1
|
Martin I, Silverberg M, Abdelgawad A, Tanaka K, Halloran BA, Nicola T, Myers ED, Desai JP, White CT, Karabayir I, Akbilgic O, Tipton L, Gentle SJ, Ambalavanan N, Peters BM, Vu LD, Jain VG, Lal CV, Cormier SA, Pierre JF, Jilling T, Talati AJ, Willis KA. The fungal microbiota modulate neonatal oxygen-induced lung injury. MICROBIOME 2025; 13:24. [PMID: 39871397 PMCID: PMC11773857 DOI: 10.1186/s40168-025-02032-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND The immature lungs of very preterm infants are exposed to supraphysiologic oxygen, contributing to bronchopulmonary dysplasia (BPD), a chronic lung disease that is the most common morbidity of prematurity. While the microbiota significantly influences neonatal health, the relationship between the intestinal microbiome, particularly micro-eukaryotic members such as fungi and yeast, and lung injury severity in newborns remains unknown. RESULTS Here, we show that the fungal microbiota modulates hyperoxia-induced lung injury severity in very low birth weight premature infants and preclinical pseudohumanized and altered fungal colonization mouse models. Instead of fungal communities dominated by Candida and Saccharomyces, the first stool microbiomes of infants who developed BPD had less interconnected community architectures with a greater diversity of rarer fungi. After using a pseudohumanized model to show that transfer to the neonatal microbiome from infants with BPD increased the severity of lung injury, we used gain and loss of function approaches to demonstrate that modulating the extent of initial neonatal fungal colonization affected the extent of BPD-like lung injury in mice. We also identified alterations in the murine intestinal microbiome and transcriptome associated with augmented lung injury. CONCLUSIONS These findings demonstrate that features of the initial intestinal fungal microbiome are associated with the later development of BPD in premature neonates and exert a microbiome-driven effect that is transferable and modifiable in murine models, which suggests both causality and a potential therapeutic strategy. Video Abstract.
Collapse
Affiliation(s)
- Isaac Martin
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mary Silverberg
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ahmed Abdelgawad
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kosuke Tanaka
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brian A Halloran
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Teodora Nicola
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erin D Myers
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jay P Desai
- Division of Neonatology, Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Catrina T White
- Division of Neonatology, Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ibrahim Karabayir
- Division of Cardiology, Department of Medicine, Wake Forest University School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Oguz Akbilgic
- Division of Cardiology, Department of Medicine, Wake Forest University School of Medicine, Wake Forest University, Winston-Salem, NC, USA
- Epidemiological Cardiology Research Center, Wake Forest University, Winston-Salem, NC, USA
| | - Laura Tipton
- Departments of Biology and Mathematics & Statistics, James Madison University, Harrisonburg, VA, USA
| | - Samuel J Gentle
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Luan D Vu
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Viral G Jain
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charitharth V Lal
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agricultural and Life Sciences, The University of WI-Madison, Madison, WI, USA
| | - Tamás Jilling
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ajay J Talati
- Division of Neonatology, Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Obstetrics and Gynecology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kent A Willis
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
2
|
Xu F, Wu Q, Yang L, Sun H, Li J, An Z, Li H, Wu H, Song J, Chen W, Wu W. Modification of gut and airway microbiota on ozone-induced airway inflammation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176604. [PMID: 39353487 DOI: 10.1016/j.scitotenv.2024.176604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Ground-level ozone (O3) has been shown to induce airway inflammation, the underlying mechanisms remain unclear. The aim of this study was to determine whether gut and airway microbiota dysbiosis, and airway metabolic alterations were associated with O3-induced airway inflammation. Thirty-six 8-week-old male C57BL/6 N mice were divided into 2 groups: sterile water group and broad-spectrum antibiotics group (Abx). Each group was further divided into two subgroups, filtered air group (Air) and O3 group (O3), with 9 mice in each subgroup. Mice in the Air and O3 groups were exposed to filtered air or 1 ppm O3, 4 h/d for 5 consecutive days, respectively. Mice in Abx + Air and Abx + O3 groups were exposed to filtered air or O3, respectively, after drinking broad-spectrum Abx. 24 h after the final O3 exposure, mouse feces and bronchoalveolar lavage fluids (BALF) were collected and subjected to measurements of airway oxidative stress and inflammation biomarkers, 16S rRNA sequencing and metabolite profiling. Hematoxylin-eosin staining of lung tissues was applied to examine the pathological changes of lung tissue. The results showed that O3 exposure resulted in airway oxidative stress and inflammation, as well as gut and airway microbiota dysbiosis, and airway metabolism alteration. Abx pre-treatment markedly changed gut and airway microbiota and promoted O3-induced metabolic disorder and airway inflammation. Spearman correlation analyses indicated that inter-related gut and airway microbiota dysbiosis and airway metabolic disorder were associated with O3-induced airway inflammation. Together, inhaled O3 causes airway inflammation, which may implicate gut and airway microbiota dysbiosis and airway metabolic alterations.
