1
|
Hsieh LL, Looney M, Figueroa A, Massaccesi G, Stavrakis G, Anaya EU, D'Alessio FR, Ordonez AA, Pekosz AS, DeFilippis VR, Karakousis PC, Karaba AH, Cox AL. Bystander monocytic cells drive infection-independent NLRP3 inflammasome response to SARS-CoV-2. mBio 2024; 15:e0081024. [PMID: 39240187 PMCID: PMC11481483 DOI: 10.1128/mbio.00810-24] [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: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
The pathogenesis of COVID-19 is associated with a hyperinflammatory immune response. Monocytes and macrophages play a central role in this hyperinflammatory response to SARS-CoV-2. NLRP3 inflammasome activation has been observed in monocytes of patients with COVID-19, but the mechanism and consequences of inflammasome activation require further investigation. In this study, we inoculated a macrophage-like THP-1 cell line, primary differentiated human nasal epithelial cell (hNEC) cultures, and primary monocytes with SARS-CoV-2. We found that the activation of the NLRP3 inflammasome in macrophages does not rely on viral replication, receptor-mediated entry, or actin-dependent entry. SARS-CoV-2 productively infected hNEC cultures without triggering the production of inflammasome cytokines IL-18 and IL-1β. Importantly, these cytokines did not inhibit viral replication in hNEC cultures. SARS-CoV-2 inoculation of primary monocytes led to inflammasome activation and induced a macrophage phenotype in these cells. Monocytic cells from bronchoalveolar lavage (BAL) fluid, but not from peripheral blood, of patients with COVID-19, showed evidence of inflammasome activation, expressed the proinflammatory marker CD11b, and displayed oxidative burst. These findings highlight the central role of activated macrophages, as a result of direct viral sensing, in COVID-19 and support the inhibition of IL-1β and IL-18 as potential therapeutic strategies to reduce immunopathology without increasing viral replication. IMPORTANCE Inflammasome activation is associated with severe COVID-19. The impact of inflammasome activation on viral replication and mechanistic details of this activation are not clarified. This study advances our understanding of the role of inflammasome activation in macrophages by identifying TLR2, NLRP3, ASC, and caspase-1 as dependent factors in this activation. Further, it highlights that SARS-CoV-2 inflammasome activation is not a feature of nasal epithelial cells but rather activation of bystander macrophages in the airway. Finally, we demonstrate that two pro inflammatory cytokines produced by inflammasome activation, IL-18 and IL-1β, do not restrict viral replication and are potential targets to ameliorate pathological inflammation in severe COVID-19.
Collapse
Affiliation(s)
- Leon L. Hsieh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Monika Looney
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexis Figueroa
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guido Massaccesi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Georgia Stavrakis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Eduardo U. Anaya
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Franco R. D'Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alvaro A. Ordonez
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew S. Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Victor R. DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Petros C. Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew H. Karaba
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L. Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Liddie JM, Bind MA, Karra M, Sunderland EM. County-level associations between drinking water PFAS contamination and COVID-19 mortality in the United States. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2024:10.1038/s41370-024-00723-5. [PMID: 39369072 DOI: 10.1038/s41370-024-00723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Epidemiologic and animal studies both support relationships between exposures to per- and polyfluoroalkyl substances (PFAS) and harmful effects on the immune system. Accordingly, PFAS have been identified as potential environmental risk factors for adverse COVID-19 outcomes. OBJECTIVE Here, we examine associations between PFAS contamination of U.S. community water systems (CWS) and county-level COVID-19 mortality records. Our analyses leverage two datasets: one at the subnational scale (5371 CWS serving 621 counties) and one at the national scale (4798 CWS serving 1677 counties). The subnational monitoring dataset was obtained from statewide drinking monitoring of PFAS (2016-2020) and the national monitoring dataset was obtained from a survey of unregulated contaminants (2013-2015). METHODS We conducted parallel analyses using multilevel quasi-Poisson regressions to estimate cumulative incidence ratios for the association between county-level measures of PFAS drinking water contamination and COVID-19 mortality prior to vaccination onset (Jan-Dec 2020). In the primary analyses, these regressions were adjusted for several county-level sociodemographic factors, days after the first reported case in the county, and total hospital beds. RESULTS In the subnational analysis, detection of at least one PFAS over 5 ng/L was associated with 12% higher [95% CI: 4%, 19%] COVID-19 mortality. In the national analysis, detection of at least one PFAS above the reporting limits (20-90 ng/L) was associated with 13% higher [95% CI: 8%, 19%] COVID-19 mortality. IMPACT STATEMENT Our findings provide evidence for an association between area-level drinking water PFAS contamination and higher COVID-19 mortality in the United States. These findings reinforce the importance of ongoing state and federal monitoring efforts supporting the U.S. Environmental Protection Agency's 2024 drinking water regulations for PFAS. More broadly, this example suggests that drinking water quality could play a role in infectious disease severity. Future research would benefit from study designs that combine area-level exposure measures with individual-level outcome data.
Collapse
Affiliation(s)
- Jahred M Liddie
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Marie-Abèle Bind
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mahesh Karra
- Frederick S. Pardee School of Global Studies, Boston University, Boston, MA, USA
| | - Elsie M Sunderland
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| |
Collapse
|
3
|
Das A, Wang X, Devonshire K, Lyons EJ, Popescu I, Zhou Z, Li J, Sembrat J, Pilewski J, Zou C, Alder JK, Chen BB, Snyder ME, McDyer JF. IL-10 Is Critical for Regulation of Cytotoxic CD4+NKG7+ T Cells in Lung Allograft Rejection but Is Not Required for Allograft Acceptance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:898-905. [PMID: 39072690 DOI: 10.4049/jimmunol.2400279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
Lung transplant remains the primary therapeutic option for patients with end-stage lung disease, but long-term survival rates remain suboptimal compared with other solid organ transplants. Acute cellular rejection (ACR) is a significant challenge in lung transplant recipients, with T cell-mediated mechanisms playing a major role. IL-10 is known for its immunoregulatory function, although its specific role in lung allograft rejection remains unclear. Using the mouse orthotopic lung transplant model, we investigated the role of IL-10 in regulating alloeffector T cell responses. Unexpectedly, we found that IL-10 was not required for early costimulation blockade-induced allograft acceptance. However, IL-10 deficiency or blockade resulted in increased CD4+ T cell numbers, proliferation, graft infiltration, and alloeffector responses. In the absence of IL-10, CD4+ T cell responses predominated over CD8 responses during ACR in contrast to wild-type mice. Type 1 immunity (IFN-γ) responses along with elevated CD4+NKG7+ and CD4+CD107a+ responses predominated during ACR, highlighting a critical regulatory role for IL-10 in modulating CD4+ T cell alloimmune responses. We further demonstrated increased colocalization of NKG7 and CD107a in CD4+ T cells from IL-10-deficient allografts, suggesting coordination in cytotoxic activity. Together, our findings highlight a critical role for IL-10 in regulation of cytotoxic CD4+NKG7+ T cells, an effector population that needs further investigation to elucidate their role in lung allograft rejection.
Collapse
Affiliation(s)
- Antu Das
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xingan Wang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kaitlyn Devonshire
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Emily J Lyons
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Iulia Popescu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Zihe Zhou
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jingmei Li
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John Sembrat
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Joseph Pilewski
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Chunbin Zou
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jonathan K Alder
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Bill B Chen
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Mark E Snyder
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John F McDyer
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
4
|
Thomas SM, Ankley LM, Conner KN, Rapp AW, McGee AP, LeSage F, Tanner CD, Vielma TE, Scheeres EC, Obar JJ, Olive AJ. TGFβ primes alveolar-like macrophages to induce type I IFN following TLR2 activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611226. [PMID: 39282428 PMCID: PMC11398362 DOI: 10.1101/2024.09.04.611226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Alveolar macrophages (AMs) are key mediators of lung function and are potential targets for therapies during respiratory infections. TGFβ is an important regulator of AM differentiation and maintenance, but how TGFβ directly modulates the innate immune responses of AMs remains unclear. This shortcoming prevents effective targeting of AMs to improve lung function in health and disease. Here we leveraged an optimized ex vivo AM model system, fetal-liver derived alveolar-like macrophages (FLAMs), to dissect the role of TGFβ in AMs. Using transcriptional analysis, we first globally defined how TGFβ regulates gene expression of resting FLAMs. We found that TGFβ maintains the baseline metabolic state of AMs by driving lipid metabolism through oxidative phosphorylation and restricting inflammation. To better understand inflammatory regulation in FLAMs, we next directly tested how TGFβ alters the response to TLR2 agonists. While both TGFβ (+) and TGFβ (-) FLAMs robustly responded to TLR2 agonists, we found an unexpected activation of type I interferon (IFN) responses in FLAMs and primary AMs in a TGFβ-dependent manner. Surprisingly, mitochondrial antiviral signaling protein and the interferon regulator factors 3 and 7 were required for IFN production by TLR2 agonists. Together, these data suggest that TGFβ modulates AM metabolic networks and innate immune signaling cascades to control inflammatory pathways in AMs.
Collapse
Affiliation(s)
- Sean M. Thomas
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Laurisa M. Ankley
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Kayla N. Conner
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Alexander W. Rapp
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Abigail P. McGee
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Francois LeSage
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Christopher D. Tanner
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Taryn E. Vielma
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Eleanor C. Scheeres
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Joshua J. Obar
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Andrew J. Olive
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
5
|
Mondell E, Nino G, Hong X, Wang X, Gutierrez MJ. Immune Biomarkers at Birth Predict Lower Respiratory Tract Infection Risk in a Large Birth Cohort. Pathogens 2024; 13:765. [PMID: 39338956 PMCID: PMC11435078 DOI: 10.3390/pathogens13090765] [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: 07/16/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Lower respiratory tract infections (LRTIs) remain the leading cause of infant morbidity and mortality worldwide and affect long-term respiratory health. Identifying immunological determinants of LRTI susceptibility may help stratify disease risk and identify therapies. This study aimed to identify neonatal immunological factors predicting LRTI risk in infancy. Cord blood plasma from 191 neonates from the Boston Birth Cohort was analyzed for 28 soluble immune factors. LRTI was defined as bronchiolitis, bronchitis, or pneumonia during the first year of life. Welch's t-test demonstrated significantly higher log10 transformed concentrations of IL-17 and IFNγ in the LRTI group compared to neonates without LRTI in the first year of life (p < 0.05). Risk associations were determined using multivariate survival models. There were 29 infants with LRTIs. High cord blood levels of IFNγ (aHR = 2.35, 95% CI 1.07-5.17), TNF-β (aHR = 2.86, 95% CI 1.27-6.47), MIP-1α (aHR = 2.82, 95% CI 1.22-6.51), and MIP-1β (aHR = 2.34, 95% CI 1.05-5.20) were associated with a higher risk of LRTIs. RANTES was associated with a lower risk (aHR = 0.43, 95% CI 0.19-0.97). Soluble immune factors linked to antiviral immunity (IFNγ) and cytokines mediating inflammatory responses (TNF-β), and cell homing (MIP-1α/b), at birth were associated with an increased risk of LRTIs during infancy.
Collapse
Affiliation(s)
- Ethan Mondell
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Gustavo Nino
- Division of Pulmonary and Sleep Medicine, Children’s National Hospital, George Washington University, Washington, DC 20010, USA;
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC 20010, USA
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (X.H.); (X.W.)
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (X.H.); (X.W.)
- Division of General Pediatrics and Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Maria J. Gutierrez
- Division of Pediatric Allergy, Immunology and Rheumatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Sudduth ER, López Ruiz A, Trautmann-Rodriguez M, Fromen CA. Age-dependent changes in phagocytic activity: in vivo response of mouse pulmonary antigen presenting cells to direct lung delivery of charged PEGDA nanoparticles. J Nanobiotechnology 2024; 22:476. [PMID: 39135064 PMCID: PMC11318229 DOI: 10.1186/s12951-024-02743-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Current needle-based vaccination for respiratory viruses is ineffective at producing sufficient, long-lasting local immunity in the elderly. Direct pulmonary delivery to the resident local pulmonary immune cells can create long-term mucosal responses. However, criteria for drug vehicle design rules that can overcome age-specific changes in immune cell functions have yet to be established. RESULTS Here, in vivo charge-based nanoparticle (NP) uptake was compared in mice of two age groups (2- and 16-months) within the four notable pulmonary antigen presenting cell (APC) populations: alveolar macrophages (AM), interstitial macrophages (IM), CD103+ dendritic cells (DCs), and CD11b+ DCs. Both macrophage populations exhibited preferential uptake of anionic nanoparticles but showed inverse rates of phagocytosis between the AM and IM populations across age. DC populations demonstrated preferential uptake of cationic nanoparticles, which remarkably did not significantly change in the aged group. Further characterization of cell phenotypes post-NP internalization demonstrated unique surface marker expression and activation levels for each APC population, showcasing heightened DC inflammatory response to NP delivery in the aged group. CONCLUSION The age of mice demonstrated significant preferences in the charge-based NP uptake in APCs that differed greatly between macrophages and DCs. Carefully balance of the targeting and activation of specific types of pulmonary APCs will be critical to produce efficient, age-based vaccines for the growing elderly population.
