1
|
Hong Y, Cui J, Xu G, Li N, Peng G. Intestinal IL-17 family orchestrates microbiota-driven histone deacetylation and promotes Treg differentiation to mediate the alleviation of asthma by Ma-Xing-Shi-Gan decoction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156656. [PMID: 40311598 DOI: 10.1016/j.phymed.2025.156656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/08/2025] [Accepted: 03/15/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Gut microbiota imbalance is well-known as one important trigger of allergic asthma. Ma-Xing-Shi-Gan decoction (MXSG) is a traditional Chinese medicine prescription with ideal clinical efficacy on asthma. However, whether and how MXSG exerts its efficacy on asthma through gut microbiota remains unclear. PURPOSE To investigate the underlying mechanism of MXSG against asthma using multi-omics technologies. METHODS An asthma model was established using 8-week-old C57BL/6 J mice, after which they were daily administrated with high-, medium- and low-dose MXSG for 7 days. Histopathological examinations and flow cytometry were performed to evaluate the effects of MXSG on lung immune injury. Key regulatory pathways were predicted via network pharmacology and verified using 16S rRNA sequencing, metagenomics, metabolomics, and in vivo experiments including the knockout of the targeting gene. RESULTS MXSG alleviated asthma symptoms, elevated intestinal microbial diversities, and enriched potential beneficial microbes such as Lactococcus, Lactobacillus, and Limosilactobacillus. Network pharmacology and experimental validation highlighted the IL-17/Treg signaling as crucial for asthma treatment. IL-17 knockout experiments revealed its necessity for Treg differentiation during asthma. Moreover, IL-17-deficient asthmatic mice exhibited lower levels of Lactobacillus and significant changes in microbial genes involving histone deacetylases (HDAC) and short-chain fatty acids (SCFAs). Finally, MXSG significantly boosted SCFA production and reduced HDAC9 expression, which were correlated with Treg cell ratios. CONCLUSION Our study delineates a novel mechanism where MXSG synergizes with the IL-17 family to enrich intestinal beneficial microbes (e.g. Lactobacillus) and SCFAs. This inhibits the expression of SCFA-downstream HDAC9 to promote Treg differentiation, and thus potentially alleviates asthma.
Collapse
Affiliation(s)
- Yanfei Hong
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China
| | - Jiaqi Cui
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China
| | - Guichuan Xu
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China
| | - Na Li
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China.
| | - Guiying Peng
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102401, PR China.
| |
Collapse
|
2
|
Choi S, Yoo SA, Ji KY, Jung DH, Lee S, Lee KG, Kim KM, Lee JY, Jung MA, Pyun BJ, Hur J, Choi JY, Rhee CK, Kim WU, Kim T. Asthma Alleviation by Ginsenoside Rb1 via Promotion of Treg Proliferation and Inflammatory T Cell Inhibition. Allergy 2025. [PMID: 40251907 DOI: 10.1111/all.16551] [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: 07/22/2024] [Revised: 01/09/2025] [Accepted: 02/20/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Regulatory T cells (Tregs) are living drugs with feasibility, tolerability, and therapeutic benefits. Although Tregs are linked to asthma prognosis through inflammation regulation, no therapeutic agents specifically designed to manage asthma by upregulating Tregs have been developed to date. METHODS We screened a library of 250 natural products using a cytometric bead array. Among the selected candidates, gRb1 was identified for further investigation. The effects of gRb1 on Treg and Th17 populations were evaluated in mouse asthma models and human PBMCs from both healthy donors and asthma patients using flow cytometry and cytokine analysis. RESULTS In inflammatory conditions, ginsenoside Rb1 (gRb1, a major ginseng component) increased IL-10- and TGF-β-expressing Treg populations and decreased the Th17 population; activated phospho-STAT5 and NFAT1 in Tregs; inhibited NFAT1 activation in conventional T cells (Tconvs); increased Treg proliferation and Tconv-Treg differentiation, inhibiting Tconv proliferation; and reduced inflammatory cytokine secretion by Tconvs. In asthma model mice, suppression of asthma symptoms by gRb1 was associated with elevated Treg and lower Th17, Th1, and Th2 counts. gRb1 treatment of stimulated PBMCs from patients with asthma and healthy donors increased IL-10- and TGF-β-expressing Treg populations and decreased IL-17A-, IL-22-, IFN-γ-, and TNF-α-expressing T-cell populations. CONCLUSIONS gRb1 alleviate asthma by shifting the Treg-inflammatory T cell balance. These findings suggest a strategy for enhancing Treg activity through treatment with gRb1. This may provide a novel therapeutic approach for asthma and related disorders.