Collapse
Affiliation(s)
- Fei Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Qiong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Lin Yang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Han Sun
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Juan Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Huijun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Hui Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Song
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
3
|
Wu T, Liu H, Xu R, Li Z, Wei Y. Differences in cellular and molecular processes in exposure to PM 2.5 and O 3. ENVIRONMENT INTERNATIONAL 2024; 192:109052. [PMID: 39406161 DOI: 10.1016/j.envint.2024.109052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/06/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Epidemiological and toxicological studies have shown that PM2.5 and O3 could pose significant risks to human health, such as an increased incidence of respiratory and cardiovascular diseases. Usually, the adverse health outcomes induced by PM2.5 and O3 exposure are similar. However, PM2.5 and O3 have distinct physical and chemical properties, with PM2.5 being a solid-liquid mixture and O3 being a strongly oxidizing gaseous pollutant. Therefore, we speculated that there are some differences in biological processes induced by PM2.5 and O3 exposure. In the present study, we investigated the differences induced by PM2.5 and O3 exposure from the perspective of cellular and molecular processes. Firstly, the pulmonary epithelial cells (BEAS-2B) were exposed to different concentrations of PM2.5 or O3 at different durations. Then, we chose experimental models with the concentrations and duration at which the cell survival rate was 50 % after exposure to PM2.5 and O3, which were 100 μg/mL for 24 h for PM2.5, and 200 ppb for 4 h for O3. Our findings indicate that PM2.5 infiltrates cells via endocytosis without causing significant damage to cell membranes, while O3 induces lipid peroxidation at the cell surface. Moreover, the detection of mitochondrial function showed that the content of ATP was significantly reduced after exposure to both PM2.5 and O3. However, we found a significant difference in mtDNA copy number. PM2.5 exposure increased the mtDNA copy number by up-regulating the expression of fission genes (Fis1, Mff, Dnm1). O3 exposure decreased it by up-regulating the expression of fusion gene (Mfn1, Mfn2) and down-regulating the expression of fission gene (Fis1, Dnm1). These results indicate that although both PM2.5 and O3 exposure induced almost exactly similar adverse health outcomes, significant differences do exist in cellular and molecular processes.
Collapse
Affiliation(s)
- Tingting Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; College of Environmental Science And Engineering, Tongji University, Shanghai, China
| | - Hao Liu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Rongrong Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; College of Environmental Science And Engineering, Tongji University, Shanghai, China
| | - Zhigang Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China.
| | - Yongjie Wei
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; College of Environmental Science And Engineering, Tongji University, Shanghai, China; Center for Global Health, School of Public Health, Nanjing Medical University, China.
| |
Collapse
|
4
|
Burak MF, Stanley TL, Lawson EA, Campbell SL, Lynch L, Hasty AH, Domingos AI, Dixit VD, Hotamışlıgil GS, Sheedy FJ, Dixon AE, Brinkley TE, Hill JA, Donath MY, Grinspoon SK. Adiposity, immunity, and inflammation: interrelationships in health and disease: a report from 24th Annual Harvard Nutrition Obesity Symposium, June 2023. Am J Clin Nutr 2024; 120:257-268. [PMID: 38705359 PMCID: PMC11347817 DOI: 10.1016/j.ajcnut.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
The rapidly evolving field of immunometabolism explores how changes in local immune environments may affect key metabolic and cellular processes, including that of adipose tissue. Importantly, these changes may contribute to low-grade systemic inflammation. In turn, chronic low-grade inflammation affecting adipose tissue may exacerbate the outcome of metabolic diseases. Novel advances in our understanding of immunometabolic processes may critically lead to interventions to reduce disease severity and progression. An important example in this regard relates to obesity, which has a multifaceted effect on immunity, activating the proinflammatory pathways such as the inflammasome and disrupting cellular homeostasis. This multifaceted effect of obesity can be investigated through study of downstream conditions using cellular and systemic investigative techniques. To further explore this field, the National Institutes of Health P30 Nutrition Obesity Research Center at Harvard, in partnership with Harvard Medical School, assembled experts to present at its 24th Annual Symposium entitled "Adiposity, Immunity, and Inflammation: Interrelationships in Health and Disease" on 7 June, 2023. This manuscript seeks to synthesize and present key findings from the symposium, highlighting new research and novel disease-specific advances in the field. Better understanding the interaction between metabolism and immunity offers promising preventative and treatment therapies for obesity-related immunometabolic diseases.