Collapse
Affiliation(s)
- Emma R Sudduth
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA
| | - Aida López Ruiz
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA
| | - Michael Trautmann-Rodriguez
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA.
| |
Collapse
|
7
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
8
|
Sasidharan A, Grosche A, Xu X, Kinane TB, Angoli D, Vidyasagar S. Select amino acids recover cytokine-altered ENaC function in human bronchial epithelial cells. PLoS One 2024; 19:e0307809. [PMID: 39052685 PMCID: PMC11271875 DOI: 10.1371/journal.pone.0307809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
The airway epithelium plays a pivotal role in regulating mucosal immunity and inflammation. Epithelial barrier function, homeostasis of luminal fluid, and mucociliary clearance are major components of mucosal defense mechanisms. The epithelial sodium channel (ENaC) is one of the key players in controlling airway fluid volume and composition, and characteristic cytokines cause ENaC and barrier dysfunctions following pulmonary infections or allergic reactions. Given the limited understanding of the requisite duration and magnitude of cytokines to affect ENaC and barrier function, available treatment options for restoring normal ENaC activity are limited. Previous studies have demonstrated that distinct amino acids can modulate epithelial ion channel activities and barrier function in intestines and airways. Here, we have investigated the time- and concentration-dependent effect of representative cytokines for Th1- (IFN-γ and TNF-α), Th2- (IL-4 and IL-13), and Treg-mediated (TGF-β1) immune responses on ENaC activity and barrier function in human bronchial epithelial cells. When cells were exposed to Th1 and Treg cytokines, ENaC activity decreased gradually while barrier function remained largely unaffected. In contrast, Th2 cytokines had an immediate and profound inhibitory effect on ENaC activity that was subsequently followed by epithelial barrier disruption. These functional changes were associated with decreased membrane protein expression of α-, β-, and γ-ENaC, and decreased mRNA levels of β- and γ-ENaC. A proprietary blend of amino acids was developed based on their ability to prevent Th2 cytokine-induced ENaC dysfunction. Exposure to the select amino acids reversed the inhibitory effect of IL-13 on ENaC activity by increasing mRNA levels of β- and γ-ENaC, and protein expression of γ-ENaC. This study indicates the beneficial effect of select amino acids on ENaC activity in an in vitro setting of Th2-mediated inflammation suggesting these amino acids as a novel therapeutic approach for correcting this condition.
Collapse
Affiliation(s)
- Anusree Sasidharan
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Astrid Grosche
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Xiaodong Xu
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - T. Bernard Kinane
- Pediatric Pulmonary Division, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Damiano Angoli
- Pediatric Pulmonary Division, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
9
|
Iqbal H, Rhee DK. Intranasal Immunization of Pneumococcal pep27 Mutant Attenuates Allergic and Inflammatory Diseases by Upregulating Skin and Mucosal Tregs. Vaccines (Basel) 2024; 12:737. [PMID: 39066375 PMCID: PMC11281725 DOI: 10.3390/vaccines12070737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Conventional immunization methods such as intramuscular injections lack effective mucosal protection against pathogens that enter through the mucosal surfaces. Moreover, conventional therapy often leads to adverse events and compromised immunity, followed by complicated outcomes, leading to the need to switch to other options. Thus, a need to develop safe and effective treatment with long-term beneficial outcomes to reduce the risk of relapse is mandatory. Mucosal vaccines administered across mucosal surfaces, such as the respiratory or intestinal mucosa, to prompt robust localized and systemic immune responses to prevent the public from acquiring pathogenic diseases. Mucosal immunity contains a unique immune cell milieu that selectively identify pathogens and limits the transmission and progression of mucosal diseases, such as allergic dermatitis and inflammatory bowel disease (IBD). It also offers protection from localized infection at the site of entry, enables the clearance of pathogens on mucosal surfaces, and leads to the induction of long-term immunity with the ability to shape regulatory responses. Regulatory T (Treg) cells have been a promising strategy to suppress mucosal diseases. To find advances in mucosal treatment, we investigated the therapeutic effects of intranasal pep27 mutant immunization. Nasal immunization protects mucosal surfaces, but nasal antigen presentation appears to entail the need for an adjuvant to stimulate immunogenicity. Here, a novel method is developed to induce Tregs via intranasal immunization without an adjuvant to potentially overcome allergic diseases and gut and lung inflammation using lung-gut axis communication in animal models. The implementation of the pep27 mutant for these therapies should be preceded by studies on Treg resilience through clinical translational studies on dietary changes.
Collapse
Affiliation(s)
- Hamid Iqbal
- Department of Pharmacy, CECOS University, Peshawar 25000, Pakistan;
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
10
|
Foster R, Veras MM, Bachi ALL, do Amaral JB, Yariwake VY, Waked D, Rodrigues ACB, Farrajota M, Pires RP, Pantaleão K, dos Santos JDMB, Damian FH, Saldiva PH, Vaisberg MW. Inflammatory Status in Trained and Untrained Mice at Different Pollution Levels. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:821. [PMID: 39063399 PMCID: PMC11276537 DOI: 10.3390/ijerph21070821] [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: 05/01/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024]
Abstract
Atmospheric pollution can be defined as a set of changes that occur in the composition of the air, making it unsuitable and/or harmful and thereby generating adverse effects on human health. The regular practice of physical exercise (PE) is associated with the preservation and/or improvement of health; however, it can be influenced by neuroimmunoendocrine mechanisms and external factors such as air pollution, highlighting the need for studies involving the practice of PE in polluted environments. Herein, 24 male C57BL/6 mice were evaluated, distributed into four groups (exposed to a high concentration of pollutants/sedentary, exposed to a high concentration of pollutants/exercised, exposed to ambient air/sedentary, and exposed to ambient air/exercised). The exposure to pollutants occurred in the environmental particle concentrator (CPA) and the physical training was performed on a treadmill specially designed for use within the CPA. Pro- and anti-inflammatory markers in blood and bronchoalveolar lavage (BALF), BALF cellularity, and lung tissue were evaluated. Although the active group exposed to a high concentration of pollution showed a greater inflammatory response, both the correlation analysis and the ratio between pro- and anti-inflammatory cytokines demonstrated that the exercised group presented greater anti-inflammatory activity, suggesting a protective/adaptative effect of exercise when carried out in a polluted environment.
Collapse
Affiliation(s)
- Roberta Foster
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| | - Mariana Matera Veras
- Experimental Atmospheric Pollution Laboratory, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (M.M.V.); (V.Y.Y.); (D.W.); (A.C.B.R.)
| | - Andre Luis Lacerda Bachi
- Post-Graduation Program in Health Science, University of Santo Amaro, São Paulo 04743-030, Brazil
| | - Jonatas Bussador do Amaral
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| | - Victor Yuji Yariwake
- Experimental Atmospheric Pollution Laboratory, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (M.M.V.); (V.Y.Y.); (D.W.); (A.C.B.R.)
| | - Dunia Waked
- Experimental Atmospheric Pollution Laboratory, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (M.M.V.); (V.Y.Y.); (D.W.); (A.C.B.R.)
| | - Ana Clara Bastos Rodrigues
- Experimental Atmospheric Pollution Laboratory, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (M.M.V.); (V.Y.Y.); (D.W.); (A.C.B.R.)
| | - Marilia Farrajota
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| | - Robério Pereira Pires
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| | - Karina Pantaleão
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| | | | - Francys Helen Damian
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| | - Paulo Hilário Saldiva
- Experimental Atmospheric Pollution Laboratory, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (M.M.V.); (V.Y.Y.); (D.W.); (A.C.B.R.)
| | - Mauro Walter Vaisberg
- Department of Otorhinolaryngology and Head and Neck Surgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo 04021-001, Brazil; (R.F.); (J.B.d.A.); (M.F.); (R.P.P.); (K.P.); (M.W.V.)
| |
Collapse
|
11
|
M Yusoff NNF, Ahmad S, Wan Abdul Rahman WF, Mohamud R, C Boer J, Plebanski M, Abdullah B, Chen X, Tengku Din TADAA. CD4+ Foxp3+ Regulatory T-cells in Modulating Inflammatory Microenvironment in Chronic Rhinosinusitis with Nasal Polyps: Progress and Future Prospect. Cytokine 2024; 178:156557. [PMID: 38452440 DOI: 10.1016/j.cyto.2024.156557] [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: 11/15/2023] [Revised: 01/26/2024] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a subtype of chronic rhinosinusitis (CRS) characterized by the presence of nasal polyps (NP) in the paranasal mucosa. Despite the complex etiology, NP is believed to result from chronic inflammation. The long-term aftermath of the type 2 response is responsible for symptoms seen in NP patients, i.e. rhinorrhea, hyposmia, and nasal obstruction. Immune cellular tolerogenic mechanisms, particularly CD4 + Foxp3 + regulatory T cells (Tregs), are crucial to curtail inflammatory responses. Current evidence suggests impaired Treg activity is the main reason underlying the compromise of self-tolerance, contributing to the onset of CRSwNP. There is compelling evidence that tumor necrosis factor 2 (TNFR2) is preferentially expressed by Tregs, and TNFR2 is able to identify the most potent suppressive subset of Tregs. Tumor necrosis factor (TNF)-TNFR2 interaction plays a decisive role in the activation and expansion of Tregs. This review summarizes current understanding of Tregs biology, focusing on the discussion of the recent advances in the study of TNF-TNFR2 axis in the upregulation of Treg function as a negative feedback mechanism in the control of chronic inflammation. The role of dysregulation of Tregs in the immunopathogenesis of CRSwNP will be analyzed. The future perspective on the harnessing Tregs-mediated self-tolerant mechanism in the management of CRSwNP will be introduced.
Collapse
Affiliation(s)
- Nur Najwa Farahin M Yusoff
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | | | - Rohimah Mohamud
- 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, Victoria 3083, Australia
| | - Magdalena Plebanski
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Baharudin Abdullah
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | | |
Collapse
|
12
|
Macchione M, Yoshizaki K, Frias DP, Maier K, Smelan J, Prado CM, Mauad T. Fragrances as a trigger of immune responses in different environments. Toxicol In Vitro 2024; 96:105769. [PMID: 38142785 DOI: 10.1016/j.tiv.2023.105769] [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: 08/03/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Fragrances can cause allergic skin reactions, expressed as allergic contact dermatitis and reactions in the respiratory tract that range from acute temporary upper airway irritation to obstructive lung disease. These adverse health effects may result from the stimulation of a specific (adaptive) immune response. Th1 cells, which essentially produce interleukin-2 (IL-2) and interferon-γ (IFN-γ), play a key role in allergic contact dermatitis and also on allergic sensitization to common allergens (e.g., nickel and fragrance). It has been shown that fragrance allergy leads to Th2/Th22 production of IL-4, IL-5 and IL-13, controlling the development of IgE and mediating hypersensitivity reactions in the lung, such as asthma. Cytokines released during immune response modulate the expression of cytochrome P450 (CYPs) proteins, which can result in alterations of the pharmacological effects of substances in inflammatory diseases. The mechanisms linking environment and immunity are still not completely understood but it is known that aryl hydrocarbon receptor (AhR) is a sensor with conserved ligand-activated transcription factor, highly expressed in cells that controls complex transcriptional programs which are ligand and cell type specific, with CYPs as targeted genes. This review focuses on these important aspects of immune responses of the skin and respiratory tract cells, describing some in vitro models applied to evaluate the mechanisms involved in fragrance-induced allergy.
