulmonary hypertension (PH) patients. The effects of TMAO on PH, however, has not been elucidated. In the present study, we found that circulating TMAO was elevated in intermediate to high-risk PH patients when compared to healthy control or low-risk PH patients. In monocrotaline-induced rat PH models, circulating TMAO was elevated; and reduction of TMAO using 3,3-dimethyl-1-butanol (DMB) significantly decreased right ventricle systolic pressure, pulmonary vascular muscularization in both monocrotaline-induced rat PH and hypoxia induced mice PH models. RNA sequencing of rat lungs revealed that DMB treatment significant suppressed the pathways involved in cytokine-cytokine receptor interaction, and cytokine and chemokine signaling. Protein-protein interaction analysis of the differentially expressed transcripts regulated by DMB showed 5 hub genes with a strong connectivity of proinflammatory cytokines and chemokines including Kng1, Cxcl1, Cxcl2, CxcL6 and Il6. In vitro, TMAO significantly increased the expression of Kng1, Cxcl1, Cxcl2, CxcL6 and Il6 in bone marrow derived macrophage. And TMAO-treated conditioned medium from macrophage increased the proliferation and migration of pulmonary artery smooth muscle cells; but TMAO treatment did not change the proliferation or migration of pulmonary artery smooth muscle cells. In conclusion, our study demonstrates that TMAO is increased in severe PH, and the reduction of TMAO decreases pulmonary vascular muscularization and alleviates PH via suppressing the macrophage production of chemokines and cytokines.
Collapse
Affiliation(s)
- Yuhang Huang
- State Key Laboratory of Respiratory Disease, 555049, Guangzhou, China
| | - Fanjie Lin
- State Key Laboratory of Respiratory Disease, 555049, Guangzhou, China
| | - Ruidi Tang
- State Key Laboratory of Respiratory Disease, 555049, Guangzhou, China
| | - Changlei Bao
- State Key Laboratory of Respiratory Disease, 555049, Guangzhou, China
| | - Qingxun Zhou
- Guangzhou Medical University, Guangzhou Institute of Respiratory Diseases, Guangzhou, China
| | - Kaiwen Ye
- Guangzhou Medical University, 26468, Guangzhou Institute of Respiratory Diseases, Guangzhou, China
| | - Yi Shen
- State Key Laboratory of Respiratory Disease, 555049, Guangzhou, China
| | - Chunli Liu
- Guangzhou Institute of Respiratory Disease, 518877, Respiratory Medicine, Guangzhou, China
| | - Cheng Hong
- Guangzhou Medical University The First Associated Hospital, Guangzhou Institute of Respiratory Diseases, Guangzhou, China
| | - Kai Yang
- Guangzhou Institute of Respiratory Diseases, Guangzhou, China
| | - Haiyang Tang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- The University of Arizona, 8041, Medicine, Tucson, Arizona, United States
| | - Wenju Lu
- Guangzhou Medical University The First Associated Hospital, Guangzhou Institute of Respiratory Diseases, Guangzhou, China
| | - Tao Wang
- Guangzhou Institute of Respiratory Disease, 518877, Respiratory Medicine, Guangzhou, China;
| |
Collapse
|
33
|
Moutsoglou DM. 2021 American Thoracic Society BEAR Cage Winning Proposal: Microbiome Transplant in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:13-16. [PMID: 34758276 PMCID: PMC8865595 DOI: 10.1164/rccm.202108-1833ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
34
|
Chaudhari SN, McCurry MD, Devlin AS. Chains of evidence from correlations to causal molecules in microbiome-linked diseases. Nat Chem Biol 2021; 17:1046-1056. [PMID: 34552222 PMCID: PMC8480537 DOI: 10.1038/s41589-021-00861-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 07/16/2021] [Indexed: 12/27/2022]
Abstract
Human-associated microorganisms play a vital role in human health, and microbial imbalance has been linked to a wide range of disease states. In this Review, we explore recent efforts to progress from correlative studies that identify microorganisms associated with human disease to experiments that establish causal relationships between microbial products and host phenotypes. We propose that successful efforts to uncover phenotypes often follow a chain of evidence that proceeds from (1) association studies; to (2) observations in germ-free animals and antibiotic-treated animals and humans; to (3) fecal microbiota transplants (FMTs); to (4) identification of strains; and then (5) molecules that elicit a phenotype. Using this experimental 'funnel' as our guide, we explore how the microbiota contributes to metabolic disorders and hypertension, infections, and neurological conditions. We discuss the potential to use FMTs and microbiota-inspired therapies to treat human disease as well as the limitations of these approaches.