Collapse
Affiliation(s)
- Susanna Choi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Seung-Ah Yoo
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kon-Young Ji
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Dong Ho Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang-Gu Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ki-Myo Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul National University Boramae Hospital, Seoul, Republic of Korea
| | - Joo Young Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Myung-A Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Bo-Jeong Pyun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Jung Hur
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| |
Collapse
|
3
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 311] [Impact Index Per Article: 155.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
4
|
van der Burg N, Stenberg H, Ekstedt S, Diamant Z, Bornesund D, Ankerst J, Kumlien Georén S, Cardell LO, Bjermer L, Erjefält J, Tufvesson E. Neutrophil phenotypes in bronchial airways differentiate single from dual responding allergic asthmatics. Clin Exp Allergy 2023; 53:65-77. [PMID: 35437872 PMCID: PMC10083921 DOI: 10.1111/cea.14149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/18/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Allergic asthmatics with both an early (EAR) and a late allergic reaction (LAR) following allergen exposure are termed 'dual responders' (DR), while 'single responders' (SR) only have an EAR. Mechanisms that differentiate DR from SR are largely unknown, particularly regarding the role and phenotypes of neutrophils. Therefore, we aimed to study neutrophils in DR and SR asthmatics. METHODS Thirty-four allergic asthmatics underwent an inhaled allergen challenge, samples were collected before and up to 24 h post-challenge. Cell differentials were counted from bronchial lavage, alveolar lavage and blood; and tissue neutrophils were quantified in immune-stained bronchial biopsies. Lavage neutrophil nuclei lobe segmentation was used to classify active (1-4 lobes) from suppressive neutrophils (≥5 lobes). Levels of transmigration markers: soluble (s)CD62L and interleukin-1Ra, and activity markers: neutrophil elastase (NE), DNA-histone complex and dsDNA were measured in lavage fluid and plasma. RESULTS Compared with SR at baseline, DR had more neutrophils in their bronchial airways at baseline, both in the lavage (p = .0031) and biopsies (p = .026) and elevated bronchial neutrophils correlated with less antitransmigratory IL-1Ra levels (r = -0.64). DR airways had less suppressive neutrophils and more 3-lobed (active) neutrophils (p = .029) that correlated with more bronchial lavage histone (p = .020) and more plasma NE (p = .0016). Post-challenge, DR released neutrophil extracellular trap factors in the blood earlier and had less pro-transmigratory sCD62L during the late phase (p = .0076) than in SR. CONCLUSION DR have a more active airway neutrophil phenotype at baseline and a distinct neutrophil response to allergen challenge that may contribute to the development of an LAR. Therefore, neutrophil activity should be considered during targeted diagnosis and bio-therapeutic development for DR.
Collapse
Affiliation(s)
- Nicole van der Burg
- Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Henning Stenberg
- Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund University, Lund, Sweden.,Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sandra Ekstedt
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Zuzana Diamant
- Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund University, Lund, Sweden.,Department of Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Leuven, Belgium.,Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Daisy Bornesund
- Department of Experimental Medical Science, Cell and Tissue biology, Lund University, Lund, Sweden
| | - Jaro Ankerst
- Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Susanna Kumlien Georén
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Olaf Cardell
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Department of Otorhinolaryngology, Head & Neck Surgery, Institute of Clinical Sciences, Skane University Hospital, Lund, Sweden
| | - Leif Bjermer
- Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Jonas Erjefält
- Department of Experimental Medical Science, Cell and Tissue biology, Lund University, Lund, Sweden
| | - Ellen Tufvesson
- Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Zhou Y, Hu L, Zhang H, Zhang H, Liu J, Zhao X, Wang J, Wang Q. Guominkang formula alleviate inflammation in eosinophilic asthma by regulating immune balance of Th1/2 and Treg/Th17 cells. Front Pharmacol 2022; 13:978421. [PMID: 36330091 PMCID: PMC9624229 DOI: 10.3389/fphar.2022.978421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/14/2022] [Indexed: 09/17/2023] Open
Abstract
The number of patients with allergic asthma is rising yearly, and hormonal drugs, such as dexamethasone, have unique advantages and certain limitations. In the treatment of allergic diseases especially allergic asthma, increasing the percentage or the function of immunosuppressive cells, such as Treg cells, may achieve a good effect. On the basis of good clinical results, we found that Guominkang (GMK) especially high-concentration GMK can achieve a similar effect with dexamethasone in controlling the symptoms of allergic asthma and inhibiting inflammation of allergic asthma. In our study, GMK can inhibit the recruitment of inflammatory cells, decrease mucus production, and reduce airway resistance. Besides, GMK can reconstruct the cellular immune balance of Th1/2 and Treg/Th17 cells. Metabolome results show that DL-glutamine, L-pyroglutamic acid, prostaglandin b1, prostaglandin e2, and 3,4-dihydroxyhydrocinnamic acid are the metabolic biomarkers and are associated with Th1/2 and Treg/Th17 cell balance. GMK can also change the gut microbiota in the allergic asthma mouse model. The genus_Muriculum, genus_(Clostridium) GCA900066575, genus_klebsiella, genus_Desulfovibrio, genus_Rikenellaceae RC9 gut group, family_Chitinophagaceae, family_Nocardioidaceae, and genus_Corynebacterium are gut microbiota biomarkers treated by GMK. Among these biomarkers, genus_Muriculum is the gut microbiota biomarker associated with Th1/2 and Treg/Th17 cell balance. Interestingly, we first found that DL-glutamine, L-pyroglutamic acid, prostaglandin b1, prostaglandin e2, and 3,4-dihydroxyhydrocinnamic acid are all associated with genus_Muriculum. GMK will be a new strategy for the treatment of eosinophilic asthma, and biomarkers will also be a new research direction.