Collapse
Affiliation(s)
- Mehmet Furkan Burak
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Sophia L Campbell
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Lydia Lynch
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Ana I Domingos
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, United Kingdom
| | - Vishwa D Dixit
- Department of Pathology, Department of Comparative Medicine, Department of Immunobiology, Yale School of Medicine, and Yale Center for Research on Aging, New Haven, CT, United States
| | - Gökhan S Hotamışlıgil
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Frederick J Sheedy
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Anne E Dixon
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Tina E Brinkley
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Marc Y Donath
- Department of Biomedicine, University of Basel, Basel, Switzerland; Clinic of Endocrinology, Diabetes & Metabolism, University Hospital Basel, Basel, Switzerland
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
5
|
Xiao H, Fang LT, Tang AZ, Chen HL, Xu ML, Wei XS, Pang GD, Li CQ. Mycobacterium vaccae alleviates allergic airway inflammation and airway hyper-responsiveness in asthmatic mice by altering intestinal microbiota. Immunology 2024; 171:595-608. [PMID: 38205925 DOI: 10.1111/imm.13750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Host immunity can influence the composition of the gut microbiota and consequently affect disease progression. Previously, we reported that a Mycobacterium vaccae vaccine could ameliorate allergic inflammation in asthmatic mice by regulating inflammatory immune processes. Here, we investigated the anti-inflammatory effects of M. vaccae on allergic asthma via gut microbiota modulation. An ovalbumin (OVA)-induced asthmatic murine model was established and treated with M. vaccae. Gut microbiota profiles were determined in 18 BALB/c mice using 16S rDNA gene sequencing and metabolomic profiling was performed using liquid chromatography quadrupole time-of-flight mass spectrometry. Mycobacterium vaccae alleviated airway hyper-reactivity and inflammatory infiltration in mice with OVA-induced allergic asthma. The microbiota of asthmatic mice is disrupted and that this can be reversed with M. vaccae. Additionally, a total of 24 differential metabolites were screened, and the abundance of PI(14:1(9Z)/18:0), a glycerophospholipid, was found to be correlated with macrophage numbers (r = 0.52, p = 0.039). These metabolites may affect chemokine (such as macrophage chemoattractant protein-1) concentrations in the serum, and ultimately affect pulmonary macrophage recruitment. Our data demonstrated that M. vaccae might alleviate airway inflammation and hyper-responsiveness in asthmatic mice by reversing imbalances in gut microbiota. These novel mechanistic insights are expected to pave the way for novel asthma therapeutic strategies.