Collapse
Affiliation(s)
- M Macchione
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil.
| | - K Yoshizaki
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - D P Frias
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - K Maier
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - J Smelan
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - C M Prado
- Federal University of Sao Paulo, Santos, Brazil
| | - T Mauad
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| |
Collapse
|
13
|
He J, Li J, Lin Q, Ni H, Huang S, Cheng H, Ding X, Huang Y, Yu H, Xu Y, Nie H. Anti-CD20 treatment attenuates Th2 cell responses: implications for the role of lung follicular mature B cells in the asthmatic mice. Inflamm Res 2024; 73:433-446. [PMID: 38345634 DOI: 10.1007/s00011-023-01847-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND B cells were believed to act as antigen-presenting cells (APCs) to promote T helper type 2 (Th2) cell responses. However, the role of lung B cells and its subpopulations in Th2 cell responses in asthma remains unclear. OBJECTIVE We leveraged an anti-CD20 monoclonal antibody (mAb) treatment that has been shown to selectively deplete B cells in mice and investigated whether this treatment modulates Th2 cell responses and this modulation is related to lung follicular mature (FM) B cells in a murine model of asthma. METHODS AND RESULTS We used a house dust mite (HDM)-induced asthma mouse model and found that anti-CD20 mAb treatment attenuates Th2 cell responses. Meanwhile, anti-CD20 mAb treatment did dramatically reduce the number of B cells, especially FM B cells in the lungs, but did not impact the frequency of other immune cell types, including lung T cells, dendritic cells, natural killer cells, and regulatory T cells in wild-type mice. Moreover, we found that the suppressive effect of anti-CD20 mAb treatment on Th2 cell responses could be reversed upon adoptive transfer of lung FM B cells, but not lung CD19+ B cells without FM B cells in asthmatic mice. CONCLUSIONS These findings reveal that anti-CD20 mAb treatment alleviates Th2 cell responses, possibly by depleting lung FM B cells in a Th2-driven asthma model. This implies a potential therapeutic approach for asthma treatment through the targeting of lung FM B cells.
Collapse
Affiliation(s)
- Jilong He
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Jingling Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Qibin Lin
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Haiyang Ni
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Sisi Huang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Hong Cheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Xuhong Ding
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Yi Huang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Hongying Yu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Yaqing Xu
- Department of Geriatric Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
14
|
Russo S, Kwiatkowski M, Wolters JC, Gerding A, Hermans J, Govorukhina N, Bischoff R, Melgert BN. Effects of lysine deacetylase inhibitor treatment on LPS responses of alveolar-like macrophages. J Leukoc Biol 2024; 115:435-449. [PMID: 37811856 DOI: 10.1093/jleuko/qiad121] [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: 05/26/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages are key immune cells that can adapt their metabolic phenotype in response to different stimuli. Lysine deacetylases are important enzymes regulating inflammatory gene expression and lysine deacetylase inhibitors have been shown to exert anti-inflammatory effects in models of chronic obstructive pulmonary disease. We hypothesized that these anti-inflammatory effects may be associated with metabolic changes in macrophages. To validate this hypothesis, we used an unbiased and a targeted proteomic approach to investigate metabolic enzymes, as well as liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry, to quantify metabolites in combination with the measurement of functional parameters in primary murine alveolar-like macrophages after lipopolysaccharide-induced activation in the presence or absence of lysine deacetylase inhibition. We found that lysine deacetylase inhibition resulted in reduced production of inflammatory mediators such as tumor necrosis factor α and interleukin 1β. However, only minor changes in macrophage metabolism were observed, as only one of the lysine deacetylase inhibitors slightly increased mitochondrial respiration while no changes in metabolite levels were seen. However, lysine deacetylase inhibition specifically enhanced expression of proteins involved in ubiquitination, which may be a driver of the anti-inflammatory effects of lysine deacetylase inhibitors. Our data illustrate that a multiomics approach provides novel insights into how macrophages interact with cues from their environment. More detailed studies investigating ubiquitination as a potential driver of lysine deacetylase inhibition will help developing novel anti-inflammatory drugs for difficult-to-treat diseases such as chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Sara Russo
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Marcel Kwiatkowski
- Functional Proteo-Metabolomics, Department of Biochemistry, University of Innsbruck, Innrain 80-82, Innsbruck 6020, Austria
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Albert Gerding
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Jos Hermans
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Natalia Govorukhina
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Barbro N Melgert
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| |
Collapse
|
15
|
Yi P, Yu W, Xiong Y, Dong Y, Huang Q, Lin Y, Du Y, Hua F. IL-35: New Target for Immunotherapy Targeting the Tumor Microenvironment. Mol Cancer Ther 2024; 23:148-158. [PMID: 37988561 DOI: 10.1158/1535-7163.mct-23-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/15/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023]
Abstract
Interleukin 35(IL-35) is a newly discovered inhibitory cytokine of the IL12 family. More recently, IL-35 was found to be increased in the tumor microenvironment (TME) and peripheral blood of many patients with cancer, indicating that it plays an important role in the TME. Tumors secrete cytokines that recruit myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) into the TME to promote malignant progression, which is a great challenge for cancer treatment. Radiotherapy causes serious adverse effects, and tumor resistance to immune checkpoint inhibitors is still an unsolved challenge. Thus, new cancer therapy approaches are urgently needed. Numerous studies have shown that IL-35 can recruit immunosuppressive cells to enable tumor immune escape by promoting the conversion of immune cells into a tumor growth-promoting phenotype as well as facilitating tumor angiogenesis. IL-35-neutralizing antibodies were found to boost the chemotherapeutic effect of gemcitabine and considerably reduce the microvascular density of pancreatic cancer in mice. Therefore, targeting IL-35 in the TME provides a promising cancer treatment target. In addition, IL-35 may be used as an independent prognostic factor for some tumors in the near future. This review intends to reveal the interplay of IL-35 with immune cells in the TME, which may provide new options for the treatment of cancer.
Collapse
Affiliation(s)
- Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Wenjun Yu
- Fuzhou First People's Hospital of Jiangxi Province, Fuzhou City, Jiangxi Province, P.R. China
| | - Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yao Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Qiang Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yunfei Du
- Department of Anesthesiology, Nanchang Central Hospital, Nanchang, Jiangxi, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| |
Collapse
|
16
|
Cao M, Wang Z, Lan W, Xiang B, Liao W, Zhou J, Liu X, Wang Y, Zhang S, Lu S, Lang J, Zhao Y. The roles of tissue resident macrophages in health and cancer. Exp Hematol Oncol 2024; 13:3. [PMID: 38229178 PMCID: PMC10790434 DOI: 10.1186/s40164-023-00469-0] [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: 10/08/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
As integral components of the immune microenvironment, tissue resident macrophages (TRMs) represent a self-renewing and long-lived cell population that plays crucial roles in maintaining homeostasis, promoting tissue remodeling after damage, defending against inflammation and even orchestrating cancer progression. However, the exact functions and roles of TRMs in cancer are not yet well understood. TRMs exhibit either pro-tumorigenic or anti-tumorigenic effects by engaging in phagocytosis and secreting diverse cytokines, chemokines, and growth factors to modulate the adaptive immune system. The life-span, turnover kinetics and monocyte replenishment of TRMs vary among different organs, adding to the complexity and controversial findings in TRMs studies. Considering the complexity of tissue associated macrophage origin, macrophages targeting strategy of each ontogeny should be carefully evaluated. Consequently, acquiring a comprehensive understanding of TRMs' origin, function, homeostasis, characteristics, and their roles in cancer for each specific organ holds significant research value. In this review, we aim to provide an outline of homeostasis and characteristics of resident macrophages in the lung, liver, brain, skin and intestinal, as well as their roles in modulating primary and metastatic cancer, which may inform and serve the future design of targeted therapies.
Collapse
Affiliation(s)
- Minmin Cao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zihao Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wanying Lan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- Guixi Community Health Center of the Chengdu High-Tech Zone, Chengdu, China
| | - Binghua Xiang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjun Liao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaomeng Liu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yiling Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shichuan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shun Lu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Zhao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
17
|
Barbier E, Carpentier J, Simonin O, Gosset P, Platel A, Happillon M, Alleman LY, Perdrix E, Riffault V, Chassat T, Lo Guidice JM, Anthérieu S, Garçon G. Oxidative stress and inflammation induced by air pollution-derived PM 2.5 persist in the lungs of mice after cessation of their sub-chronic exposure. ENVIRONMENT INTERNATIONAL 2023; 181:108248. [PMID: 37857188 DOI: 10.1016/j.envint.2023.108248] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/21/2023]
Abstract
More than 7 million early deaths/year are attributable to air pollution. Current health concerns are especially focused on air pollution-derived particulate matter (PM). Although oxidative stress-induced airway inflammation is one of the main adverse outcome pathways triggered by air pollution-derived PM, the persistence of both these underlying mechanisms, even after exposure cessation, remained poorly studied. In this study, A/JOlaHsd mice were also exposed acutely (24 h) or sub-chronically (4 weeks), with or without a recovery period (12 weeks), to two urban PM2.5 samples collected during contrasting seasons (i.e., autumn/winter, AW or spring/summer, SS). The distinct intrinsic oxidative potentials (OPs) of AW and SS PM2.5, as evaluated in acellular conditions, were closely related to their respective physicochemical characteristics and their respective ability to really generate ROS over-production in the mouse lungs. Despite the early activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) cell signaling pathway by AW and, in a lesser degree, SS PM2.5, in the murine lungs after acute and sub-chronic exposures, the critical redox homeostasis was not restored, even after the exposure cessation. Accordingly, an inflammatory response was reported through the activation of the nuclear factor-kappa B (NF-κB) cell signaling pathway activation, the secretion of cytokines, and the recruitment of inflammatory cells, in the murine lungs after the acute and sub-chronic exposures to AW and, in a lesser extent, to SS PM2.5, which persisted after the recovery period. Taken together, these original results provided, for the first time, new relevant insights that air pollution-derived PM2.5, with relatively high intrinsic OPs, induced oxidative stress and inflammation, which persisted admittedly at a lower level in the lungs after the exposure cessation, thereby contributing to the occurrence of molecular and cellular adverse events leading to the development and/or exacerbation of future chronic inflammatory lung diseases and even cancers.
Collapse
Affiliation(s)
- Emeline Barbier
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR4483-IMPECS, France
| | - Jessica Carpentier
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR4483-IMPECS, France
| | - Ophélie Simonin
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR4483-IMPECS, France
| | - Pierre Gosset
- Service d'Anatomo-pathologie, Hôpital Saint Vincent de Paul, Lille, France
| | - Anne Platel
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR4483-IMPECS, France
| | - Mélanie Happillon
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR4483-IMPECS, France
| | - Laurent Y Alleman
- IMT Nord Europe, Institut Mines-Télécom, Univ. Lille, Centre for Energy and Environment, Lille, France
| | - Esperanza Perdrix
- IMT Nord Europe, Institut Mines-Télécom, Univ. Lille, Centre for Energy and Environment, Lille, France
| | - Véronique Riffault
- IMT Nord Europe, Institut Mines-Télécom, Univ. Lille, Centre for Energy and Environment, Lille, France
| | - Thierry Chassat
- Institut Pasteur de Lille, Plateforme d'Expérimentation et de Haute Technologie Animale, Lille, France
| | | | | | - Guillaume Garçon
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR4483-IMPECS, France.
| |
Collapse
|
18
|
Elean M, Raya Tonetti F, Fukuyama K, Arellano-Arriagada L, Namai F, Suda Y, Gobbato N, Nishiyama K, Villena J, Kitazawa H. Immunobiotic Ligilactobacillus salivarius FFIG58 Confers Long-Term Protection against Streptococcus pneumoniae. Int J Mol Sci 2023; 24:15773. [PMID: 37958756 PMCID: PMC10648150 DOI: 10.3390/ijms242115773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Previously, we isolated potentially probiotic Ligilactobacillus salivarius strains from the intestines of wakame-fed pigs. The strains were characterized based on their ability to modulate the innate immune responses triggered by the activation of Toll-like receptor (TLR)-3 or TLR4 signaling pathways in intestinal mucosa. In this work, we aimed to evaluate whether nasally administered L. salivarius strains are capable of modulating the innate immune response in the respiratory tract and conferring long-term protection against the respiratory pathogen Streptococcus pneumoniae. Infant mice (3-weeks-old) were nasally primed with L. salivarius strains and then stimulated with the TLR3 agonist poly(I:C). Five or thirty days after the last poly(I:C) administration mice were infected with pneumococci. Among the strains evaluated, L. salivarius FFIG58 had a remarkable ability to enhance the protection against the secondary pneumococcal infection by modulating the respiratory immune response. L. salivarius FFIG58 improved the ability of alveolar macrophages to produce interleukin (IL)-6, interferon (IFN)-γ, IFN-β, tumor necrosis factor (TNF)-α, IL-27, chemokine C-C motif ligand 2 (CCL2), chemokine C-X-C motif ligand 2 (CXCL2), and CXCL10 in response to pneumococcal challenge. Furthermore, results showed that the nasal priming of infant mice with the FFIG58 strain protected the animals against secondary infection until 30 days after stimulation with poly(I:C), raising the possibility of using nasally administered immunobiotics to stimulate trained immunity in the respiratory tract.