Collapse
Affiliation(s)
- Yumei Zhou
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linhan Hu
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Honglei Zhang
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Haiyun Zhang
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Juntong Liu
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ji Wang
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Wang
- School of Chinese Medicine, National Institute of TCM Body Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Kim TB, Ionescu L, Avdimiretz N, Murdoch F, Larsen IM, Motyka B, West LJ, Urschel S. Alterations in the immune phenotype of thymectomized children and the development of atopic disorders after heart transplantation. Pediatr Transplant 2022; 26:e14252. [PMID: 35187796 DOI: 10.1111/petr.14252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 01/04/2022] [Accepted: 02/03/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Atopic disorders are more common in children after heart transplant (HTx). We hypothesized that HTx at an early age and thymus excision (TE) affect development of T and B cells, especially regulatory T cells (Tregs), which help maintain tolerance. METHODS In this single-center study including 24 patients transplanted between 2013 and 2018, we investigated lymphocyte patterns in relation to these factors using flow cytometry. Clinical data were collected from standardized questionnaires and medical charts. Patients were stratified into TE and non-TE groups as well as patients with and without post-transplant atopy development/worsening. RESULTS 64% of TE patients experienced new or worsening asthma/eczema post-transplant compared to 20% of non-TE patients. TE patients had higher total Treg proportions (CD4+CD25+CD127lo) than non-TE patients (p = .043), but borderline significantly lower naïve Tregs (CD45RA+CD27-) (p = .057). Memory CD4+ T cells were higher in TE patients in trend (p = .084). Total Tregs did not differ between atopic/nonatopic groups, although naïve Tregs were significantly lower in atopic patients (p = .028). Memory CD4+ T cells were higher in atopic patients in trend (p = .082). IgM+IgD+ B cells were higher in nonatopic patients in trend (p = .064). CONCLUSIONS New/worsening atopy is more common in thymectomized HTx children and is associated with alterations in T-cell profiles. Avoiding TE may prevent these alterations and reduce incidence of atopy post-HTx.
Collapse
Affiliation(s)
- Tiffany B Kim
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lavinia Ionescu
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nicholas Avdimiretz
- Department of Pediatric Respirology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Faye Murdoch
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Ingrid M Larsen
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Bruce Motyka
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lori J West
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Simon Urschel
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Stollery Children's Hospital, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Nozari P, Mokhtari P, Nemati M, Zainodini N, Taghipour Z, Asadi F, Ayoobi F, Jafarzadeh A. Investigation of the effect of IFN-γ/TNF-α-treated mesenchymal stem cells on Th9- and Treg cell-related parameters in a mouse model of ovalbumin-induced allergic asthma. Immunopharmacol Immunotoxicol 2022; 44:773-785. [PMID: 35620857 DOI: 10.1080/08923973.2022.2082977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Th9- and regulatory T (Treg) cells exert pro- and anti-allergic activity, respectively. Mesenchymal stem cell (MSC)-related immunomodulatory impacts can be enhanced by inflammatory cytokines. Here, the modulatory effects of IFN-γ/TNF-α-induced MSCs on Th9- and Treg cell-related parameters were investigated using an asthma model. METHODS Allergic asthma was induced in BALB/c mice using sensitized and challenging with ovalbumin (OVA). The asthmatic groups were treated intraperitoneally with PBS, MSCs, IFN-γ-induced MSCs, TNF-α-induced MSCs and "IFN-γ + TNF-α"-induced MSCs before the challenge phase. The mice were sacrificed 24 hours after challenge. The serum IL-9 and IL-35 levels, as well as gene expression of IL-9, PU.1, IL-35-EBI3 and FOXP3 in the lung tissues were assessed using ELISA and real time-PCR, respectively. RESULTS The differences of Th9 and Treg-related parameters were not significant between untreated asthmatic mice and those treated with non-induced MSCs. In comparison with untreated asthmatic group, treatment with IFN-γ-induced MSCs significantly reduced serum IL-9 levels, reduced lung expression of IL-9 and PU.1, while increasing serum IL-35 levels as well as lung expression of FOXP3; treatment with TNF-α-induced MSCs significantly reduced serum IL-9 levels as well as lung expression of IL-9, and treatment with "IFN-γ + TNF-α"-induced MSCs significantly modulated all investigated Th9 and Treg-related parameters. In comparison to mice treated with non-induced MSCs, serum IL-9 levels were remarkably decreased in mice treated with IFN-γ-induced and "IFN-γ + TNF-α"-induced MSCs. CONCLUSIONS IFN-γ-and "IFN-γ + TNF-α" treated MSCs exerted almost comparable impacts, but were more efficient than TNF-α-exposed MSCs. Thus, IFN-γ alone can be sufficient to promote immunomodulatory effects of MSCs.
Collapse
Affiliation(s)
- Parvin Nozari
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Pejman Mokhtari
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nahid Zainodini
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Taghipour
- Department of Histology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Asadi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Ayoobi
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
8
|
Paris JL, de la Torre P, Flores AI. New Therapeutic Approaches for Allergy: A Review of Cell Therapy and Bio- or Nano-Material-Based Strategies. Pharmaceutics 2021; 13:2149. [PMID: 34959429 PMCID: PMC8707403 DOI: 10.3390/pharmaceutics13122149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023] Open
Abstract
Allergy constitutes a major health issue due to its large prevalence. The established therapeutic approaches (allergen avoidance, antihistamines, and corticosteroids) do not address the underlying causes of the pathology, highlighting the need for other long-term treatment options. Antigen-specific immunotherapy enables the long-term control of allergic diseases by promoting immunological tolerance to the allergen. However, efficacious immunotherapies are not available for all possible allergens, and the risk of undesired reactions during therapy remains a concern, especially in patients with severe allergic reactions. In this context, two types of therapeutic strategies appear especially promising for the future in the context of allergy: cell therapy and bio- or nano-material-based therapy. In this review, the main strategies developed this far in these two types of strategies are discussed, with several examples illustrating the different approaches.