Collapse
Affiliation(s)
- Huan Xiao
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li-Ting Fang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - An-Zhou Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hong-Liu Chen
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mei-Li Xu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao-Shua Wei
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guo-Dong Pang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao-Qian Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Kurihara Y, Tashiro H, Konomi Y, Sadamatsu H, Ihara S, Takamori A, Kimura S, Sueoka-Aragane N, Takahashi K. Thymic stromal lymphopoietin contributes to ozone-induced exacerbations of eosinophilic airway inflammation via granulocyte colony-stimulating factor in mice. Allergol Int 2024; 73:313-322. [PMID: 38145912 DOI: 10.1016/j.alit.2023.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Ozone is one of the triggers of asthma, but its impact on the pathophysiology of asthma, such as via airway inflammation and airway hyperresponsiveness (AHR), is not fully understood. Thymic stromal lymphopoietin (TSLP) is increasingly seen as a crucial molecule associated with asthma severity, such as corticosteroid resistance. METHODS Female BALB/c mice sensitized and challenged with house dust mite (HDM) were exposed to ozone at 2 ppm for 3 h. Airway inflammation was assessed by the presence of inflammatory cells in bronchoalveolar lavage fluid and concentrations of cytokines including TSLP in lung. Anti-TSLP antibody was administered to mice to block the signal. Survival and adhesion of bone marrow-derived eosinophils in response to granulocyte colony-stimulating factor (G-CSF) were evaluated. RESULTS Ozone exposure increased eosinophilic airway inflammation and AHR in mice sensitized and challenged with HDM. In addition, TSLP, but not IL-33 and IL-25, was increased in lung by ozone exposure. To confirm whether TSLP signaling is associated with airway responses to ozone, an anti-TSLP antibody was administered, and it significantly attenuated eosinophilic airway inflammation, but not AHR. Interestingly, G-CSF, but not type 2 cytokines such as IL-4, IL-5, and IL-13, was regulated by TSLP signaling associated with eosinophilic airway inflammation, and G-CSF prolonged survival and activated eosinophil adhesion. CONCLUSIONS The present data show that TSLP contributes to ozone-induced exacerbations of eosinophilic airway inflammation and provide greater understanding of ozone-induced severity mechanisms in the pathophysiology of asthma related to TSLP and G-CSF.
Collapse
Affiliation(s)
- Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan
| | - Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan.
| | - Yoshie Konomi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan
| | - Hironori Sadamatsu
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan
| | - Satoshi Ihara
- Department of Graduate School of Science and Engineering, Saga University, Saga, Japan
| | - Ayako Takamori
- Clinical Research Center, Saga University Hospital, Saga, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University Hospital, Saga, Japan
| |
Collapse
|
7
|
Tashiro H, Kurihara Y, Kuwahara Y, Takahashi K. Impact of obesity in asthma: Possible future therapies. Allergol Int 2024; 73:48-57. [PMID: 37659887 DOI: 10.1016/j.alit.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 09/04/2023] Open
Abstract
Obesity is one of the factors associated with the severity of asthma. Obesity is associated with aggravation of the pathophysiology of asthma, including exacerbations, airway inflammation, decreased pulmonary function, and airway hyperresponsiveness. The present review addresses the characteristics of asthma with obesity, focusing especially on the heterogeneity caused by the degree of type 2 inflammation, sex differences, the onset of asthma, and race differences. To understand the severity mechanisms in asthma and obesity, such as corticosteroid resistance, fatty acids, gut microbiome, and cytokines, several basic research studies are evaluated. Finally, possible future therapies, including weight reduction, microbiome-targeted therapies, and other molecular targeted therapies are addressed. We believe that the present review will contribute to better understanding of the severity mechanisms and the establishment of novel treatments for severe asthma patients with obesity.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan.
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
8
|
Tian Y, Xu P, Wu X, Gong Z, Yang X, Zhu H, Zhang J, Hu Y, Li G, Sang N, Yue H. Lung injuries induced by ozone exposure in female mice: Potential roles of the gut and lung microbes. ENVIRONMENT INTERNATIONAL 2024; 183:108422. [PMID: 38217903 DOI: 10.1016/j.envint.2024.108422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
Ozone (O3) is one of the most harmful pollutants affecting health. However, the potential effects of O3 exposure on microbes in the gut-lung axis related to lung injuries remain elusive. In this study, female mice were exposed to 0-, 0.5- and 1-ppm O3 for 28 days, followed by routine blood tests, lung function tests and histopathological examination of the colon, nasal cavity and lung. Mouse faeces and lungs were collected for 16s rRNA sequencing to assess the overall microbiological profile and screen for key differential enriched microbes (DEMs). The key DEMs in faecal samples were Butyricimonas, Rikenellaceae RC9 and Escherichia-Shigella, whereas those in lung samples were DNF00809, Fluviicola, Bryobacter, Family XII AD3011 group, Sharpea, MND1 and unclassified Phycisphaeraceae. After a search in microbe-disease databases, these key DEMs were found to be associated with lung diseases such as lung neoplasms, cystic fibrosis, pneumonia, chronic obstructive pulmonary disease, respiratory distress syndrome and bronchiectasis. Subsequently, we used transcriptomic data from Gene Expression Omnibus (GEO) with exposure conditions similar to those in this study to cross-reference with Comparative Toxicogenomic Database (CTD). Il-6 and Ccl2 were identified as the key causative genes and were validated. The findings of this study suggest that exposure to O3 leads to significant changes in the microbial composition of the gut and lungs. These changes are associated with increased levels of inflammatory factors in the lungs and impaired lung function, resulting in an increased risk of lung disease. Altogether, this study provides novel insights into the role of microbes present in the gut-lung axis in O3 exposure-induced lung injury.