Collapse
Affiliation(s)
- Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (F.R.T.); (L.A.-A.)
| | - Fernanda Raya Tonetti
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (F.R.T.); (L.A.-A.)
| | - Kohtaro Fukuyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Luciano Arellano-Arriagada
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (F.R.T.); (L.A.-A.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (F.N.); (K.N.)
| | - Fu Namai
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 980-8572, Japan;
| | - Nadia Gobbato
- Laboratory of Immunology, Microbiology Institute, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, Tucuman 4000, Argentina;
| | - Keita Nishiyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (F.R.T.); (L.A.-A.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (F.N.); (K.N.)
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| |
Collapse
|
19
|
Almeida BR, Barros BCSC, Barros DTL, Orikaza CM, Suzuki E. Paracoccidioides brasiliensis Induces α3 Integrin Lysosomal Degradation in Lung Epithelial Cells. J Fungi (Basel) 2023; 9:912. [PMID: 37755020 PMCID: PMC10532483 DOI: 10.3390/jof9090912] [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: 08/14/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Studies on the pathogen-host interaction are crucial for the understanding of the mechanisms involved in the establishment, maintenance, and spread of infection. In recent years, our research group has observed that the P. brasiliensis species interact with integrin family receptors and increase the expression of α3 integrin in lung epithelial cells within 5 h of infection. Interestingly, α3 integrin levels were reduced by approximately 99% after 24 h of infection with P. brasiliensis compared to non-infected cells. In this work, we show that, during infection with this fungus, α3 integrin is increased in the late endosomes of A549 lung epithelial cells. We also observed that the inhibitor of the lysosomal activity bafilomycin A1 was able to inhibit the decrease in α3 integrin levels. In addition, the silencing of the charged multivesicular body protein 3 (CHMP3) inhibited the reduction in α3 integrin levels induced by P. brasiliensis in A549 cells. Thus, together, these results indicate that this fungus induces the degradation of α3 integrin in A549 lung epithelial cells by hijacking the host cell endolysosomal pathway.
Collapse
Affiliation(s)
| | | | | | | | - Erika Suzuki
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Ed. Antonio C. M. Paiva, São Paulo 04023-062, SP, Brazil; (B.R.A.)
| |
Collapse
|
20
|
Dentice Maidana S, Imamura Y, Elean M, Albarracín L, Nishiyama K, Suda Y, Kurata S, Jure MÁ, Kitazawa H, Villena J. Oral Administration of Lacticaseibacillus rhamnosus CRL1505 Modulates Lung Innate Immune Response against Klebsiella pneumoniae ST25. Microorganisms 2023; 11:1148. [PMID: 37317122 DOI: 10.3390/microorganisms11051148] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
Orally administered Lacticaseibacillus rhamnosus CRL1505 enhances respiratory immunity, providing protection against respiratory viruses and Streptococcus pneumoniae. However, the capacity of the CRL1505 strain to improve respiratory immunity against Gram-negative bacterial infections has not been evaluated before. The aim of this work was to evaluate whether the Lcb. rhamnosus CRL1505 was able to beneficially regulate the respiratory innate immune response and enhance the resistance to hypermucoviscous KPC-2-producing Klebsiella pneumoniae of the sequence type 25 (ST25). BALB/c mice were treated with the CRL1505 strain via the oral route and then nasally challenged with K. pneumoniae ST25 strains LABACER 01 or LABACER 27. Bacterial cell counts, lung injuries and the respiratory and systemic innate immune responses were evaluated after the bacterial infection. The results showed that K. pneumoniae ST25 strains increased the levels of TNF-α, IL-1β, IL-6, IFN-γ, IL-17, KC and MPC-1 in the respiratory tract and blood, as well as the numbers of BAL neutrophils and macrophages. Mice treated with Lcb. rhamnosus CRL1505 had significantly lower K. pneumoniae counts in their lungs, as well as reduced levels of inflammatory cells, cytokines and chemokines in the respiratory tract and blood when compared to infected controls. Furthermore, higher levels of the regulatory cytokines IL-10 and IL-27 were found in the respiratory tract and blood of CRL1505-treated mice than controls. These results suggest that the ability of Lcb. rhamnosus CRL1505 to help with the control of detrimental inflammation in lungs during K. pneumoniae infection would be a key feature to improve the resistance to this pathogen. Although further mechanistic studies are necessary, Lcb. rhamnosus CRL1505 can be proposed as a candidate to improve patients' protection against hypermucoviscous KPC-2-producing strains belonging to the ST25, which is endemic in the hospitals of our region.
Collapse
Affiliation(s)
- Stefania Dentice Maidana
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
- Laboratory of Antimicrobials, Institute of Microbiology "Luis C. Verna", Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, Tucuman 4000, Argentina
| | - Yoshiya Imamura
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
| | - Leonardo Albarracín
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
| | - Keita Nishiyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 980-8572, Japan
| | - Shoichiro Kurata
- Laboratory of Molecular Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - María Ángela Jure
- Laboratory of Antimicrobials, Institute of Microbiology "Luis C. Verna", Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, Tucuman 4000, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| |
Collapse
|
21
|
Kozik AJ, Begley LA, Lugogo N, Baptist A, Erb-Downward J, Opron K, Huang YJ. Airway microbiota and immune mediator relationships differ in obesity and asthma. J Allergy Clin Immunol 2023; 151:931-942. [PMID: 36572355 PMCID: PMC10566565 DOI: 10.1016/j.jaci.2022.11.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Asthma and obesity are both complex conditions characterized by chronic inflammation, and obesity-related severe asthma has been associated with differences in the microbiome. However, whether the airway microbiome and microbiota-immune response relationships differ between obese persons with or without nonsevere asthma is unestablished. OBJECTIVE We compared the airway microbiome and microbiota-immune mediator relationships between obese and nonobese subjects, with and without mild-moderate asthma. METHODS We performed cross-sectional analyses of the airway (induced sputum) microbiome and cytokine profiles from blood and sputum using 16S ribosomal RNA gene and internal transcribed spacer region sequencing to profile bacteria and fungi, and multiplex immunoassays. Analysis tools included QIIME 2, linear discriminant analysis effect size (aka LEfSe), Piphillin, and Sparse inverse covariance estimation for ecological association inference (aka SPIEC-EASI). RESULTS Obesity, irrespective of asthma status, was associated with significant differences in sputum bacterial community structure and composition (unweighted UniFrac permutational analysis of variance, P = .02), including a higher relative abundance of Prevotella, Gemella, and Streptococcus species. Among subjects with asthma, additional differences in sputum bacterial composition and fungal richness were identified between obese and nonobese individuals. Correlation network analyses demonstrated differences between obese and nonobese asthma in relationships between cytokine mediators, and these together with specific airway bacteria involving blood PAI-1, sputum IL-1β, GM-CSF, IL-8, TNF-α, and several Prevotella species. CONCLUSION Obesity itself is associated with an altered sputum microbiome, which further differs in those with mild-moderate asthma. The distinct differences in airway microbiota and immune marker relationships in obese asthma suggest potential involvement of airway microbes that may affect mechanisms or outcomes of obese asthma.
Collapse
Affiliation(s)
- Ariangela J Kozik
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Mich.
| | - Lesa A Begley
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Njira Lugogo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Alan Baptist
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Ann Arbor, Mich
| | - John Erb-Downward
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Kristopher Opron
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Yvonne J Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Mich; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
22
|
Putri DU, Huang CK, Ou TY, Lin CF, Lee MC, Hung CS, Lee CH. Persistent dysregulation of cellular immunity following COVID-19 recovery despite minimal post-COVID-19 sequelae manifestation. J Infect 2023; 86:486-488. [PMID: 36822411 PMCID: PMC9942484 DOI: 10.1016/j.jinf.2023.02.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023]
Affiliation(s)
- Denise Utami Putri
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan
| | - Chun-Kai Huang
- Department of Laboratory Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan
| | - Tsong-Yih Ou
- Division of Infectious Diseases, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan; Department of Nursing, Cardinal Tien College of Healthcare and Management, Taipei 231038, Taiwan
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan; Core Laboratory of Immune Monitoring, Office of Research and Development, Taipei Medical University, Taipei 110301, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan; International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Ming-Che Lee
- Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Sheng Hung
- Department of Laboratory Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan; School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chih-Hsin Lee
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| |
Collapse
|
23
|
Rebuli ME, Stanley Lee A, Nurhussien L, Tahir UA, Sun WY, Kimple AJ, Ebert CS, Almond M, Jaspers I, Rice MB. Nasal biomarkers of immune function differ based on smoking and respiratory disease status. Physiol Rep 2023; 11:e15528. [PMID: 36780897 PMCID: PMC9925276 DOI: 10.14814/phy2.15528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/04/2022] [Accepted: 11/13/2022] [Indexed: 02/15/2023] Open
Abstract
Respiratory biomarkers have the potential to identify airway injury by revealing inflammatory processes within the respiratory tract. Currently, there are no respiratory biomarkers suitable for clinical use to identify patients that warrant further diagnostic work-up, counseling, and treatment for toxic inhalant exposures or chronic airway disease. Using a novel, noninvasive method of sampling the nasal epithelial lining fluid, we aimed to investigate if nasal biomarker patterns could distinguish healthy nonsmoking adults from active smokers and those with chronic upper and lower airway disease in this exploratory study. We compared 28 immune mediators from healthy nonsmoking adults (n = 32), former smokers with COPD (n = 22), chronic rhinosinusitis (CRS) (n = 22), and smoking adults without airway disease (n = 13). Using ANOVA, multinomial logistic regressions, and weighted gene co-expression network analysis (WGCNA), we determined associations between immune mediators and each cohort. Six mediators (IL-7, IL-10, IL-13, IL-12p70, IL-15, and MCP-1) were lower among disease groups compared to healthy controls. Participants with lower levels of IL-10, IL-12p70, IL-13, and MCP-1 in the nasal fluid had a higher odds of being in the COPD or CRS group. The cluster analysis identified groups of mediators that correlated with disease status. Specifically, the cluster of IL-10, IL-12p70, and IL-13, was positively correlated with healthy and negatively correlated with COPD groups, and two clusters were correlated with active smoking. In this exploratory study, we preliminarily identified groups of nasal mucosal mediators that differed by airway disease and smoking status. Future prospective, age-matched studies that control for medication use are needed to validate these patterns and determine if nasosorption has diagnostic utility for upper and lower airway disease or injury.
Collapse
Affiliation(s)
- Meghan E. Rebuli
- Department of Pediatrics and Curriculum in Toxicology and Environmental MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Center for Environmental Medicine, Asthma and Lung BiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Anna Stanley Lee
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Lina Nurhussien
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Usman A. Tahir
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Wendy Y. Sun
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Adam J. Kimple
- Department of Otolaryngology‐Head and Neck SurgeryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Charles S. Ebert
- Department of Otolaryngology‐Head and Neck SurgeryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Martha Almond
- Center for Environmental Medicine, Asthma and Lung BiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Ilona Jaspers
- Department of Pediatrics and Curriculum in Toxicology and Environmental MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Center for Environmental Medicine, Asthma and Lung BiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Mary B. Rice
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
24
|
Zhou H, Qian Y, Liu J. MicroRNA-127 promotes anti-microbial ability in porcine alveolar macrophages via S1PR3/TLR signaling pathway. J Vet Sci 2023; 24:e20. [PMID: 37012029 PMCID: PMC10071279 DOI: 10.4142/jvs.22110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND As Actinobacillus pleuropneumonniae (APP) infection causes considerable losses in the pig industry, there is a growing need to develop effective therapeutic interventions that leverage host immune defense mechanisms to combat these pathogens. OBJECTIVES To demonstrate the role of microRNA (miR)-127 in controlling bacterial infection against APP. Moreover, to investigate a signaling pathway in macrophages that controls the production of anti-microbial peptides. METHODS Firstly, we evaluated the effect of miR-127 on APP-infected pigs by cell count/enzyme-linked immunosorbent assay (ELISA). Then the impact of miR-127 on immune cells was detected. The cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6 were evaluated by ELISA. The expression of cytokines (anti-microbial peptides [AMPs]) was assessed using quantitative polymerase chain reaction. The expression level of IL-6, TNF-α and p-P65 were analyzed by western blot. The expression of p65 in the immune cells was investigated by immunofluorescence. RESULTS miR-127 showed a protective effect on APP-infected macrophage. Moreover, the protective effect might depend on its regulation of macrophage bactericidal activity and the generation of IL-22, IL-17 and AMPs by targeting sphingosine-1-phosphate receptor3 (SIPR3), the element involved in the Toll-like receptor (TLR) cascades. CONCLUSIONS Together, we identify that miR-127 is a regulator of S1PR3 and then regulates TLR/nuclear factor-κB signaling in macrophages with anti-bacterial acticity, and it might be a potential target for treating inflammatory diseases caused by APP.