Collapse
Affiliation(s)
- Juan L. Paris
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain;
- Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, 29590 Málaga, Spain
| | - Paz de la Torre
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| |
Collapse
|
9
|
Van Zeebroeck L, Arroyo Hornero R, Côrte-Real BF, Hamad I, Meissner TB, Kleinewietfeld M. Fast and Efficient Genome Editing of Human FOXP3 + Regulatory T Cells. Front Immunol 2021; 12:655122. [PMID: 34408743 PMCID: PMC8365355 DOI: 10.3389/fimmu.2021.655122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
FOXP3+ regulatory T cells (Tregs) are central for maintaining peripheral tolerance and immune homeostasis. Because of their immunosuppressive characteristics, Tregs are a potential therapeutic target in various diseases such as autoimmunity, transplantation and infectious diseases like COVID-19. Numerous studies are currently exploring the potential of adoptive Treg therapy in different disease settings and novel genome editing techniques like CRISPR/Cas will likely widen possibilities to strengthen its efficacy. However, robust and expeditious protocols for genome editing of human Tregs are limited. Here, we describe a rapid and effective protocol for reaching high genome editing efficiencies in human Tregs without compromising cell integrity, suitable for potential therapeutic applications. By deletion of IL2RA encoding for IL-2 receptor α-chain (CD25) in Tregs, we demonstrated the applicability of the method for downstream functional assays and highlighted the importance for CD25 for in vitro suppressive function of human Tregs. Moreover, deletion of IL6RA (CD126) in human Tregs elicits cytokine unresponsiveness and thus may prevent IL-6-mediated instability of Tregs, making it an attractive target to potentially boost functionality in settings of adoptive Treg therapies to contain overreaching inflammation or autoimmunity. Thus, our rapid and efficient protocol for genome editing in human Tregs may advance possibilities for Treg-based cellular therapies.
Collapse
Affiliation(s)
- Lauren Van Zeebroeck
- Vlaams Instituut voor Biotechnologie (VIB) Laboratory of Translational Immunomodulation, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Rebeca Arroyo Hornero
- Vlaams Instituut voor Biotechnologie (VIB) Laboratory of Translational Immunomodulation, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Beatriz F. Côrte-Real
- Vlaams Instituut voor Biotechnologie (VIB) Laboratory of Translational Immunomodulation, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Ibrahim Hamad
- Vlaams Instituut voor Biotechnologie (VIB) Laboratory of Translational Immunomodulation, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Torsten B. Meissner
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Markus Kleinewietfeld
- Vlaams Instituut voor Biotechnologie (VIB) Laboratory of Translational Immunomodulation, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
10
|
Mthembu N, Ikwegbue P, Brombacher F, Hadebe S. Respiratory Viral and Bacterial Factors That Influence Early Childhood Asthma. FRONTIERS IN ALLERGY 2021; 2:692841. [PMID: 35387053 PMCID: PMC8974778 DOI: 10.3389/falgy.2021.692841] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Asthma is a chronic respiratory condition characterised by episodes of shortness of breath due to reduced airway flow. The disease is triggered by a hyperreactive immune response to innocuous allergens, leading to hyper inflammation, mucus production, changes in structural cells lining the airways, and airway hyperresponsiveness. Asthma, although present in adults, is considered as a childhood condition, with a total of about 6.2 million children aged 18 and below affected globally. There has been progress in understanding asthma heterogeneity in adults, which has led to better patient stratification and characterisation of multiple asthma endotypes with distinct, but overlapping inflammatory features. The asthma inflammatory profile in children is not well-defined and heterogeneity of the disease is less described. Although many factors such as genetics, food allergies, antibiotic usage, type of birth, and cigarette smoke exposure can influence asthma development particularly in children, respiratory infections are thought to be the major contributing factor in poor lung function and onset of the disease. In this review, we focus on viral and bacterial respiratory infections in the first 10 years of life that could influence development of asthma in children. We also review literature on inflammatory immune heterogeneity in asthmatic children and how this overlaps with early lung development, poor lung function and respiratory infections. Finally, we review animal studies that model early development of asthma and how these studies could inform future therapies and better understanding of this complex disease.
Collapse
Affiliation(s)
- Nontobeko Mthembu
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Paul Ikwegbue
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Health Science Faculty, International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Sabelo Hadebe
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Kammala AK, Yang C, Panettieri RA, Das R, Subramanian H. G Protein-Coupled Receptor Kinase 2 (GRK2) Regulates T Cell Response in a Murine Model of House Dust Mite-Induced Asthma. FRONTIERS IN ALLERGY 2021; 2:656886. [PMID: 35386975 PMCID: PMC8974720 DOI: 10.3389/falgy.2021.656886] [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] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 11/19/2022] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2) is an adapter protein that modulates G protein-coupled receptor (GPCR) signaling. It also regulates the functions and activity of other intracellular proteins in many cell types. Accordingly, GRK2 is thought to contribute to disease progression by a variety of mechanisms related to its multifunctional roles. Indeed, GRK2 levels are enhanced in patient samples as well as in preclinical models of several diseases. We have previously shown that GRK2 regulates mast cell functions, and thereby contributes to exacerbated inflammation during allergic reactions. In the current study, we observed that GRK2 levels are enhanced in the lungs of human asthma patients and in mice sensitized to house dust mite extract (HDME) allergen. Consistent with these findings, interleukin (IL)-4 and IL-13 levels were reduced in the lungs of GRK2+/- mice in a HMDE mouse model of asthma. Because Th2 cells are the major source of these cytokines during asthma, we determined the role of GRK2 in regulating T cell-specific responses in our HMDE mouse model. We observed a significant reduction of airway hyperresponsiveness (AHR), lung eosinophil and lymphocyte counts, serum IgE, Th2 cytokines (IL-4 and IL-13), goblet cell hyperplasia and mucus production in mice that had reduced GRK2 expression specifically in T cells. Collectively, our studies reveal an important role for GRK2 in regulating T cell response during asthma pathogenesis and further elucidation of the mechanisms through which GRK2 modulates airway inflammation will lead to the development of new therapeutic strategies for asthma.