Collapse
Affiliation(s)
- Snehal N Chaudhari
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Megan D McCurry
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - A Sloan Devlin
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Luo L, Chen Q, Yang L, Zhang Z, Xu J, Gou D. MSCs Therapy Reverse the Gut Microbiota in Hypoxia-Induced Pulmonary Hypertension Mice. Front Physiol 2021; 12:712139. [PMID: 34531759 PMCID: PMC8438532 DOI: 10.3389/fphys.2021.712139] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cell (MSC) therapy is a promising therapeutic approach based on its strong effect on pulmonary hypertension (PH) in rats. However, the detailed mechanism of MSC therapy remains unknown. Alterations in the gut microbiota were found in both type 1 pulmonary arterial hypertension patients and hypoxia/SU5416- or monocrotaline (MCT)-induced PH rats. However, whether the therapeutic mechanism of MSCs is associated with the gut microbiota is poorly understood. Here, we found that gut microbiota homeostasis was disrupted in hypoxia-induced PH mice due to the increased Firmicutes-to-Bacteroidetes (F/B) ratio; enhanced abundances of harmful Marinifilaceae, Helicobacteraceae, and Lactobacillaceae; and decreased abundances of beneficial Bacteroidaceae, Prevotellaceae, Tannerellaceae, and Lachnospiraceae. Unexpectedly, reverses of the increase in disease-associated microbiota and decrease in anti-inflammatory and immunomodulatory functional microbiota were observed in the MSC-treated group. We also identified harmful Erysipelotrichaceae, Alphaproteobacteria, Christensenella timonensis, Coriobacteriales, and Rhodospirillales that may serve as gut microbiota biomarkers of hypoxia-induced PH mice. Micrococcaales, Nesterenkonia, Anaerotruncus, and Tyzzerella may serve as gut microbiota biomarkers of MSC-treated mice. In summary, MSC treatment suppresses hypoxia-induced pulmonary hypertension in mice, and alterated gut microbiota may play a role in the development and progression of PH. The mechanism of MSC therapy is associated with various metabolic pathways of the gut microbiota in hypoxia model PH mice.
Collapse
Affiliation(s)
- Lingjie Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Vascular Disease Research Center, Carson International Cancer Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China.,School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Qinhua Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Vascular Disease Research Center, Carson International Cancer Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Vascular Disease Research Center, Carson International Cancer Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhenxia Zhang
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Jihong Xu
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Vascular Disease Research Center, Carson International Cancer Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
36
|
Karoor V, Strassheim D, Sullivan T, Verin A, Umapathy NS, Dempsey EC, Frank DN, Stenmark KR, Gerasimovskaya E. The Short-Chain Fatty Acid Butyrate Attenuates Pulmonary Vascular Remodeling and Inflammation in Hypoxia-Induced Pulmonary Hypertension. Int J Mol Sci 2021; 22:9916. [PMID: 34576081 PMCID: PMC8467617 DOI: 10.3390/ijms22189916] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive cardiovascular disorder in which local vascular inflammation leads to increased pulmonary vascular remodeling and ultimately to right heart failure. The HDAC inhibitor butyrate, a product of microbial fermentation, is protective in inflammatory intestinal diseases, but little is known regarding its effect on extraintestinal diseases, such as PH. In this study, we tested the hypothesis that butyrate is protective in a Sprague-Dawley (SD) rat model of hypoxic PH. Treatment with butyrate (220 mg/kg intake) prevented hypoxia-induced right ventricular hypertrophy (RVH), hypoxia-induced increases in right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, and permeability. A reversal effect of butyrate (2200 mg/kg intake) was observed on elevated RVH. Butyrate treatment also increased the acetylation of histone H3, 25-34 kDa, and 34-50 kDa proteins in the total lung lysates of butyrate-treated animals. In addition, butyrate decreased hypoxia-induced accumulation of alveolar (mostly CD68+) and interstitial (CD68+ and CD163+) lung macrophages. Analysis of cytokine profiles in lung tissue lysates showed a hypoxia-induced upregulation of TIMP-1, CINC-1, and Fractalkine and downregulation of soluble ICAM (sICAM). The expression of Fractalkine and VEGFα, but not CINC-1, TIMP-1, and sICAM was downregulated by butyrate. In rat microvascular endothelial cells (RMVEC), butyrate (1 mM, 2 and 24 h) exhibited a protective effect against TNFα- and LPS-induced barrier disruption. Butyrate (1 mM, 24 h) also upregulated tight junctional proteins (occludin, cingulin, claudin-1) and increased the acetylation of histone H3 but not α-tubulin. These findings provide evidence of the protective effect of butyrate on hypoxic PH and suggest its potential use as a complementary treatment for PH and other cardiovascular diseases.