Collapse
Affiliation(s)
- Honglei Zhou
- School of Pet Science and Technology, Jiangsu Agri-animal Husbandry Vocational College Jiangsu 225300, China
| | - Yujia Qian
- Taizhou Jianyouda Pharma Co., LTD, Jiangsu 225300, China
| | - Jing Liu
- School of Pet Science and Technology, Jiangsu Agri-animal Husbandry Vocational College Jiangsu 225300, China
| |
Collapse
|
25
|
Yang Z, Roth K, Ding J, Kassotis CD, Mor G, Petriello MC. Exposure to a mixture of per-and polyfluoroalkyl substances modulates pulmonary expression of ACE2 and circulating hormones and cytokines. Toxicol Appl Pharmacol 2022; 456:116284. [PMID: 36270329 PMCID: PMC10325118 DOI: 10.1016/j.taap.2022.116284] [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: 07/05/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 01/01/2023]
Abstract
Genetic and environmental factors impact on the interindividual variability of susceptibility to communicable and non-communicable diseases. A class of ubiquitous chemicals, Per- and polyfluoroalkyl substances (PFAS) have been linked in epidemiological studies to immunosuppression and increased susceptibility to viral infections, but possible mechanisms are not well elucidated. To begin to gain insight into the role of PFAS in susceptibility to one such viral infection, Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), male and female C57BL/6 J mice were exposed to control water or a mixture of 5 PFAS (PFOS, PFOA, PFNA, PFHxS, Genx) for 12 weeks and lungs were isolated for examination of expression of SARS-CoV-2-related receptors Angiotensin-Converting Enzyme 2 (ACE2) and others. Secondary analyses included circulating hormones and cytokines which have been shown to directly or indirectly impact on ACE2 expression and severity of viral infections. Changes in mRNA and protein expression were analyzed by RT-qPCR and western blotting and circulating hormones and cytokines were determined by ELISA and MESO QuickPlex. The PFAS mixture decreased Ace2 mRNA 2.5-fold in male mice (p < 0.0001), with no significant change observed in females. In addition, TMPRSS2, ANPEP, ENPEP and DPP4 (other genes implicated in COVID-19 infection) were modulated due to PFAS. Plasma testosterone, but not estrogen were strikingly decreased due to PFAS which corresponded to PFAS-mediated repression of 4 representative pulmonary AR target genes; hemoglobin, beta adult major chain (Hbb-b1), Ferrochelatase (Fech), Collagen Type XIV Alpha 1 Chain (Col14a1), 5'-Aminolevulinate Synthase 2 (Alas2). Finally, PFAS modulated circulating pro and anti-inflammatory mediators including IFN-γ (downregulated 3.0-fold in females; p = 0.0301, 2.1-fold in males; p = 0.0418) and IL-6 (upregulated 5.6-fold in males; p = 0.030, no change in females). In conclusion, our data indicate long term exposure to a PFAS mixture impacts mechanisms related to expression of ACE2 in the lung. This work provides a mechanistic rationale for important future studies of PFAS exposure and subsequent viral infection.
Collapse
Affiliation(s)
- Zhao Yang
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Jiahui Ding
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48202, USA
| | - Gil Mor
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA
| | - Michael C Petriello
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
26
|
Cytokines help suggest aplastic anemia with pulmonary bacterial or co-fungal infection. Sci Rep 2022; 12:18373. [PMID: 36319826 PMCID: PMC9626605 DOI: 10.1038/s41598-022-22503-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/17/2022] [Indexed: 12/31/2022] Open
Abstract
Although aplastic anemia (AA) does not come under the category of blood malignant diseases, the infection that frequently occurs in this bone marrow failure can make it worse. Pulmonary infection is the most prevalent but limiting clinical diagnosis. To find biomarkers predicting bacterial or bacterial-combined fungal infections in the lungs, we reviewed 287 AA medical records including 151 without any infection, 87 with pure pulmonary bacterial infection, and 49 with bacterial and fungal infection were reviewed. There were substantial changes in IL-17F, IL-17A, IFN-γ, IL-6, IL-8, and IL-10 levels between the non-infected and lung bacterial infection groups (P < 0.05). Further, a significant variation in IL-17A, TNF-β, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-22, and IL-12p70, between the uninfected group and the pulmonary bacterial and fungal infection group (P < 0.05) was observed. The results further revealed significant differences in TNF-β, IL-12p70, IL-6, IL-8, and IL-10 between the pulmonary bacterial infection group and the fungal infection group (P < 0.05). Moreover, by calculating ROC and cut-off values, we determined that IL-6 (AUC = 0.98, Cut-off = 14.28 pg/ml, P = 0.0000) had a significant advantage than other cytokines, body temperature (AUC = 0.61, P = 0.0050), PCT (AUC = 0.57, P = 0.0592), and CRP (AUC = 0.60, P = 0.0147) in the detection of lungs bacterial infections. In addition, IL-6 (AUC = 1.00, Cut-off = 51.50 pg/ml, P = 0.000) and IL-8 (AUC = 0.87, Cut-off = 60.53 pg/ml, P = 0.0000) showed stronger advantages than other cytokines, body temperature (AUC = 0.60, P = 0.0324), PCT (AUC = 0.72, Cut-off = 0.63 ng/ml, P = 0.0000) and CRP (AUC = 0.79, Cut-off = 5.79 mg/l, P = 0.0000) in distinguishing bacteria from fungi. This may suggest that IL-8 may play a role in differentiating co-infected bacteria and fungi. Such advantages are repeated in severe aplastic anemia (SAA) and very severe aplastic anemia (VSAA).In conclusion, aberrant IL-6 elevations in AA patients may predict the likelihood of bacterial lung infection. The concurrent increase of IL-6 and IL-8, on the other hand, should signal bacterial and fungal infections in patients.These findings may help to suggest bacterial or fungal co-infection in patients with AA (Focus on VSAA and SAA).
Collapse
|
27
|
Leszczyńska K, Jakubczyk D, Górska S. The NLRP3 inflammasome as a new target in respiratory disorders treatment. Front Immunol 2022; 13:1006654. [PMID: 36203607 PMCID: PMC9531678 DOI: 10.3389/fimmu.2022.1006654] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years a continuous increase in new cases of respiratory disorders, such as rhinitis, asthma, and chronic obstructive pulmonary disease (COPD), has been observed. The exact pathomechanism of these diseases is still blurry, resulting in the lack of targeted and effective therapy. The conventional use of treatment strategies, such as antihistamine drugs and/or glucocorticosteroids act mainly symptomatically and have significant side effects. Specific allergen immunotherapy is only useful in the management of specific allergies and selected patients. Therefore, new therapeutic solutions are constantly being sought. The novelty of recent years has been the association between NLRP3 inflammasome activation and the development of airway inflammatory diseases. This seems to be an interesting therapeutic target that may support or even replace traditional therapies in the future. The review presented, discusses the contribution of NLRP3 inflammasome to the development of allergic rhinitis, allergic asthma, and COPD. Moreover, the modulatory properties of probiotics as potential inhibitors of NLRP3 inflammasome are emphasised.
Collapse
|
28
|
Agrawal S, Monteiro C, Baca CF, Mohammadi R, Subramanian V, de Melo Bento CA, Agrawal A. Metabolites and growth factors produced by airway epithelial cells induce tolerance in macrophages. Life Sci 2022; 302:120659. [PMID: 35623392 PMCID: PMC10081865 DOI: 10.1016/j.lfs.2022.120659] [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: 02/14/2022] [Revised: 05/13/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022]
Abstract
Macrophages play a role in preventing inflammation in the respiratory tract. To investigate the mechanisms that lead to tolerance in macrophages, we examined the crosstalk between airway epithelial cells (AECs) and macrophages using a 2D coculture model. Culture of macrophages with AECs led to a significant inhibition of LPS induced pro-inflammatory responses. More importantly, AECs induced the secretion of TGF-β and IL-10 from macrophages even in the absence of LPS stimulation. In addition, the expression of inhibitory molecule, CD200R was also upregulated on AEC exposed macrophages. Furthermore, the AECs exposed macrophages induced significantly increased level of T regulatory cells. Investigation into the possible mechanisms indicated that a combination of growth factor, G-CSF, and metabolites, Kynurenine and lactic acid produced by AECs is responsible for inducing tolerance in macrophages. Interestingly, all these molecules had differential effect on macrophages with G-CSF inducing TGF-β, Kynurenine elevating IL-10, and lactic acid upregulating CD200R. Furthermore, a cocktail of these factors/metabolites induced similar changes in macrophages as AEC exposure. Altogether, these data identify factors secreted by AECs that enhance tolerance in the respiratory tract. These mediators thus have the potential to be used for therapeutic purposes to modulate respiratory inflammation following local viral infections and lung diseases.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, CA, USA 92617
| | - Clarice Monteiro
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, CA, USA 92617; Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California Irvine, CA 92617, USA; Sue and Bill Stem Cell Center, University of California Irvine, CA 92617, USA
| | - Veedamali Subramanian
- Division of Gastroenterology, Department of Medicine, University of California Irvine, CA 92617, USA
| | - Cleonice Alves de Melo Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, CA, USA 92617.
| |
Collapse
|
29
|
Chen S, Xia J, Zhang Y, Zhan Q. IL35 attenuated LPS-induced acute lung injury by regulating macrophage polarization. Mol Biol Rep 2022; 49:5811-5820. [PMID: 35748972 PMCID: PMC9244303 DOI: 10.1007/s11033-022-07293-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/27/2022] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Interleukin 35 (IL35) has been reported to play a role in acute lung injury (ALI); however, the current results regarding the relationship between IL35 and ALI are inconsistent. Therefore, we aimed to further determine the function of IL35 in ALI in mice and the potential mechanism in this paper. MATERIALS AND METHODS Hematoxylin-eosin (HE) staining and Masson staining were used to evaluate lung injury in mice. Immunohistochemical staining was used to evaluate the expression of IL35 p35, TLR4 and MD2 and the Bax/Bcl2 and p-P65/P65 ratios. The expression levels of IL35 EBi3, CD68, CD206 and MPO were assessed by immunofluorescence staining. RT-PCR was used to examine the expression levels of IL1β and IL6. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was performed to detect apoptotic cells. RESULTS Overexpression of IL35 alleviated LPS-induced ALI in mice. IL35 overexpression decreased the expression of CD68 and increased the expression of CD206 in mice with ALI. Furthermore, upregulation of IL35 expression obviously reduced the expression of MPO, IL1β and IL6 in the lung tissues of mice with ALI. Mechanistically, IL35 suppressed the TLR4/NFκB-P65 pathway, leading to the promotion of the M1 to M2 macrophage transition and alleviation of inflammation in mice with ALI. CONCLUSIONS IL35 relieved LPS-induced inflammation and ALI in mice by regulating M1/M2 macrophage polarization and inhibiting the activation of the TLR4/NFκB-P65 pathway.
Collapse
Affiliation(s)
- Shengsong Chen
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No 9, Dongdan Santao, Dongcheng District, 100730, Beijing, P. R. China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No 9, Dongdan Santao, Dongcheng District, 100730, Beijing, P. R. China.
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
| |
Collapse
|
30
|
The Contribution of Viral Proteins to the Synergy of Influenza and Bacterial Co-Infection. Viruses 2022; 14:v14051064. [PMID: 35632805 PMCID: PMC9143653 DOI: 10.3390/v14051064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
A severe course of acute respiratory disease caused by influenza A virus (IAV) infection is often linked with subsequent bacterial superinfection, which is difficult to cure. Thus, synergistic influenza-bacterial co-infection represents a serious medical problem. The pathogenic changes in the infected host are accelerated as a consequence of IAV infection, reflecting its impact on the host immune response. IAV infection triggers a complex process linked with the blocking of innate and adaptive immune mechanisms required for effective antiviral defense. Such disbalance of the immune system allows for easier initiation of bacterial superinfection. Therefore, many new studies have emerged that aim to explain why viral-bacterial co-infection can lead to severe respiratory disease with possible fatal outcomes. In this review, we discuss the key role of several IAV proteins-namely, PB1-F2, hemagglutinin (HA), neuraminidase (NA), and NS1-known to play a role in modulating the immune defense of the host, which consequently escalates the development of secondary bacterial infection, most often caused by Streptococcus pneumoniae. Understanding the mechanisms leading to pathological disorders caused by bacterial superinfection after the previous viral infection is important for the development of more effective means of prevention; for example, by vaccination or through therapy using antiviral drugs targeted at critical viral proteins.