Collapse
Affiliation(s)
- Ananth K. Kammala
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Canchai Yang
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Reynold A. Panettieri
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, United States
| | - Rupali Das
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Hariharan Subramanian
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
12
|
Birmingham JM, Chesnova B, Wisnivesky JP, Calatroni A, Federman J, Bunyavanich S, Busse PJ. The Effect of Age on T-Regulatory Cell Number and Function in Patients With Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:646-654. [PMID: 34212550 PMCID: PMC8255355 DOI: 10.4168/aair.2021.13.4.646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/01/2020] [Accepted: 12/25/2020] [Indexed: 11/20/2022]
Abstract
T-regulatory cells (Tregs) play a key role in suppressing effector cells and maintaining self-tolerance. Studies of younger adults and children suggest that insufficient differentiation and functional defects of Tregs may contribute to the development of asthma; however, data from older patients with asthma are limited. To address the effects of aging on the relationship of Treg frequency and function with clinical outcomes, we collected induced sputum (differential cell count and Treg frequency) and peripheral blood (Treg function and frequency) from aged (> 60 years of age) and younger (20–40 years old) patients with asthma. In younger patients, low Treg suppression was associated with significantly higher mean numbers of emergency department (ED) (1.8 vs. 0.17, P = 0.02) and urgent care visits (2.3 vs. 0.17, P = 0.01) for asthma, and decreased asthma control (mean Asthma Control Test [ACT] score, 17 vs. 21.3, P = 0.01) compared to those with high Treg suppression. In older patients, however, a lower Treg function was not significantly associated with ACT scores (18.2 vs. 13.4, P = 0.10), or the number of ED (P = 0.9) or urgent care visits (P = 0.2). Our data suggest that Tregs have a weak relationship with asthma control and clinical asthma outcomes in older patients and differ from findings in younger patients, where Tregs are more likely to play a protective role.
Collapse
Affiliation(s)
- Janette M Birmingham
- Divisions of Allergy and Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bogdana Chesnova
- Divisions of Allergy and Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan P Wisnivesky
- Divisions of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Divisions of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Jessie Federman
- Divisions of Allergy and Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula J Busse
- Divisions of Allergy and Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
13
|
Azid NA, Ahmad S, Boer JC, Al-Hatamleh MAI, Mohammad N, Mohd Ashari NS, Tan HT, Chen X, Plebanski M, Mohamud R. A profile of TNFR2 + regulatory T cells and CD103 + dendritic cells in the peripheral blood of patients with asthma. Hum Immunol 2020; 81:634-643. [PMID: 32771274 DOI: 10.1016/j.humimm.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023]
Abstract
The interaction of tolerogenic CD103+ dendritic cells (DCs) with regulatory T (Tregs) cells modulates immune responses by inducing immune tolerance. Hence, we determined the proportion of these cells in the peripheral blood mononuclear cells (PBMC) of asthmatic patients. We observed lower trends of CD11b-CD103+ DCs and CD86 within CD11b-CD103+ DCs, while increased levels of Foxp3 expressing CD25+/-TNFR2+ cells in asthmatics. There was a positive correlation in the expression of Foxp3 within CD3+CD4+CD25+TNFR2+ Tregs and CD11b-CD103+ as well as the expression of CD86 within HLA-DR+CD11c+CD11b-CD103+ DCs. In conclusion, we suggest that the increased levels of Tregs in blood could continuously suppress the T helper 2 (Th2) cells activation in the circulation which is also supported by the increase of anti-inflammatory cytokines IL-10 and TNF. Overall, functional immunoregulation of the regulatory cells, particularly Tregs, exhibit immune suppression and induce immune tolerance linked with the immune activation by the antigen presenting cells (APC).
Collapse
Affiliation(s)
- Nor Azrini Azid
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Jennifer C Boer
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia.
| | - Mohammad A I Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Nurashikin Mohammad
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan, Malaysia.
| | - Noor Suryani Mohd Ashari
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan, Malaysia.
| | - Hern Tze Tan
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, China.
| | - Magdalena Plebanski
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia.
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan, Malaysia.
| |
Collapse
|
14
|
Huang J, Liu J, Xian Y, Liu B, Wu Y, Zhan C, Liang W, Luo W, Lai K, Chen R. Elevated Circulating CD4 +CD25 +CD127 -/low Regulatory T Cells in Patients with Non-asthmatic Eosinophilic Bronchitis. Lung 2020; 198:491-497. [PMID: 32367413 DOI: 10.1007/s00408-020-00358-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/24/2020] [Indexed: 01/11/2023]
Abstract
PURPOSE Non-asthmatic eosinophilic bronchitis (NAEB) is a common cause of chronic cough. It is characterized by sputum eosinophilia like asthma but lacks airway hyperresponsiveness. Regulatory T cells (Tregs) are recognized as immune suppressors and are involved in the pathogenesis of asthma. However, the relationship between Tregs and NAEB remains unknown. This study aimed to preliminarily explore the role of Tregs in NAEB by comparing circulating Tregs levels to asthma and healthy controls. METHODS Fractional exhaled nitric oxide (FeNO), spirometry with bronchial provocation test, sputum induction and blood routine test were performed in all subjects. Peripheral blood mononuclear cells were used to detect the Tregs (CD4+CD25+CD127-/low) by flow cytometry. Relationship between the levels of circulating Tregs and clinical indexes was also observed. RESULTS A total of 15 patients with NAEB, 20 patients with asthma and 11 healthy controls were included. The absolute numbers of circulating Tregs in the NAEB group (49.8 ± 18.9 × 103 cells/ml) and asthma group (53.3 ± 18.7 × 103 cells/ml) were higher than that in healthy control group (32.7 ± 11.6 × 103 cells/ml) (both P < 0.01). In total, the level of circulating Tregs showed positive correlation with FeNO (r = 0.30, P < 0.05). CONCLUSION Tregs may play a key role not only in asthmatic patients, but also in patients with NAEB, as reflected by the elevated Tregs in peripheral blood.