Collapse
Affiliation(s)
- Yuchai Tian
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Pengchong Xu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaoyun Wu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Zhihua Gong
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China; Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tong ji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, PR China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Xiaowen Yang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huizhen Zhu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Jiyue Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Yangcheng Hu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
9
|
Willis KA, Silverberg M, Martin I, Abdelgawad A, Karabayir I, Halloran BA, Myers ED, Desai JP, White CT, Lal CV, Ambalavanan N, Peters BM, Jain VG, Akbilgic O, Tipton L, Jilling T, Cormier SA, Pierre JF, Talati AJ. The fungal intestinal microbiota predict the development of bronchopulmonary dysplasia in very low birthweight newborns. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.29.23290625. [PMID: 37398134 PMCID: PMC10312873 DOI: 10.1101/2023.05.29.23290625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
RATIONALE Bronchopulmonary dysplasia (BPD) is the most common morbidity affecting very preterm infants. Gut fungal and bacterial microbial communities contribute to multiple lung diseases and may influence BPD pathogenesis. METHODS We performed a prospective, observational cohort study comparing the multikingdom fecal microbiota of 144 preterm infants with or without moderate to severe BPD by sequencing the bacterial 16S and fungal ITS2 ribosomal RNA gene. To address the potential causative relationship between gut dysbiosis and BPD, we used fecal microbiota transplant in an antibiotic-pseudohumanized mouse model. Comparisons were made using RNA sequencing, confocal microscopy, lung morphometry, and oscillometry. RESULTS We analyzed 102 fecal microbiome samples collected during the second week of life. Infants who later developed BPD showed an obvious fungal dysbiosis as compared to infants without BPD (NoBPD, p = 0.0398, permutational multivariate ANOVA). Instead of fungal communities dominated by Candida and Saccharomyces, the microbiota of infants who developed BPD were characterized by a greater diversity of rarer fungi in less interconnected community architectures. On successful colonization, the gut microbiota from infants with BPD augmented lung injury in the offspring of recipient animals. We identified alterations in the murine intestinal microbiome and transcriptome associated with augmented lung injury. CONCLUSIONS The gut fungal microbiome of infants who will develop BPD is dysbiotic and may contribute to disease pathogenesis.
Collapse
|
10
|
Jackson TW, House JS, Henriquez AR, Schladweiler MC, Jackson KM, Fisher AA, Snow SJ, Alewel DI, Motsinger-Reif AA, Kodavanti UP. Multi-tissue transcriptomic and serum metabolomic assessment reveals systemic implications of acute ozone-induced stress response in male Wistar Kyoto rats. Metabolomics 2023; 19:81. [PMID: 37690105 PMCID: PMC11955933 DOI: 10.1007/s11306-023-02043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Air pollutant exposures have been linked to systemic disease; however, the underlying mechanisms between responses of the target tissue and systemic effects are poorly understood. A prototypic inducer of stress, ozone causes respiratory and systemic multiorgan effects through activation of a neuroendocrine stress response. The goal of this study was to assess transcriptomic signatures of multiple tissues and serum metabolomics to understand how neuroendocrine and adrenal-derived stress hormones contribute to multiorgan health outcomes. Male Wistar Kyoto rats (12-13 weeks old) were exposed to filtered air or 0.8 ppm ozone for 4-hours, and blood/tissues were collected immediately post-exposure. Each tissue had distinct expression profiles at baseline. Ozone changed 1,640 genes in lung, 274 in hypothalamus, 2,516 in adrenals, 1,333 in liver, 1,242 in adipose, and 5,102 in muscle (adjusted p-value < 0.1, absolute fold-change > 50%). Serum metabolomic analysis identified 863 metabolites, of which 447 were significantly altered in ozone-exposed rats (adjusted p-value < 0.1, absolute fold change > 20%). A total of 6 genes were differentially expressed in all 6 tissues. Glucocorticoid signaling, hypoxia, and GPCR signaling were commonly changed, but ozone induced tissue-specific changes in oxidative stress, immune processes, and metabolic pathways. Genes upregulated by TNF-mediated NFkB signaling were differentially expressed in all ozone-exposed tissues, but those defining inflammatory response were tissue-specific. Upstream predictor analysis identified common mediators of effects including glucocorticoids, although the specific genes responsible for these predictors varied by tissue. Metabolomic analysis showed major changes in lipids, amino acids, and metabolites linked to the gut microbiome, concordant with transcriptional changes identified through pathway analysis within liver, muscle, and adipose tissues. The distribution of receptors and transcriptional mechanisms underlying the ozone-induced stress response are tissue-specific and involve induction of unique gene networks and metabolic phenotypes, but the shared initiating triggers converge into shared pathway-level responses. This multi-tissue transcriptomic analysis, combined with circulating metabolomic assessment, allows characterization of the systemic inhaled pollutant-induced stress response.