Collapse
|
31
|
Chung O, Jung YE, Lee KW, An YJ, Kim J, Roh YR, Bhak J, Park K, Weber JA, Cheong J, Cha SS, Lee JH, Yim HS. The Analyses of Cetacean Virus-Responsive Genes Reveal Evolutionary Marks in Mucosal Immunity-Associated Genes. Biochem Genet 2022; 60:2299-2312. [PMID: 35334059 PMCID: PMC8949644 DOI: 10.1007/s10528-022-10221-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 03/09/2022] [Indexed: 11/06/2022]
Abstract
Viruses are the most common and abundant organisms in the marine environment. To better understand how cetaceans have adapted to this virus-rich environment, we compared cetacean virus-responsive genes to those from terrestrial mammals. We identified virus-responsive gene sequences in seven species of cetaceans, which we compared with orthologous sequences in seven terrestrial mammals. As a result of evolution analysis using the branch model and the branch-site model, 21 genes were selected using at least one model. IFN-ε, an antiviral cytokine expressed at mucous membranes, and its receptor IFNAR1 contain cetacean-specific amino acid substitutions that might change the interaction between the two proteins and lead to regulation of the immune system against viruses. Cetacean-specific amino acid substitutions in IL-6, IL-27, and the signal transducer and activator of transcription (STAT)1 are also predicted to alter the mucosal immune response of cetaceans. Since mucosal membranes are the first line of defense against the external environment and are involved in immune tolerance, our analysis of cetacean virus-responsive genes suggests that genes with cetacean-specific mutations in mucosal immunity-related genes play an important role in the protection and/or regulation of immune responses against viruses.
Collapse
Affiliation(s)
| | - Ye-Eun Jung
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Kyeong Won Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea
| | - Young Jun An
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea
| | - Jungeun Kim
- Personal Genomics Institute, Genome Research Foundation, Cheongju, 28160, Republic of Korea
| | - Yoo-Rim Roh
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea.,Department of Marine Biotechnology, Korea University of Science and Technology, Daejeon, 306-350, Republic of Korea
| | - Jong Bhak
- Clinomics, Ulsan, 44919, Republic of Korea.,Personal Genomics Institute, Genome Research Foundation, Cheongju, 28160, Republic of Korea.,Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Kiejung Park
- Sangmyung University, Cheonan, 31066, Republic of Korea
| | - Jessica A Weber
- Department of Biology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Jaehun Cheong
- Department of Molecular Biology, Pusan National University, Busan, 46241, Republic of Korea
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jung-Hyun Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea. .,Department of Marine Biotechnology, Korea University of Science and Technology, Daejeon, 306-350, Republic of Korea.
| | - Hyung-Soon Yim
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea.
| |
Collapse
|
32
|
Xu M, Li N, Fan X, Zhou Y, Bi S, Shen A, Wang B. Differential Effects of Toll-Like Receptor Signaling on the Activation of Immune Responses in the Upper Respiratory Tract. Microbiol Spectr 2022; 10:e0114421. [PMID: 35196817 PMCID: PMC8865572 DOI: 10.1128/spectrum.01144-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/28/2022] [Indexed: 11/20/2022] Open
Abstract
Vaccination through the upper respiratory tract (URT) is highly effective for the prevention of respiratory infectious diseases. Toll-like receptor (TLR)-based adjuvants are immunostimulatory and considered potential adjuvant candidates. However, the patterns of immune response to different TLRs at the URT have not been revealed. In this study, SPF mice were preexposed to TLR agonists intranasally to simulate the status of humans. Inflammatory response to TLR agonists and TLR signal-mediated adaptive immune responses were analyzed. The results revealed that similar to human tonsils, inflammatory response to stimulation with TLR4 or TLR2 agonist was attenuated in agonist-exposed mice but not in mice without this exposure. In contrast, TLR9 or TLR3 agonist preexposure did not affect the inflammatory response to restimulation by matching agonists. For the adaptive immune response, after agonist preexposure the antibody response to antigens adjuvanted with TLR4 or TLR2 agonist was substantially restricted, whereas, both antibody and T cell responses to antigens adjuvanted with TLR9 or TLR3 agonist were activated as robustly as in mice without agonist exposure. Moreover, we demonstrate that the mechanisms underlying the differential activation of TLRs are regulated at the level of TLR expression in innate and adaptive immune cells. These results indicate that TLRs on the cell surface (TLR4 and 2) and in the endolysosomal compartments (TLR9 and 3) display distinct immune response patterns. The findings provide important information for the use of TLR agonists as mucosal adjuvants and enhance our understanding of immune responses to bacterial and viral infections in the respiratory mucosa. IMPORTANCE Agonists of TLRs are potential adjuvant candidates for mucosal vaccination. We demonstrated that the TLR-mediated inflammatory and antibody responses in the URT of SPF mice exposed to extracellular TLR agonists were substantially restricted. In contrast, inflammatory and adaptive immune responses, including B and T cell activation, were not desensitized in mice exposed to intracellular TLR agonists. The distinct responsive patterns of extra and intracellular TLRs regulated at TLR expression in immune cells. The results indicated that TLRs differentially impact the innate and adaptive immune response in the URT, which contributes to the selection of TLR-based mucosal adjuvants and helps understand the difference between the immune response in bacterial and viral infections.
Collapse
Affiliation(s)
- Meiyi Xu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Ning Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xin Fan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Ya Zhou
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Shuai Bi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Adong Shen
- Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Beijing, China
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
The interplay between SARS-CoV-2 infected airway epithelium and immune cells modulates regulatory/inflammatory signals. iScience 2022; 25:103854. [PMID: 35128349 PMCID: PMC8802491 DOI: 10.1016/j.isci.2022.103854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 01/27/2022] [Indexed: 01/08/2023] Open
Abstract
To assess the cross-talk between immune cells and respiratory tract during SARS-CoV-2 infection, we analyzed the relationships between the inflammatory response induced by SARS-CoV-2 replication and immune cells phenotype in a reconstituted organotypic human airway epithelium (HAE). The results indicated that immune cells failed to inhibit SARS-CoV-2 replication in the HAE model. In contrast, immune cells strongly affected the inflammatory profile induced by SARS-CoV-2 infection, dampening the production of several immunoregulatory/inflammatory signals (e.g., IL-35, IL-27, and IL-34). Moreover, these mediators were found inversely correlated with innate immune cell frequency (NK and γδ T cells) and directly with CD8 T cells. The enriched signals associated with NK and CD8 T cells highlighted the modulation of pathways induced by SARS-CoV-2 infected HAE. These findings are useful to depict the cell-cell communication mechanisms necessary to develop novel therapeutic strategies aimed to promote an effective immune response. HAE as a model to study the cross-talk between infected epithelium and immune cells Immune cells failed to inhibit SARS-CoV-2 replication Immune cells dampen the production of several signals induced by SARS-CoV-2 infection Decrease of NK/γδ T and increase of CD8 T cells in SARS-CoV-2 infected HAE co-culture
Collapse
|
34
|
Lad N, Murphy A, Parenti C, Nelson C, Williams N, Sharpe G, McTernan P. Asthma and obesity: endotoxin another insult to add to injury? Clin Sci (Lond) 2021; 135:2729-2748. [PMID: 34918742 PMCID: PMC8689194 DOI: 10.1042/cs20210790] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Low-grade inflammation is often an underlying cause of several chronic diseases such as asthma, obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Defining the mediators of such chronic low-grade inflammation often appears dependent on which disease is being investigated. However, downstream systemic inflammatory cytokine responses in these diseases often overlap, noting there is no doubt more than one factor at play to heighten the inflammatory response. Furthermore, it is increasingly believed that diet and an altered gut microbiota may play an important role in the pathology of such diverse diseases. More specifically, the inflammatory mediator endotoxin, which is a complex lipopolysaccharide (LPS) derived from the outer membrane cell wall of Gram-negative bacteria and is abundant within the gut microbiota, and may play a direct role alongside inhaled allergens in eliciting an inflammatory response in asthma. Endotoxin has immunogenic effects and is sufficiently microscopic to traverse the gut mucosa and enter the systemic circulation to act as a mediator of chronic low-grade inflammation in disease. Whilst the role of endotoxin has been considered in conditions of obesity, cardiovascular disease and T2DM, endotoxin as an inflammatory trigger in asthma is less well understood. This review has sought to examine the current evidence for the role of endotoxin in asthma, and whether the gut microbiota could be a dietary target to improve disease management. This may expand our understanding of endotoxin as a mediator of further low-grade inflammatory diseases, and how endotoxin may represent yet another insult to add to injury.
Collapse
Affiliation(s)
- Nikita Lad
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Alice M. Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Cristina Parenti
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Carl P. Nelson
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Neil C. Williams
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Graham R. Sharpe
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Philip G. McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| |
Collapse
|
35
|
Okajima T, Shigemori S, Namai F, Ogita T, Sato T, Shimosato T. Free Feeding of CpG-Oligodeoxynucleotide Particles Prophylactically Attenuates Allergic Airway Inflammation and Hyperresponsiveness in Mice. Front Immunol 2021; 12:738041. [PMID: 34867960 PMCID: PMC8639529 DOI: 10.3389/fimmu.2021.738041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
CpG-oligodeoxynucleotides (CpG-ODNs) constitute an attractive alternative for asthma treatment. However, very little evidence is available from studies on the oral administration of CpG-ODNs in animals. Previously, we developed acid-resistant particles (named ODNcap) as an oral delivery device for ODNs. Here, we showed that free feeding of an ODNcap-containing feed prophylactically attenuates allergic airway inflammation, hyperresponsiveness, and goblet cell hyperplasia in an ovalbumin-induced asthma model. Using transcriptomics-driven approaches, we demonstrated that injury of pulmonary vein cardiomyocytes accompanies allergen inhalation challenge, but is inhibited by ODNcap feeding. We also showed the participation of an airway antimicrobial peptide (Reg3γ) and fecal microbiota in the ODNcap-mediated effects. Collectively, our findings suggest that daily oral ingestion of ODNcap may provide preventive effects on allergic bronchopulmonary insults via regulation of mechanisms involved in the gut-lung connection.
Collapse
Affiliation(s)
- Takuma Okajima
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Suguru Shigemori
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Fu Namai
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Tasuku Ogita
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takashi Sato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
36
|
Guarino C, Pedicelli I, Perna F, Di Spirito V, Fiorentino G, Procaccini F, Rea G. E-cigarette, or vaping, product use Associated Lung Injury (EVALI): new scenarios for physicians and radiologists. Monaldi Arch Chest Dis 2021; 92. [PMID: 34865457 DOI: 10.4081/monaldi.2021.1962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/06/2021] [Indexed: 11/23/2022] Open
Abstract
A 59-year-old female ex-smoker with 40 pack year smoking history and a 5-year current e-cigarette (EC) use history, presented with progressive dyspnea on exertion and daily cough for 2 months. A CT scan showed a consolidation area with air bronchogram in the middle lobe and non-calcific bilateral nodules, which could be attributed to community-acquired pneumonia. The patient was treated with empiric antibiotics and systemic steroids for 10 days. Infectious, neoplastic and autoimmune pathologies were excluded, whereas a broncho-alveolar lavage revealed an accumulation of lipids in the cytoplasm of the alveolar macrophages. Despite the recommendation of vaping cessation, the patient continued to use EC. A new CT exam, carried out after 18 months, showed reversed halo sign (RHS), patchy ground-glass opacity (GGO), pleuro-parenchymal bands, and indeed perilobular pattern, suggestive of organizing pneumonia (OP). The final diagnosis was E-cigarette, or vaping, product use Associated Lung Injury (EVALI)- related OP.