Collapse
Affiliation(s)
- Jieru Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.,Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaxing Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Yansi Xian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.,Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bixia Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.,Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yinglin Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.,Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chen Zhan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Wanqin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Wei Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Kefang Lai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Ruchong Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
15
|
Zhang X, Olsen N, Zheng SG. The progress and prospect of regulatory T cells in autoimmune diseases. J Autoimmun 2020; 111:102461. [PMID: 32305296 DOI: 10.1016/j.jaut.2020.102461] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Treg) are an important immune cell population, playing a crucial role in regulating immune tolerance and preventing autoimmune diseases. These cells consist of various cell sub-populations and generally have an immunoregulatory or suppressive role against immune responses. They also have a different cell heterogeneity and each populations has own biological characteristics. Treg deficiency, reduction, instability, reduced vitality and dysfunction all account for multiple autoimmune diseases. In this review, we have systemically reviewed Treg classification, phenotypic features, regulation of Foxp3 expression, plasticity and stability of Treg as well as their relationship with several important autoimmune diseases. We particularly focus on why and how inflammatory and diet environments affect the functional capacity and underlying mechanisms of Treg cell populations. We also summarize new advances in technologies which help to analyze and dissect these cells in molecular levels in-depth. We also clarify the possible clinical relevance on application of these cells in patients with autoimmune diseases. The advantages and weaknesses have been carefully discussed as well. We also propose the possible approaches to overcome these weaknesses of Treg cells in complicate environments. Thus, we have displayed the updated knowledge of Treg cells, which provides an overall insight into the role and mechanisms of Treg cells in autoimmune diseases.
Collapse
Affiliation(s)
- Ximei Zhang
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey, 17033, USA
| | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA.
| |
Collapse
|
16
|
Wang N, Yuan J, Karim MR, Zhong P, Sun YP, Zhang HY, Wang YF. Effects of Mitophagy on Regulatory T Cell Function in Patients With Myasthenia Gravis. Front Neurol 2020; 11:238. [PMID: 32318017 PMCID: PMC7154095 DOI: 10.3389/fneur.2020.00238] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
Objective: This study was conducted to determine whether regulatory T cells (CD4+CD25+T, Tregs) show abnormal mitophagy as well as the function of Tregs in patients with myasthenia gravis (MG). Methods: CD4+T cells and CD4+CD25+Treg cells were obtained from 15 patients with MG (MG group) and 15 controls (N group). Tregs from the MG group were subjected to rapamycin-induced culture for 48 h (Rapa group) and 3-methyladenine-induced culture for 48 h (3-MA group). The levels of mitophagy in Tregs were then observed through electron and confocal microscopy. Expression of the autophagy-related protein LC3-II was detected by western blotting, and mitochondrial function in each group was evaluated by flow cytometry. Inhibition of Treg cell proliferation was detected by flow cytometry. Results: Mitophagy in the MG group was lower than that in the N group; it was higher in the Rapa group compared to that in the MG group and lowered in the 3-MA group than in the MG group. Expression of the autophagy-related protein LC3-II was lower in the MG group than in the N group, higher in the Rapa group than in the MG group, and lower in the 3-MA group than in the MG group. The mitochondrial membrane potential was lower in the MG group compared to that in the N group; it was higher in the Rapa group than in the MG group and lowered in the 3-MA group than in the MG group. Inhibition of Treg proliferation was lower in the MG group than in the N group; it was higher in the Rapa group than in the MG group and lowered in the 3-MA group than in the MG group. Conclusion: The decreased mitochondrial membrane potential and mitophagy in Tregs in the MG group may be related to a decreased inhibition of Treg proliferation. The mitochondrial membrane potential was increased after adding the autophagy agent Rapa to enhance mitophagy, and the proliferation inhibition function of Tregs was also enhanced. The autophagy agent 3-MA down-regulated mitophagy, which decreased the mitochondrial membrane potential and inhibitory effect of Tregs. These results reveal the possible cellular immune mechanism of Treg dysfunction in MG.
Collapse
Affiliation(s)
- Na Wang
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China
| | - Jiang Yuan
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China
| | - Md Rezaul Karim
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China.,Biomedical Research Institute of Hubei University of Medicine, Shiyan, China
| | - Ping Zhong
- Department of Preventive Medicine, Hubei University of Medicine, Shiyan, China
| | - Yan-Peng Sun
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China
| | - Hong-Yan Zhang
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China
| | - Yun-Fu Wang
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China.,Biomedical Research Institute of Hubei University of Medicine, Shiyan, China
| |
Collapse
|
17
|
Mustafa AS. Vaccine Potential of Mycobacterial Antigens against Asthma. Med Princ Pract 2020; 29:404-411. [PMID: 32422630 PMCID: PMC7511680 DOI: 10.1159/000508719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/17/2020] [Indexed: 12/16/2022] Open
Abstract
Asthma is a cause of substantial burden of disease in the world, including both premature deaths and reduced quality of life. A leading hypothesis to explain the worldwide increase of asthma is the "hygiene hypothesis," which suggests that the increase in the prevalence of asthma is due to the reduction in exposure to infections/microbial antigens. In allergic asthma, the most common type of asthma, antigen-specific T helper (Th)2 and Th17 cells and their cytokines are primary mediators of the pathological consequences. In contrast, Th1 and T regulatory (Treg) cells and their cytokines play a protective role. This article aims to review the information on the effect of mycobacteria and their antigens in modulating Th2/Th17 responses towards Th1/Treg responses and protection against asthma in humans and animal models.