Collapse
Affiliation(s)
- Thomas W Jackson
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - John S House
- Division of Intramural Research, National Institute of Environmental Health Sciences, Department of Health and Human Services, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Andres R Henriquez
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | | | - Anna A Fisher
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Sam J Snow
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
- ICF, Durham, NC, USA
| | - Devin I Alewel
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Allison A Motsinger-Reif
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| |
Collapse
|
11
|
Tashiro H, Takahashi K, Uchida M, Kurihara Y, Sadamatsu H, Takamori A, Kimura S, Sueoka-Aragane N. Effect of Azithromycin on Exacerbations in Asthma Patients with Obesity: Protocol for a Multi-Center, Prospective, Single-Arm Intervention Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1861. [PMID: 36767227 PMCID: PMC9915079 DOI: 10.3390/ijerph20031861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Obesity is associated with severe asthma, but no specific treatment has been established. The gut microbiome is increasingly recognized as a crucial factor, but specific treatments focused on the gut microbiome have not been established. Recently, azithromycin has been found to have the capacity to attenuate exacerbations, a characteristic of severe asthma. The effect of azithromycin on obesity-induced severe asthma is not understood. METHODS The purpose of the present study is to clarify the effect of azithromycin on exacerbations in asthmatic patients with obesity. To explore the mechanism, the gut microbiome, metabolites of microbes such as short-chain fatty acids, and blood inflammatory cytokines will be analyzed to evaluate the correlation with the effect of azithromycin on exacerbations in obesity-induced severe asthma. A multi-center, prospective, single-arm intervention study is planned. DISCUSSION The present study will allow us to evaluate the effect of azithromycin on exacerbations, particularly in asthma patients with obesity, and explore biomarkers, targeting molecules including the gut microbiome, which are correlated with decreased exacerbations. The present results could contribute to identifying new therapeutic prospects and targeted microbes or molecules associated with severe clinical characteristics in asthmatic patients with obesity. TRIAL REGISTRATION This study has been registered as a prospective study with the University Hospital Medical Information Network (UMIN0000484389) and the Japan Registry of Clinical Trials (jRCTs071220023).
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
- TARGET Investigator Group, Saga 8498501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
- TARGET Investigator Group, Saga 8498501, Japan
| | - Masaru Uchida
- TARGET Investigator Group, Saga 8498501, Japan
- Division of Internal Medicine, Japan Community Health Care Organization Saga Central Hospital, Saga 8498522, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
- TARGET Investigator Group, Saga 8498501, Japan
| | - Hironori Sadamatsu
- TARGET Investigator Group, Saga 8498501, Japan
- Division of Respiratory Medicine, Saga Prefectural Medical Center Koseikan, Saga 8408571, Japan
| | - Ayako Takamori
- Clinical Research Center, Saga University Hospital, Saga 8498501, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
| |
Collapse
|
12
|
Rodríguez-Sánchez S, Valiente N, Seseña S, Cabrera-Pinto M, Rodríguez A, Aranda A, Palop L, Fernández-Martos CM. Ozone modified hypothalamic signaling enhancing thermogenesis in the TDP-43 A315T transgenic model of Amyotrophic Lateral Sclerosis. Sci Rep 2022; 12:20814. [PMID: 36460700 PMCID: PMC9718766 DOI: 10.1038/s41598-022-25033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/23/2022] [Indexed: 12/04/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), a devastating progressive neurodegenerative disease, has no effective treatment. Recent evidence supports a strong metabolic component in ALS pathogenesis. Indeed, metabolic abnormalities in ALS correlate to disease susceptibility and progression, raising additional therapeutic targets against ALS. Ozone (O3), a natural bioactive molecule, has been shown to elicit beneficial effects to reduce metabolic disturbances and improved motor behavior in TDP-43A315T mice. However, it is fundamental to determine the mechanism through which O3 acts in ALS. To characterize the association between O3 exposure and disease-associated weight loss in ALS, we assessed the mRNA and protein expression profile of molecular pathways with a main role in the regulation of the metabolic homeostasis on the hypothalamus and the brown adipose tissue (BAT) at the disease end-stage, in TDP-43A315T mice compared to age-matched WT littermates. In addition, the impact of O3 exposure on the faecal bacterial community diversity, by Illumina sequencing, and on the neuromuscular junctions (NMJs), by confocal imaging, were analysed. Our findings suggest the effectiveness of O3 exposure to induce metabolic effects in the hypothalamus and BAT of TDP-43A315T mice and could be a new complementary non-pharmacological approach for ALS therapy.