Collapse
Affiliation(s)
- Carmine Guarino
- Unit of Bronchology, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples.
| | - Ilaria Pedicelli
- Unit of Bronchology, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples.
| | - Francesco Perna
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Naples.
| | - Valentina Di Spirito
- Unit of Pathophysiology and Respiratory Rehabilitation, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples.
| | - Giuseppe Fiorentino
- Unit of Pathophysiology and Respiratory Rehabilitation, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples.
| | - Fabio Procaccini
- Department of Diagnostic Imaging, Polydiagnostic Center Vega s.r.l., Naples.
| | - Gaetano Rea
- Department of Diagnostic Imaging, Monaldi Hospital, Azienda Ospedaliera dei Colli, Naples.
| |
Collapse
|
37
|
Gao R, Zu W, Liu Y, Li J, Li Z, Wen Y, Wang H, Yuan J, Cheng L, Zhang S, Zhang Y, Zhang S, Liu W, Lan X, Liu L, Li F, Zhang Z. Quasispecies of SARS-CoV-2 revealed by single nucleotide polymorphisms (SNPs) analysis. Virulence 2021; 12:1209-1226. [PMID: 34030593 PMCID: PMC8158041 DOI: 10.1080/21505594.2021.1911477] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/28/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
New SARS-CoV-2 mutants have been continuously indentified with enhanced transmission ever since its outbreak in early 2020. As an RNA virus, SARS-CoV-2 has a high mutation rate due to the low fidelity of RNA polymerase. To study the single nucleotide polymorphisms (SNPs) dynamics of SARS-CoV-2, 158 SNPs with high confidence were identified by deep meta-transcriptomic sequencing, and the most common SNP type was C > T. Analyses of intra-host population diversity revealed that intra-host quasispecies' composition varies with time during the early onset of symptoms, which implicates viral evolution during infection. Network analysis of co-occurring SNPs revealed the most abundant non-synonymous SNP 22,638 in the S glycoprotein RBD region and 28,144 in the ORF8 region. Furthermore, SARS-CoV-2 variations differ in an individual's respiratory tissue (nose, throat, BALF, or sputum), suggesting independent compartmentalization of SARS-CoV-2 populations in patients. The positive selection analysis of the SARS-CoV-2 genome uncovered the positive selected amino acid G251V on ORF3a. Alternative allele frequency spectrum (AAFS) of all variants revealed that ORF8 could bear alternate alleles with high frequency. Overall, the results show the quasispecies' profile of SARS-CoV-2 in the respiratory tract in the first two months after the outbreak.
Collapse
Affiliation(s)
- Rongsui Gao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Wenhong Zu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yang Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Junhua Li
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Zeyao Li
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yanling Wen
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jing Yuan
- Department of Infectious Diseases, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Disease, Shenzhen, Guangdong Province, China
| | - Lin Cheng
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Shengyuan Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yu Zhang
- National Supercomputing Center in Shenzhen (Shenzhen Cloud Computing Center), Shenzhen, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Weilong Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xun Lan
- Department of Basic Medical Sciences at School of Medicine, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Lei Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Feng Li
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong Province, China
- Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong Province, China
| |
Collapse
|
38
|
Rubey KM, Brenner JS. Nanomedicine to fight infectious disease. Adv Drug Deliv Rev 2021; 179:113996. [PMID: 34634395 PMCID: PMC8665093 DOI: 10.1016/j.addr.2021.113996] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/09/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022]
Abstract
The ubiquity and potency of antibiotics may give the false impression that infection is a solved problem. Unfortunately, even bacterial infections, the target of antibiotics, remain a major cause of illness and death. Several major unmet needs persist: biofilms, such as those on implanted hardware, largely resist antibiotics; the inflammatory host response to infection often produces more damage than the infection itself; and systemic antibiotics often decimate the gut microbiome, which can predispose to additional infections and even predispose to non-infectious diseases. Additionally, there is an increasing threat from multi-drug resistant microorganisms, though market forces may continue to inhibit innovation in this realm. These numerous unmet infection-related needs provide attractive goals for innovation of targeted drug delivery technologies, especially those of nanomedicine. Here we review several of those innovations in pre-clinical development, the two such therapies which have made it to clinical use, and the opportunities for further technology development for treating infections.
Collapse
Affiliation(s)
- Kathryn M Rubey
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jacob S Brenner
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Peng J, Siracusa MC. Basophils in antihelminth immunity. Semin Immunol 2021; 53:101529. [PMID: 34815162 PMCID: PMC8715908 DOI: 10.1016/j.smim.2021.101529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022]
Abstract
It has been appreciated that basophilia is a common feature of helminth infections for approximately 50 years. The ability of basophils to secrete IL-4 and other type 2 cytokines has supported the prevailing notion that basophils contribute to antihelminth immunity by promoting optimal type 2 T helper (Th2) cell responses. While this appears to be the case in several helminth infections, emerging studies are also revealing that the effector functions of basophils are extremely diverse and parasite-specific. Further, new reports now suggest that basophils can restrict type 2 inflammation in a manner that preserves the integrity of helminth-affected tissue. Finally, exciting data has also demonstrated that basophils can regulate inflammation by participating in neuro-immune interactions. This article will review the current state of basophil biology and describe how recent studies are transforming our understanding of the role basophils play in the context of helminth infections.
Collapse
Affiliation(s)
- Jianya Peng
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Mark C Siracusa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
40
|
Wang X, Lv Z, Han B, Li S, Yang Q, Wu P, Li J, Han B, Deng N, Zhang Z. The aggravation of allergic airway inflammation with dibutyl phthalate involved in Nrf2-mediated activation of the mast cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 789:148029. [PMID: 34082215 DOI: 10.1016/j.scitotenv.2021.148029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/06/2021] [Accepted: 05/22/2021] [Indexed: 06/12/2023]
Abstract
Dibutyl phthalate (DBP)-an organic pollutant-is ubiquitous in the environment. DBP as an immune adjuvant is related to the development of multiple allergic diseases. However, the current research involving DBP-induced pulmonary toxicity remains poorly understood. Therefore, this research aimed to explore the adverse effect and potential mechanism of DBP exposure on the lungs in rats. In our study, ovalbumin was used to build a rat model of allergic airway inflammation to study any harmful effect of DBP exposure on lung tissues. Rats were treated by intragastric administration of DBP (500 mg kg-1 or 750 mg kg-1) and/or subcutaneous injection of SFN (4 mg kg-1). The results of histopathological analysis, cell count, and myeloperoxidase showed that DBP promoted the inflammatory damage of lungs. In the lung tissues, the detection of terminal deoxynucleotidyl transferase dUNT nick end labeling and oxidative stress indices showed that DBP significantly increased the level of apoptosis and oxidative stress. Western blot analysis indicated that DBP raised the expression level of thymic stromal lymphopoietin and reduced the nuclear expression level of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was further verified by quantitative real-time PCR. Meanwhile, DBP treatment markedly up-regulated the inflammatory cytokines such as IL-4 and IL-13, and rat mast cell protease-2, a marker secreted by mast cells (MCs). Conversely, sulforaphane, a Nrf2 inducer, ameliorated the pulmonary damage induced by DBP in the above. Altogether, our data provides a new insight into the impacts of the activation of MCs on the DBP-induced pulmonary toxicity as well as the safety evaluation of DBP.
Collapse
Affiliation(s)
- Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ning Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China.
| |
Collapse
|
41
|
Alobaidi A, Alsamarai A, Alsamarai MA. Inflammation in Asthma Pathogenesis: Role of T cells, Macrophages, Epithelial Cells and Type 2 Inflammation. Antiinflamm Antiallergy Agents Med Chem 2021; 20:317-332. [PMID: 34544350 DOI: 10.2174/1871523020666210920100707] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Asthma is a chronic disease with abnormal inflammatory and immunological responses. The disease initiated by antigens in subjects with genetic susceptibility. However, environmental factors play a role in the initiation and exacerbation of asthma attack. Asthma is T helper 2 (Th2)-cell-mediated disease. Recent studies indicated that asthma is not a single disease entity, but it is with multiple phenotypes and endotypes. The pathophysiological changes in asthma included a series of subsequent continuous vicious circle of cellular activation contributed to induction of chemokines and cytokines that potentiate inflammation. The heterogeneity of asthma influenced the treatment response. The asthma pathogenesis driven by varied set of cells such as eosinophils, basophils, neutrophils, mast cells, macrophages, epithelial cells and T cells. In this review the role of T cells, macrophage, and epithelial cells are discussed.
Collapse
Affiliation(s)
- Amina Alobaidi
- Kirkuk University College of Veterinary Medicine, Kirkuk. Iraq
| | - Abdulghani Alsamarai
- Aalborg Academy College of Medicine [AACOM], Denmark. Tikrit University College of Medicine, [TUCOM], Tikrit. Iraq
| | | |
Collapse
|
42
|
Knoll R, Schultze JL, Schulte-Schrepping J. Monocytes and Macrophages in COVID-19. Front Immunol 2021; 12:720109. [PMID: 34367190 PMCID: PMC8335157 DOI: 10.3389/fimmu.2021.720109] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a contagious viral disease caused by SARS-CoV-2 that led to an ongoing pandemic with massive global health and socioeconomic consequences. The disease is characterized primarily, but not exclusively, by respiratory clinical manifestations ranging from mild common cold symptoms, including cough and fever, to severe respiratory distress and multi-organ failure. Macrophages, a heterogeneous group of yolk-sac derived, tissue-resident mononuclear phagocytes of complex ontogeny present in all mammalian organs, play critical roles in developmental, homeostatic and host defense processes with tissue-dependent plasticity. In case of infection, they are responsible for early pathogen recognition, initiation and resolution of inflammation, as well as repair of tissue damage. Monocytes, bone-marrow derived blood-resident phagocytes, are recruited under pathological conditions such as viral infections to the affected tissue to defend the organism against invading pathogens and to aid in efficient resolution of inflammation. Given their pivotal function in host defense and the potential danger posed by their dysregulated hyperinflammation, understanding monocyte and macrophage phenotypes in COVID-19 is key for tackling the disease's pathological mechanisms. Here, we outline current knowledge on monocytes and macrophages in homeostasis and viral infections and summarize concepts and key findings on their role in COVID-19. While monocytes in the blood of patients with moderate COVID-19 present with an inflammatory, interferon-stimulated gene (ISG)-driven phenotype, cellular dysfunction epitomized by loss of HLA-DR expression and induction of S100 alarmin expression is their dominant feature in severe disease. Pulmonary macrophages in COVID-19 derived from infiltrating inflammatory monocytes are in a hyperactivated state resulting in a detrimental loop of pro-inflammatory cytokine release and recruitment of cytotoxic effector cells thereby exacerbating tissue damage at the site of infection.
Collapse
Affiliation(s)
- Rainer Knoll
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L. Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | - Jonas Schulte-Schrepping
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
43
|
Mo L, Luo X, Yang G, Liu J, Yang L, Liu Z, Wang S, Liu D, Liu Z, Yang P. Epithelial cell-derived CD83 restores immune tolerance in the airway mucosa by inducing regulatory T-cell differentiation. Immunology 2021; 163:310-322. [PMID: 33539546 PMCID: PMC8207377 DOI: 10.1111/imm.13317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of generation of regulatory T cells (Treg) remains incompletely understood. Recent studies show that CD83 has immune regulatory functions. This study aims to investigate the role of epithelial cell-derived CD83 in the restoration of immune tolerance in the airway mucosa by inducing the Treg differentiation. In this study, CD83 and ovalbumin (OVA)-carrying exosomes were generated from airway epithelial cells. An airway allergy mouse model was developed to test the role of CD83/OVA-carrying exosomes in the suppression of airway allergy by inducing Treg generation. We observed that mouse airway epithelial cells expressed CD83 that could be up-regulated by CD40 ligand. The CD83 deficiency in epithelial cells retarded the Treg generation in the airway mucosa. CD83 up-regulated transforming growth factor-β-inducible early gene 1 expression in CD4+ T cells to promote Foxp3 expression. Exposure of primed CD4+ T cells to CD83/OVA-carrying exosomes promoted antigen-specific Treg generation. Administration of CD83/OVA-carrying exosomes inhibited experimental airway allergic response. In summary, airway epithelial cells express CD83 that is required in the Treg differentiation in the airway mucosa. Administration of CD83/OVA-carrying exosomes can inhibit airway allergy that has the translation potential in the treatment of airway allergic disorders.