Collapse
Affiliation(s)
- Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait,
| |
Collapse
|
18
|
Chimeric antigen receptor -T cell therapy: Applications and challenges in treatment of allergy and asthma. Biomed Pharmacother 2019; 123:109685. [PMID: 31862474 DOI: 10.1016/j.biopha.2019.109685] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/28/2019] [Accepted: 11/16/2019] [Indexed: 01/10/2023] Open
Abstract
Despite the current advancements, cancer treatment approaches have limitations restricting their cure rate. Immunotherapy techniques are among novel and promising cancer therapeutic approaches. Therapeutic antibodies and adoptive cell therapy (ACT) are the main branches of immunotherapy. T lymphocytes and genetically engineered cells are among important cells which can be used in ACT. This review has focused on recent advances in engineered cell-based immunotherapy based on T lymphocytes with chimeric antigen receptors (CARs). CARs are recombinant receptors expressing T cell signaling domains with or without co-stimulatory molecules. CAR-T cells are expanded ex vivo and re-infused to patients in order to improve their therapeutic efficacy. Nowadays, the beneficial function of CAR-T cell therapy has been indicated in various diseases including hematological malignancies, solid tumors, autoimmune diseases, and allergic diseases such as asthma. Furthermore, antigen-specific T regulatory cells (Tregs) and gene-edited T cells seem to be beneficial in controlling inflammation in allergic asthma. In fact, dysregulated function of Tregs is responsible for dominance of T helper 2 immune response and progression of allergic asthma. CAR-Treg cells can also be designed and reproduced using iTreg population to manage asthma. In addition, universal CAR-T cells can be modified to selectively target multiple antigens. The fourth generation CAR-T cells (i.e. TRUCK cells) represent novel strategies to cure asthma and allergic diseases as well. Despite the advantages of CAR-T cells, their applications can be associated with some unwanted reactions such as cytokine storm, anaphylaxis, neurotoxicity, etc. For clinical application, there is a need to prevent and manage these complications by optimizing ACT protocols.
Collapse
|
19
|
Hu W, Ma L, Yang G, Zeng X, Liu J, Cheng B, Hu T, Zhao H, Liu Z. Der p2‑A20 DNA vaccine attenuates allergic inflammation in mice with allergic rhinitis. Mol Med Rep 2019; 20:4925-4932. [PMID: 31638224 PMCID: PMC6854600 DOI: 10.3892/mmr.2019.10760] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
Allergic rhinitis (AR) is a common disease that requires more convenient, safe and effective antigen-specific immunotherapies. The present study aimed to investigate the therapeutic effect of intranasal administration of a eukaryotic expression vector co-expressing Der p2 and A20 protein (pVAX1-Der p2-A20) on mice with allergic rhinitis. The pVAX1-Der p2-A20 vaccine was prepared and encapsulated into poly(L-lactide-co-glycolide) (PLGA) nanoparticles. An allergic rhinitis Balb/c mouse model was established through intraperitoneal sensitization with recombinant Der p2 and cholera toxin followed by intranasal challenge with recombinant Der p2. The treatment effect of the DNA vaccine on nasal allergic inflammation was evaluated, and serum IgE, sIgE, IgG and cytokine levels were determined by ELISA. The percentage of CD4+CD25+Foxp3+Tregs in the spleen was detected by flow cytometry. The DNA vaccine co-expressing Der p2 and A20 was successfully constructed and encapsulated into PLGA nanoparticles. Der p2-A20 DNA vaccine intranasal administration markedly ameliorated Der p2-induced nasal allergic inflammation. The serum Der p2-specific IgE, IL-4 and IL-13 expression levels were inhibited, while the Der p2-specific IgG1, IgG2a and IFN-γ expression levels in the serum and splenic CD4+CD25+Foxp3+Treg population were significantly increased after Der p2-A20 DNA vaccine treatment. These results indicated that the Der p2-A20 DNA vaccine alleviates nasal allergic inflammation and promotes splenic Treg population in mice with allergic rhinitis.
Collapse
Affiliation(s)
- Wenhui Hu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Li Ma
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Gui Yang
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Jiangqi Liu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Baohui Cheng
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Tianyong Hu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Hailiang Zhao
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Zhiqiang Liu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
20
|
Strehl C, Ehlers L, Gaber T, Buttgereit F. Glucocorticoids-All-Rounders Tackling the Versatile Players of the Immune System. Front Immunol 2019; 10:1744. [PMID: 31396235 PMCID: PMC6667663 DOI: 10.3389/fimmu.2019.01744] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
Glucocorticoids regulate fundamental processes of the human body and control cellular functions such as cell metabolism, growth, differentiation, and apoptosis. Moreover, endogenous glucocorticoids link the endocrine and immune system and ensure the correct function of inflammatory events during tissue repair, regeneration, and pathogen elimination via genomic and rapid non-genomic pathways. Due to their strong immunosuppressive, anti-inflammatory and anti-allergic effects on immune cells, tissues and organs, glucocorticoids significantly improve the quality of life of many patients suffering from diseases caused by a dysregulated immune system. Despite the multitude and seriousness of glucocorticoid-related adverse events including diabetes mellitus, osteoporosis and infections, these agents remain indispensable, representing the most powerful, and cost-effective drugs in the treatment of a wide range of rheumatic diseases. These include rheumatoid arthritis, vasculitis, and connective tissue diseases, as well as many other pathological conditions of the immune system. Depending on the therapeutically affected cell type, glucocorticoid actions strongly vary among different diseases. While immune responses always represent complex reactions involving different cells and cellular processes, specific immune cell populations with key responsibilities driving the pathological mechanisms can be identified for certain autoimmune diseases. In this review, we will focus on the mechanisms of action of glucocorticoids on various leukocyte populations, exemplarily portraying different autoimmune diseases as heterogeneous targets of glucocorticoid actions: (i) Abnormalities in the innate immune response play a crucial role in the initiation and perpetuation of giant cell arteritis (GCA). (ii) Specific types of CD4+ T helper (Th) lymphocytes, namely Th1 and Th17 cells, represent important players in the establishment and course of rheumatoid arthritis (RA), whereas (iii) B cells have emerged as central players in systemic lupus erythematosus (SLE). (iv) Allergic reactions are mainly triggered by several different cytokines released by activated Th2 lymphocytes. Using these examples, we aim to illustrate the versatile modulating effects of glucocorticoids on the immune system. In contrast, in the treatment of lymphoproliferative disorders the pro-apoptotic action of glucocorticoids prevails, but their mechanisms differ depending on the type of cancer. Therefore, we will also give a brief insight into the current knowledge of the mode of glucocorticoid action in oncological treatment focusing on leukemia.