Collapse
Affiliation(s)
- Sara Rodríguez-Sánchez
- grid.8048.40000 0001 2194 2329Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Toledo, Spain
| | - Nicolas Valiente
- grid.10420.370000 0001 2286 1424Division of Terrestrial Ecosystem Research, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Susana Seseña
- grid.8048.40000 0001 2194 2329Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Toledo, Spain
| | - Marta Cabrera-Pinto
- grid.414883.20000 0004 1767 1847Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Ana Rodríguez
- grid.8048.40000 0001 2194 2329Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Toledo, Spain
| | - Alfonso Aranda
- grid.8048.40000 0001 2194 2329Faculty of Chemical Science and Technology, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Llanos Palop
- grid.8048.40000 0001 2194 2329Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Toledo, Spain
| | - Carmen M. Fernández-Martos
- grid.414883.20000 0004 1767 1847Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain ,grid.1009.80000 0004 1936 826XWicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania Australia
| |
Collapse
|
13
|
Tooba R, Wu TD. Obesity and asthma: A focused review. Respir Med 2022; 204:107012. [PMID: 36279813 PMCID: PMC9671155 DOI: 10.1016/j.rmed.2022.107012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/19/2022]
Affiliation(s)
- Rubabin Tooba
- Department of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Tianshi David Wu
- Department of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Innovations in Quality, Effectiveness, and Safety, Michael E. DeBakey VA Medical Center, Houston, TX, USA.
| |
Collapse
|
14
|
Obesity affects pulmonary function in Japanese adult patients with asthma, but not those without asthma. Sci Rep 2022; 12:16457. [PMID: 36180514 PMCID: PMC9525285 DOI: 10.1038/s41598-022-20924-y] [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: 05/20/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
Obesity is associated with the severity of asthma, which is characterized by airway obstruction. Pulmonary function testing is one of the important examinations for evaluating airway obstruction. However, the impact of obesity on pulmonary function in patients with asthma is not fully understood. A total of 193 patients with asthma and 2159 patients without asthma who visited Saga University Hospital were investigated retrospectively. Obesity was defined as a body mass index (BMI) greater than 25 kg/m2. Pulmonary functions including forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1) were compared between patients with and without asthma, focusing especially on obesity. FVC percent predicted and FEV1 percent predicted were significantly lower in patients with asthma than in those without asthma (p = 0.03, < 0.01 respectively). In patients with asthma, FVC percent predicted and FEV1 percent predicted were significantly lower in patients with obesity than in those without obesity (all p < 0.01). In addition, BMI was negatively correlated with FEV1 (r =- 0.21, p = 0.003) and FVC (r = - 0.15, p = 0.04), along with the percent predicted. On multivariate analysis in patients with asthma, FVC (β [95% confidence interval] 0.12 [0.02-0.22], p = 0.02) and FEV1 (0.13 [0.05-0.22], p < 0.01) were still significantly different between patients with and without obesity. However, these obesity-associated differences were not observed in patients without asthma. Obesity reduces pulmonary function, including FVC and FEV1, in patients with asthma, but not in those without asthma.