Collapse
Affiliation(s)
- Vijaya Karoor
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
| | - Derek Strassheim
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
| | - Timothy Sullivan
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (N.S.U.)
| | - Nagavedi S. Umapathy
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (N.S.U.)
- Center for Blood Disorders, Augusta University, Augusta, GA 30912, USA
| | - Edward C. Dempsey
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Center, Aurora, CO 80045, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Denver, Denver, CO 80204, USA;
| | - Kurt R. Stenmark
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Denver, CO 80204, USA
| | - Evgenia Gerasimovskaya
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Denver, CO 80204, USA
| |
Collapse
|
37
|
Hong W, Mo Q, Wang L, Peng F, Zhou Y, Zou W, Sun R, Liang C, Zheng M, Li H, Zhao D, Gao M, Pu J, Li B, Ran P, Peng G. Changes in the gut microbiome and metabolome in a rat model of pulmonary arterial hypertension. Bioengineered 2021; 12:5173-5183. [PMID: 34405758 PMCID: PMC8806624 DOI: 10.1080/21655979.2021.1952365] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The gut microbiota is widely considered to be involved in several diseases, including atherosclerosis, obesity, chronic obstructive pulmonary disease (COPD) and pulmonary arterial hypertension (PAH). This study aimed to determine if changes in the gut microbiome and metabolome play a major role in the early pathogenesis of PAH. Male Wistar rats were injected with monocrotaline (MCT) (55 mg/kg) at day 1 and injected with calcium-sensing receptor (CaSR) antagonist NPS2143 (4.5 mg/kg/d) from days 1 to 21. Fecal samples were obtained. The gut microbiota and metabolome were analyzed by 16S rRNA gene sequencing and mass spectrometry-based analysis to investigate the effect of PAH in this rat model. MCT injection had a marked effect on the composition of the gut microbiota. This finding was further confirmed by metabolomic analysis with identification of several metabolites relevant to the gut microflora. However, NPS2143 partially abrogated this intestinal flora disorder and reversed fecal metabolite abnormalities. In conclusion, our study shows correlations between changes in the gut microbiome and the metabolome in PAH, which are affected by NPS2143.
Collapse
Affiliation(s)
- Wei Hong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China.,GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiudi Mo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China.,Department of Respiratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Luyao Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Fang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China.,Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Yuming Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Weifeng Zou
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, China
| | - Ruiting Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Chunxiao Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Mengning Zheng
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Haiqing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Dongxing Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Mi Gao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinding Pu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bing Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Gongyong Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Yao K, Wang S, Gaowa N, Huang S, Li S, Shao W. Identification of the molecular mechanisms underlying brisket disease in Holstein heifers via microbiota and metabolome analyses. AMB Express 2021; 11:86. [PMID: 34185184 PMCID: PMC8241945 DOI: 10.1186/s13568-021-01246-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/01/2021] [Indexed: 11/10/2022] Open
Abstract
Brisket disease (BD) is common among Holstein heifers in high-altitude environments, and this disease may result in serious economic loss. At present, no effective treatment is available for brisket disease. In this study, liver and cecum samples were collected from five heifers with BD and five healthy heifers (HH) for analyses of the metabolome and microbiota. The mean pulmonary arterial pressure and systolic blood pressure were significantly higher in BD heifers, whereas the average breathing rate, blood oxygen saturation, and glucose level were significantly lower in BD group than in the HH group. Further, 16S rDNA data showed that the abundance of Firmicutes was significantly lower and that of Bacteroidetes was significantly higher in BD group than in the HH group. At the genus level, the BD group heifers harbored fewer Ruminococcaceae and Lachnospiraceae than the HH group. Several metabolites, including beta-D-fructose, D-ribose, 1,4-beta-D-glucan, sucrose, and glucose-6-phosphate were present at low levels in BD heifers. Moreover, the mean pulmonary arterial pressure was negatively correlated with beta-D-fructose (r = - 0.74; P = 0.013), D-ribose (r = - 0.72; P = 0.018), and acetyl-tyrosine-ethyl-ester (r = - 0.71; P = 0.022). We also found that mean pulmonary arterial pressure was negatively correlated with most of the genera, including those in the families of Lachnospiraceae and Ruminococcaceae. In summary, the decreased levels of metabolites and microbial genera might affect BD by limiting the energy supply. This study may help us better understand the role of the microbiota in BD and provide new insights into the management of feeding to decrease the rate of BD in Holstein dairy cows in the Qinghai-Tibetan plateau.