Collapse
Affiliation(s)
- Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Berlin, Germany
| | - Lisa Ehlers
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Berlin, Germany
| |
Collapse
|
21
|
Abstract
Asthma is a chronic disease of airway inflammation due to excessive T helper cell type 2 (Th2) response. Present treatment based on inhalation of synthetic glucocorticoids can only control Th2-driven chronic eosinophilic inflammation, but cannot change the immune tolerance of the body to external allergens. Regulatory T cells (Tregs) are the main negative regulatory cells of the immune response. Tregs play a great role in regulating allergic, autoimmune, graft-versus-host responses, and other immune responses. In this review, we will discuss the classification and biological characteristics, the established immunomodulatory mechanisms, and the characteristics of induced differentiation of Tregs. We will also discuss the progress of Tregs in the field of asthma. We believe that further studies on the regulatory mechanisms of Tregs will provide better treatments and control strategies for asthma.
Collapse
Affiliation(s)
- Sheng-Tao Zhao
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.,Department of Respiratory Medcine, Kunming General Hospital of Chengdu Military Region, Kunming 650032, China
| | - Chang-Zheng Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
22
|
Lamarche D, Johnstone J, Zytaruk N, Clarke F, Hand L, Loukov D, Szamosi JC, Rossi L, Schenck LP, Verschoor CP, McDonald E, Meade MO, Marshall JC, Bowdish DME, Karachi T, Heels-Ansdell D, Cook DJ, Surette MG. Microbial dysbiosis and mortality during mechanical ventilation: a prospective observational study. Respir Res 2018; 19:245. [PMID: 30526610 PMCID: PMC6286574 DOI: 10.1186/s12931-018-0950-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022] Open
Abstract
Background Host-associated microbial communities have important roles in tissue homeostasis and overall health. Severe perturbations can occur within these microbial communities during critical illness due to underlying diseases and clinical interventions, potentially influencing patient outcomes. We sought to profile the microbial composition of critically ill mechanically ventilated patients, and to determine whether microbial diversity is associated with illness severity and mortality. Methods We conducted a prospective, observational study of mechanically ventilated critically ill patients with a high incidence of pneumonia in 2 intensive care units (ICUs) in Hamilton, Canada, nested within a randomized trial for the prevention of healthcare-associated infections. The microbial profiles of specimens from 3 anatomical sites (respiratory, and upper and lower gastrointestinal tracts) were characterized using 16S ribosomal RNA gene sequencing. Results We collected 65 specimens from 34 ICU patients enrolled in the trial (29 endotracheal aspirates, 26 gastric aspirates and 10 stool specimens). Specimens were collected at a median time of 3 days (lower respiratory tract and gastric aspirates; interquartile range [IQR] 2–4) and 6 days (stool; IQR 4.25–6.75) following ICU admission. We observed a loss of biogeographical distinction between the lower respiratory tract and gastrointestinal tract microbiota during critical illness. Moreover, microbial diversity in the respiratory tract was inversely correlated with APACHE II score (r = − 0.46, p = 0.013) and was associated with hospital mortality (Median Shannon index: Discharged alive; 1.964 vs. Deceased; 1.348, p = 0.045). Conclusions The composition of the host-associated microbial communities is severely perturbed during critical illness. Reduced microbial diversity reflects high illness severity and is associated with mortality. Microbial diversity may be a biomarker of prognostic value in mechanically ventilated patients. Trial registration ClinicalTrials.gov ID NCT01782755. Registered February 4 2013. Electronic supplementary material The online version of this article (10.1186/s12931-018-0950-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daphnée Lamarche
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jennie Johnstone
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Public Health Ontario, Toronto, ON, Canada
| | - Nicole Zytaruk
- St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - France Clarke
- St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Lori Hand
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Hamilton Health Sciences, Hamilton, ON, Canada
| | - Dessi Loukov
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, Hamilton, ON, Canada
| | - Jake C Szamosi
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Laura Rossi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Louis P Schenck
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Chris P Verschoor
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Ellen McDonald
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Maureen O Meade
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Hamilton Health Sciences, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - John C Marshall
- Department of Surgery, University of Toronto, Toronto, ON, Canada.,Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
| | - Dawn M E Bowdish
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, Hamilton, ON, Canada
| | - Tim Karachi
- Hamilton Health Sciences, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Diane Heels-Ansdell
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Deborah J Cook
- St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada. .,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada. .,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada. .,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
| | | | | | | |
Collapse
|