Collapse
|
15
|
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: 35] [Impact Index Per Article: 11.7] [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
|
16
|
Yi W, Ji Y, Gao H, Pan R, Wei Q, Cheng J, Song J, He Y, Tang C, Liu X, Song S, Su H. Does the gut microbiome partially mediate the impact of air pollutants exposure on liver function? Evidence based on schizophrenia patients. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 291:118135. [PMID: 34534831 DOI: 10.1016/j.envpol.2021.118135] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Air pollution may alter the composition of gut microbiome and subsequent liver-related metabolic disorders. Schizophrenia was often accompanied by liver dysfunction. But it was still unclear whether air pollutants affected liver function in patients with schizophrenia through gut microbiome. We aimed to clarify the impacts of long-term air pollutants on the gut microbiome and liver function in schizophrenia and to evaluate the intermediary effect of microbiome. Schizophrenia patients were recruited then serum biochemical indicators were tested. Air pollutant exposure in the previous year was retrospectively estimated by inverse distance weighting. The associations among air pollutants, gut microbiome, and liver function indicators in schizophrenia were estimated. Then the mediating effect of gut microbiome was further explored. The results showed that nitrogen dioxide (NO2), carbonic oxide (CO), ozone (O3), particulate matter with aerodynamic diameter ≤10 μm (PM10), and fine particulate matter (PM2.5) explained 2.68%-10.77% of the variation in gut microbiome composition (order level) in schizophrenia (all P < 0.05). Network correlation analysis indicated that air pollutants and liver function indicators were mainly related to Firmicutes, Actinobacteria, and Proteobacteria in schizophrenia. Long-term NO2 exposure significantly increased the levels of gamma-glutamyl transpeptidase (GGT) and glutamic pyruvic transaminase (GPT) in schizophrenia. Coriobacteriales mediated 13.98% and 49.56% (all P < 0.05) of the associations of long-term NO2 with GGT and GPT, respectively. To conclude, long-term NO2 exposure is positively associated with liver dysfunction in schizophrenia, in which gut microbiome plays an intermediary role. The two pathways, "NO2-Coriobacteriales-GGT" and "NO2-Coriobacteriales-GPT", would provide scientific evidence for the intervention of schizophrenia with liver dysfunction.
Collapse
Affiliation(s)
- Weizhuo Yi
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yifu Ji
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Hua Gao
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Rubing Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Qiannan Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Jian Cheng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Jian Song
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yangyang He
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Chao Tang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Xiangguo Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Shasha Song
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Hong Su
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China.
| |
Collapse
|
17
|
Pyclik MJ, Srutkova D, Razim A, Hermanova P, Svabova T, Pacyga K, Schwarzer M, Górska S. Viability Status-Dependent Effect of Bifidobacterium longum ssp . longum CCM 7952 on Prevention of Allergic Inflammation in Mouse Model. Front Immunol 2021; 12:707728. [PMID: 34354710 PMCID: PMC8329652 DOI: 10.3389/fimmu.2021.707728] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/20/2022] Open
Abstract
The classical definition of probiotics states that bacteria must be alive to be beneficial for human organism. However, recent reports show that inactivated bacteria or their effector molecules can also possess such properties. In this study, we investigated the physical and immunomodulatory properties of four Bifidobacterium strains in the heat-treated (HT) and untreated (UN) forms. We showed that temperature treatment of bacteria changes their size and charge, which affects their interaction with epithelial and immune cells. Based on the in vitro assays, we observed that all tested strains reduced the level of OVA-induced IL-4, IL-5, and IL-13 in the spleen culture of OVA-sensitized mice. We selected Bifidobacterium longum ssp. longum CCM 7952 (Bl 7952) for further analysis. In vivo experiments confirmed that untreated Bl 7952 exhibited allergy-reducing properties when administered intranasally to OVA-sensitized mice, which manifested in significant suppression of airway inflammation. Untreated Bl 7952 decreased local and systemic levels of Th2 related cytokines, OVA-specific IgE antibodies and simultaneously inhibited airway eosinophilia. In contrast, heat-treated Bl 7952 was only able to reduce IL-4 levels in the lungs and eosinophils in bronchoalveolar lavage, but increased neutrophil and macrophage numbers. We demonstrated that the viability status of Bl 7952 is a prerequisite for the beneficial effects of bacteria, and that heat treatment reduces but does not completely abolish these properties. Further research on bacterial effector molecules to elucidate the beneficial effects of probiotics in the prevention of allergic diseases is warranted.
Collapse
Affiliation(s)
- Marcelina Joanna Pyclik
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Agnieszka Razim
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Petra Hermanova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Tereza Svabova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Katarzyna Pacyga
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|