Collapse
|
39
|
Harmful Effects of COVID-19 on Major Human Body Organs: A Review. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The world experienced the outbreak of a new pandemic disease in 2019, known as coronavirus (CoV) disease 2019 (COVID-19), which is caused by the novel severe acute respiratory syndrome-CoV-2 (SARS-CoV-2). The respiratory system is the organ system most commonly affected by COVID-19; however, several other organ systems have been reported to be affected. The SARS-CoV-2 RNA found in infected stub samples can cause lung contagion by binding to the angiotensin-converting enzyme-2 (ACE-2) receptor of the alveolar epithelial cells. The gut microbiota (GM) promote immunity, indicating that the alignment of the microbiota and corresponding metabolic processes in COVID-19 can help to identify novel biomarkers and new therapeutic targets for this disease. The cause of kidney damage in COVID-19 patients is possibly multifactorial, involving a complex mechanism that involves complement dysregulation and thrombotic microangiopathy, as well as the occurrence of a “cytokine storm” syndrome, which are immune responses that are abandoned and dysfunctional with unfavorable prognosis in severe COVID-19 cases. Furthermore, COVID-19 involves a continuous proliferation and activation of macrophages and lymphocytes. SARS-CoV-2 can also bind to the ACE-2 receptor expressed in the cerebral capillary endothelial cells that can invade the blood-brain wall, to penetrate the brain parenchyma. However, in the ongoing pandemic, there has been a surge in studies on a wide range of topics, including causes of respiratory failure, asymptomatic patients, intensive care patients, and survivors. This review briefly describes the damaging effects of COVID-19 on vital human organs and the inhibitory function of the ACE-2 receptor on the GM, which causes gut dysbiosis, and thus, this review discusses topics that have an opportunity for further investigation.
Collapse
|
40
|
Imaging data in COVID-19 patients: focused on echocardiographic findings. Int J Cardiovasc Imaging 2021; 37:1629-1636. [PMID: 33454898 PMCID: PMC7811393 DOI: 10.1007/s10554-020-02148-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/23/2020] [Indexed: 12/19/2022]
Abstract
To assess imaging data in COVID-19 patients and its association with clinical course and survival and 86 consecutive patients (52 males, 34 females, mean age = 58.8 year) with documented COVID-19 infection were included. Seventy-eight patients (91%) were in severe stage of the disease. All patients underwent transthoracic echocardiography. Mean LVEF was 48.1% and mean estimated systolic pulmonary artery pressure (sPAP) was 27.9 mmHg. LV diastolic dysfunction was mildly abnormal in 49 patients (57.6%) and moderately abnormal in 7 cases (8.2%). Pericardial effusion was present in 5/86 (minimal in size in 3 cases and mild- moderate in 2). In 32/86 cases (37.2%), the severity of infection progressed from “severe” to “critical”. Eleven patients (12.8%) died. sPAP and computed tomography score were associated with disease progression (P value = 0.002, 0.002 respectively). Tricuspid annular plane systolic excursion (TAPSE) was significantly higher in patients with no disease progression compared with those who deteriorated (P value = 0.005). Pericardial effusion (minimal, mild or moderate) was detected more often in progressive disease (P = 0.03). sPAP was significantly lower among survivors (P value = 0.007). Echocardiographic findings (including systolic PAP, TAPSE and pericardial effusion), total CT score may have prognostic and therapeutic implication in COVID-19 patients.
Collapse
|