Collapse
Affiliation(s)
- Li‐Hua Mo
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
| | - Xiang‐Qian Luo
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Gui Yang
- Department of OtolaryngologyLonggang Central HospitalShenzhenChina
| | - Jiang‐Qi Liu
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Li‐Teng Yang
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Zhi‐Qiang Liu
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Shuai Wang
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Da‐Bo Liu
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Zhi‐Gang Liu
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
| | - Ping‐Chang Yang
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
- Guangdong Provincial Key Laboratory of Regional Immunity and DiseasesShenzhenChina
| |
Collapse
|
44
|
Keratinocytes Regulate the Threshold of Inflammation by Inhibiting T Cell Effector Functions. Cells 2021; 10:cells10071606. [PMID: 34206914 PMCID: PMC8306889 DOI: 10.3390/cells10071606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Whilst the importance of keratinocytes as a first-line defense has been widely investigated, little is known about their interactions with non-resident immune cells. In this study, the impact of human keratinocytes on T cell effector functions was analyzed in an antigen-specific in vitro model of allergic contact dermatitis (ACD) to nickel sulfate. Keratinocytes partially inhibited T cell proliferation and cytokine production. This effect was dependent on the keratinocyte/T cell ratio and was partially reversible by increasing the number of autologous dendritic cells. The inhibition of T cell proliferation by keratinocytes was independent of the T cell subtype and antigen presentation by different professional antigen-presenting cells. Autologous and heterologous keratinocytes showed comparable effects, while the fixation of keratinocytes with paraformaldehyde abrogated the immunosuppressive effect. The separation of keratinocytes and T cells by a transwell chamber, as well as a cell-free keratinocyte supernatant, inhibited T cell effector functions to the same amount as directly co-cultured keratinocytes, thus proving that soluble factor/s account for the observed suppressive effects. In conclusion, keratinocytes critically control the threshold of inflammatory processes in the skin by inhibiting T cell proliferation and cytokine production.
Collapse
|
45
|
De Sanctis JB, García AH, Moreno D, Hajduch M. Coronavirus infection: An immunologists' perspective. Scand J Immunol 2021; 93:e13043. [PMID: 33783027 PMCID: PMC8250184 DOI: 10.1111/sji.13043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Coronavirus infections are frequent viral infections in several species. As soon as the severe acute respiratory syndrome (SARS) appeared in the early 2000s, most of the research focused on pulmonary disease. However, disorders in immune response and organ dysfunctions have been documented. Elderly individuals with comorbidities exhibit worse outcomes in all the coronavirus that cause SARS. Disease severity in SARS-CoV-2 infection is related to severe inflammation and tissue injury, and effective immune response against the virus is still under analysis. ACE2 receptor expression and polymorphism, age, gender and immune genetics are factors that also play an essential role in patients' clinical features and immune responses and have been partially discussed. The present report aims to review the physiopathology of SARS-CoV-2 infection and propose new research topics to understand the complex mechanisms of viral infection and viral clearance.
Collapse
Affiliation(s)
- Juan Bautista De Sanctis
- Institute of Molecular and Translational MedicineFaculty of Medicine and DentistryPalacky UniversityOlomoucCzech Republic
- Institute of ImmunologyFaculty of MedicineUniversidad Central de VenezuelaCaracasVenezuela
| | - Alexis Hipólito García
- Institute of ImmunologyFaculty of MedicineUniversidad Central de VenezuelaCaracasVenezuela
| | - Dolores Moreno
- Chair of General Pathology and PathophysiologyFaculty of MedicineCentral University of VenezuelaCaracasVenezuela
| | - Marián Hajduch
- Institute of Molecular and Translational MedicineFaculty of Medicine and DentistryPalacky UniversityOlomoucCzech Republic
| |
Collapse
|
46
|
Freire PP, Marques AH, Baiocchi GC, Schimke LF, Fonseca DL, Salgado RC, Filgueiras IS, Napoleao SM, Plaça DR, Akashi KT, Hirata TDC, El Khawanky N, Giil LM, Cabral-Miranda G, Carvalho RF, Ferreira LCS, Condino-Neto A, Nakaya HI, Jurisica I, Ochs HD, Camara NOS, Calich VLG, Cabral-Marques O. The relationship between cytokine and neutrophil gene network distinguishes SARS-CoV-2-infected patients by sex and age. JCI Insight 2021; 6:147535. [PMID: 34027897 PMCID: PMC8262322 DOI: 10.1172/jci.insight.147535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/07/2021] [Indexed: 01/11/2023] Open
Abstract
The fact that the COVID-19 fatality rate varies by sex and age is poorly understood. Notably, the outcome of SARS-CoV-2 infections mostly depends on the control of cytokine storm and the increasingly recognized pathological role of uncontrolled neutrophil activation. Here, we used an integrative approach with publicly available RNA-Seq data sets of nasopharyngeal swabs and peripheral blood leukocytes from patients with SARS-CoV-2, according to sex and age. Female and young patients infected by SARS-CoV-2 exhibited a larger number of differentially expressed genes (DEGs) compared with male and elderly patients, indicating a stronger immune modulation. Among them, we found an association between upregulated cytokine/chemokine- and downregulated neutrophil-related DEGs. This was correlated with a closer relationship between female and young subjects, while the relationship between male and elderly patients was closer still. The association between these cytokine/chemokines and neutrophil DEGs is marked by a strongly correlated interferome network. Here, female patients exhibited reduced transcriptional levels of key proinflammatory/neutrophil-related genes, such as CXCL8 receptors (CXCR1 and CXCR2), IL-1β, S100A9, ITGAM, and DBNL, compared with male patients. These genes are well known to be protective against inflammatory damage. Therefore, our work suggests specific immune-regulatory pathways associated with sex and age of patients infected with SARS-CoV-2 and provides a possible association between inverse modulation of cytokine/chemokine and neutrophil transcriptional signatures.
Collapse
Affiliation(s)
- Paula P Freire
- Department of Immunology, Institute of Biomedical Sciences, and
| | | | | | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, and
| | | | | | | | | | - Desirée R Plaça
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karen T Akashi
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nadia El Khawanky
- Department of Hematology and Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lasse M Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | | | - Robson F Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo
| | - Luis Carlos S Ferreira
- Vaccine Development Laboratory, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | | | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, and Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Canada
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, Washington
| | | | | | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, and.,Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.,Network of Immunity in Infection, Malignancy, and Autoimmunity, Universal Scientific Education and Research Network, São Paulo, Brazil
| |
Collapse
|
47
|
Khan MA, Ashoor GA, Shamma T, Alanazi F, Altuhami A, Kazmi S, Ahmed HA, Mohammed Assiri A, Clemens Broering D. IL-10 Mediated Immunomodulation Limits Subepithelial Fibrosis and Repairs Airway Epithelium in Rejecting Airway Allografts. Cells 2021; 10:1248. [PMID: 34069395 PMCID: PMC8158696 DOI: 10.3390/cells10051248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/01/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Interleukin-10 plays a vital role in maintaining peripheral immunotolerance and favors a regulatory immune milieu through the suppression of T effector cells. Inflammation-induced microvascular loss has been associated with airway epithelial injury, which is a key pathological source of graft malfunctioning and subepithelial fibrosis in rejecting allografts. The regulatory immune phase maneuvers alloimmune inflammation through various regulatory modulators, and thereby promotes graft microvascular repair and suppresses the progression of fibrosis after transplantation. The present study was designed to investigate the therapeutic impact of IL-10 on immunotolerance, in particular, the reparative microenvironment, which negates airway epithelial injury, and fibrosis in a mouse model of airway graft rejection. Here, we depleted and reconstituted IL-10, and serially monitored the phase of immunotolerance, graft microvasculature, inflammatory cytokines, airway epithelium, and subepithelial collagen in rejecting airway transplants. We demonstrated that the IL-10 depletion suppresses FOXP3+ Tregs, tumor necrosis factor-inducible gene 6 protein (TSG-6), graft microvasculature, and establishes a pro-inflammatory phase, which augments airway epithelial injury and subepithelial collagen deposition while the IL-10 reconstitution facilitates FOXP3+ Tregs, TSG-6 deposition, graft microvasculature, and thereby favors airway epithelial repair and subepithelial collagen suppression. These findings establish a potential reparative modulation of IL-10-associated immunotolerance on microvascular, epithelial, and fibrotic remodeling, which could provide a vital therapeutic option to rescue rejecting transplants in clinical settings.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (T.S.); (F.A.); (A.A.); (S.K.); (D.C.B.)
| | | | - Talal Shamma
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (T.S.); (F.A.); (A.A.); (S.K.); (D.C.B.)
| | - Fatimah Alanazi
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (T.S.); (F.A.); (A.A.); (S.K.); (D.C.B.)
| | - Abdullah Altuhami
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (T.S.); (F.A.); (A.A.); (S.K.); (D.C.B.)
| | - Shadab Kazmi
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (T.S.); (F.A.); (A.A.); (S.K.); (D.C.B.)
| | - Hala Abdalrahman Ahmed
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (H.A.A.); (A.M.A.)
| | - Abdullah Mohammed Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (H.A.A.); (A.M.A.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Dieter Clemens Broering
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia; (T.S.); (F.A.); (A.A.); (S.K.); (D.C.B.)
| |
Collapse
|
48
|
The IL-10GFP (VeRT-X) mouse strain is not suitable for the detection of IL-10 production by granulocytes during lung inflammation. PLoS One 2021; 16:e0247895. [PMID: 33979348 PMCID: PMC8115804 DOI: 10.1371/journal.pone.0247895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022] Open
Abstract
The clear and unequivocal identification of immune effector functions is essential to understand immune responses. The cytokine IL-10 is a critical immune regulator and was shown, for example, to limit pathology during various lung diseases. However, the clear identification of IL-10-producing cells is challenging and, therefore, reporter mouse lines were developed to facilitate their detection. Several such reporter lines utilize GFP, including the IL-10GFP (VeRT-X) reporter strain studied here. In line with previous reports, we found that this IL-10GFP line faithfully reports on the IL-10 production of lymphoid cells. However, we show that the IL-10GFP reporter is not suitable to analyse IL-10 production of myeloid cells during inflammation. During inflammation, the autofluorescence of myeloid cells increased to an extent that entirely masked the IL-10-specific GFP-signal. Our data illustrate a general and important technical caveat using GFP-reporter lines for the analysis of myeloid cells and suggest that previous reports on effector functions of myeloid cells using such GFP-based reporters might require re-evaluation.
Collapse
|
49
|
Kim HS, Arellano K, Park H, Todorov SD, Kim B, Kang H, Park YJ, Suh DH, Jung ES, Ji Y, Holzapfel WH. Assessment of the safety and anti-inflammatory effects of three Bacillus strains in the respiratory tract. Environ Microbiol 2021; 23:3077-3098. [PMID: 33899316 DOI: 10.1111/1462-2920.15530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022]
Abstract
Chronic respiratory diseases are part of accumulating health problems partly due to worldwide increase in air pollution. By their antimicrobial and immunomodulatory properties, some probiotics constitute promising alternatives for the prevention and treatment of chronic respiratory diseases. We have isolated Bacillus strains from Korean fermented foods and selected three potentially probiotic strains (two Bacillus subtilis and one Bacillus amyloliquefaciens) based on safety, antimicrobial efficacy, activity against airborne pathogens and their immunomodulatory properties in vivo. Safety evaluation included in silico analysis for confirming absence of virulence genes. Safety for the respiratory tract was confirmed by an in vivo pathogenicity test using a murine model. Antimicrobial activity was displayed against several airborne pathogens. Potential antimicrobial metabolites such as 2,3-butanediol and propylene glycol were identified as possible antagonistic agents. Immunomodulatory properties in vitro were confirmed by upregulation of IL-10 expression in a macrophage cell line. Intranasal instillation and inhalation in an ovalbumin (OVA)-induced lung inflammation murine model reduced T helper type 2 (Th2) cytokines at transcriptional and protein levels in the lungs. The safety and potentially beneficial role of these Bacillus strains could be demonstrated for the respiratory tract of a murine model.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Karina Arellano
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Haryung Park
- HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Svetoslav D Todorov
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Bobae Kim
- HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Hyeji Kang
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Yu Jin Park
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Dong Ho Suh
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Eun Sung Jung
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Yosep Ji
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| |
Collapse
|
50
|
Pathophysiology of Lung Disease and Wound Repair in Cystic Fibrosis. PATHOPHYSIOLOGY 2021; 28:155-188. [PMID: 35366275 PMCID: PMC8830450 DOI: 10.3390/pathophysiology28010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive, life-threatening condition affecting many organs and tissues, the lung disease being the chief cause of morbidity and mortality. Mutations affecting the CF Transmembrane Conductance Regulator (CFTR) gene determine the expression of a dysfunctional protein that, in turn, triggers a pathophysiological cascade, leading to airway epithelium injury and remodeling. In vitro and in vivo studies point to a dysregulated regeneration and wound repair in CF airways, to be traced back to epithelial CFTR lack/dysfunction. Subsequent altered ion/fluid fluxes and/or signaling result in reduced cell migration and proliferation. Furthermore, the epithelial-mesenchymal transition appears to be partially triggered in CF, contributing to wound closure alteration. Finally, we pose our attention to diverse approaches to tackle this defect, discussing the therapeutic role of protease inhibitors, CFTR modulators and mesenchymal stem cells. Although the pathophysiology of wound repair in CF has been disclosed in some mechanisms, further studies are warranted to understand the cellular and molecular events in more details and to better address therapeutic interventions.
Collapse
|